Neuropsychiatric Disorders - Miyoshi, Koho (2010)
Neuropsychiatric Disorders - Miyoshi, Koho (2010)
Neuropsychiatric Disorders - Miyoshi, Koho (2010)
Kiyoshi Maeda
Editors
Neuropsychiatric Disorders
Editors
Koho Miyoshi, M.D., Ph.D. Yasushi Morimura, M.D, Ph.D.
Director Chief Executive Officer
Jinmeikai Research Institute for Mental Jinmeikai Foundation and Hospital
Health 53-20 Kabutoyama-cho, Nishinomiya
53-20 Kabutoyama-cho, Nishinomiya Hyogo 662-0001, Japan
Hyogo 662-0001, Japan
Ohmura Hospital
The former Professor 200 Kitayama-Ohmura, Miki
Department of Neuropsychiatry Hyogo 673-0404, Japan
Kyoto University Graduate School
of Medicine
Kyoto, Japan
© Springer 2010
This work is subject to copyright. All rights are reserved, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting,
reproduction on microfilm or in any other way, and storage in data banks.
The use of general descriptive names, registered names, trademarks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective
laws and regulations and therefore free for general use.
Product liability: The publishers cannot guarantee the accuracy of any information about dosage and
application contained in this book. In every individual case the user must check such information by
consulting the relevant literature.
v
vi Preface
Part I Introduction
vii
viii Contents
Emmeline Agars
Brain and Ageing Program, School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia
David B. Arciniegas
Brain Injury Rehabilitation Unit, HealthONE Spalding Rehabilitation Hospital,
Aurora, CO, USA;
Department of Psychiatry, University of Colorado School of Medicine,
Aurora, CO, USA
Simone Brockman
School of Psychiatry and Clinical Neurosciences, University of Western Australia,
Perth, WA, Australia;
Fremantle Hospital, Fremantle, WA, Australia
Luis Ignacio Brusco
Alzheimer Center, School of Medicine, Buenos Aires University,
Buenos Aires, Argentina
Robertas Bunevičius
Institute of Psychophysiology and Rehabilitation, Kaunas University of Medicine,
Palanga, Lithuania
Anthony S. David
Department of Psychiatry, Section of Cognitive Neuropsychiatry,
Institute of Psychiatry, King’s College London, London, UK
Masaaki Fukutake
Department of Psychiatry, Kobe University School of Medicine, Kobe, Japan
Eduardo Gastelumendi
Peruvian Psychiatric Society; Peruvian Psychoanalytic Society, Lima, Perú
Moises Gaviria
Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA;
Advocate Christ Medical Center, Oak Lawn, IL, USA
xi
xii Contributors
James Gilleen
Department of Psychiatry, Section of Cognitive Neuropsychiatry,
Institute of Psychiatry, King’s College London, London, UK
Kathryn Greenwood
Department of Psychiatry, Section of Cognitive Neuropsychiatry,
Institute of Psychiatry, King’s College London, London, UK
Takashi Haraguchi
Department of Neurology, South Okayama Medical Center, Okayama, Japan
Ryota Hashimoto
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Akira Homma
Department of Psychiatry, Center for Dementia Care Research and Training
in Tokyo, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
Hideki Ishizu
Zikei Hospital, Okayama, Japan
Michael D. Kopelman
Institute of Psychiatry, King’s College London, London, UK;
Academic Unit of Neuropsychiatry, Adamson Centre, London, UK
Kenji Kosaka
Yokohama Houyuu Hospital, Yokohama, Japan
Takashi Kudo
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Shigetoshi Kuroda
Zikei Hospital, Okayama, Japan;
Department of Neuropsychiatry, Okayama University Graduate
School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
Kiyoshi Maeda
Department of Occupational Therapy, Kobe Gakuin University
School of Rehabilitation, Kobe, Japan
Ian Grant McKeith
Wolfson Research Centre, Institute for Ageing and Health, Newcastle
University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
Yoshio Mitsuyama
Psychogeriatric Center of Daigo Hospital, Miyazaki, Japan
Koho Miyoshi
Jinmeikai Research Institute for Mental Health, Nishinomiya, Hyogo, Japan;
The former Professor, Department of Neuropsychiatry, Kyoto University
Graduate School of Medicine, Kyoto, Japan
Contributors xiii
Takashi Morihara
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Yasushi Morimura
Jinmeikai Foundation and Hospital, Nishinomiya, Japan
Yoshio Morita
Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Japan
Marco Mula
Division of Neurology, Maggiore Hospital, Amedeo Avogadro University,
Novara, Italy
Naoki Nishiguchi
Department of Neuropsychiatry, Kinki University School of Medicine,
Osakasayama, Japan
Yoshitaka Ohigashi
International Center, Graduate School of Human and Environmental Studies,
Kyoto University, Kyoto, Japan
Masayasu Okochi
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Spyridon Papapetropoulos
Department of Neurology, Miller School of Medicine, University of Miami,
Miami, FL, USA;
Biogen Idec, Cambridge, MA, USA
Arthur J. Prange, Jr.
Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA
Perminder Sachdev
School of Psychiatry, University of New South Wales, Sydney, NSW, Australia;
Neuropsychiatric Institute, Prince of Wales Hospital, Euroa Centre, Randwick,
NSW, Australia
Blake K. Scanlon
Department of Neurology, Miller School of Medicine, University of Miami,
Miami, FL, USA;
Department of Psychology, University of Miami, Coral Gables, FL, USA
Osamu Shirakawa
Department of Neuropsychiatry, Kinki University School of Medicine,
Osakasayama, Japan
Ingmar Skoog
Neuropsychiatric Epidemiology Unite, Department of Psychiatry,
Institute of Neuroscience and Physiology, Sahlgrenska Academy at Göteborg
University, Göteborg, Sweden
xiv Contributors
Sergio E. Starkstein
School of Psychiatry and Clinical Neurosciences, University of Western Australia,
WA, Australia
Fremantle Hospital, Fremantle, WA, Australia
Shinji Tagami
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Masatoshi Takeda
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Yasuyuki Tanabe
Department of Neurology, South Okayama Medical Center, Okayama, Japan
Toshihisa Tanaka
Department of Psychiatry, Osaka University Medical School, Suita, Japan
Seishi Terada
Department of Neuropsychiatry, Okayama University Graduate
School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
Michael Trimble
Department of Neuropsychiatry, Institute of Neurology, London, UK
Julian Trollor
Brain and Ageing Program, School of Psychiatry, University of New South Wales,
Sydney, NSW, Australia;
Department of Developmental Disability Neuropsychiatry, School of Psychiatry,
University of New South Wales, Sydney, NSW, Australia
Rhonda DePaul Verzal
Director of Neuropsychiatry, Advocate Christ Medical Center,
University of Illinois at Chicago, Chicago, IL, USA
Makiko Yamada
Molecular Imaging Center, National Institute of Radiological Sciences,
Chiba, Japan
Osamu Yokota
Department of Neuropsychiatry, Okayama University Graduate
School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
Clinical Manifestations
of Neuropsychiatric Disorders
Introduction
Almost all brain disorders may cause psychiatric symptoms [1]. Psychiatric symp-
toms caused by organic brain disorders could be called “neuropsychiatric” symp-
toms. Psychiatric manifestations caused by organic brain disease have been
traditionally called organic mental disorders. In DSM-IV-TR, however, the term
“organic psychosis” is not applied to the psychosis caused by organic cerebral dis-
orders. Because the “organic” or neurobiological bases of psychoses, including
Neuropsychiatric Symptoms
apathy, hallucination, delusion, and behavioral disorders. These three components form
complex clinical manifestations of neuropsychiatric disorders.
Multiple neuropsychiatric symptoms, such as cognitive impairment, disturbance
of consciousness, anxiety, mood disorders, hallucination, delusion, behavioral
change, and apathy, commonly occur concurrently in the course of cerebral
disorders (Fig. 2). The psychiatric symptoms are usually accompanied by other
psychiatric symptoms. Namely, depression, neurotic states, behavioral changes,
apathy, and cognitive impairment occur simultaneously in Alzheimer’s disease, and
mood change, apathy, visual hallucination, and cognitive impairment are encoun-
tered concurrently in patients with Parkinson’s disease. Any of the neuropsychiatric
symptoms rarely occur independently in the course of illness. Clinical manifesta-
tions of neuropsychiatric disorders usually consist of the multiple components of
psychiatric symptoms.
Anxiety commonly occurs in the initial stage of a neuropsychiatric disorder [6, 7].
However, it usually becomes less obvious as the progress of cognitive decline
continues. Anxiety, intermingled with depressive mood or dysphoria, is one of the
most common neuropsychiatric symptoms in Alzheimer’s disease and vascular
dementia. Somatic distress, such as headache, vertigo, dizziness, fatigability, list-
lessness, and feeling of weakness are occasional complaints. Excessive anxiety is
usually accompanied by restlessness, irritability, muscle tension, fears, and respiratory
symptoms of anxiety.
Apathy
Mood Disorder
Delirium
Personality changes and language disturbances with less marked memory impair-
ment are the main characteristics of cortical dementia in frontotemporal dementia.
The early clinical features of frontotemporal dementia are changes of character
and social behavior rather than impairment of memory and intellect. With the
progression of the disease, impairment of cognitive functions, including memory,
becomes obvious and slowly increases in severity. Stereotyped speech with a promi-
nent reduction of vocabulary is conspicuous in the advanced stage of illness.
Semantic dementia, as well as progressive nonfluent aphasia, is the characteristic
clinical symptom of this type of dementia. Frontotemporal dementia with motor
neuron disease is clinically characterized by the complication with motor neuron
disease.
Subcortical dementia in progressive supranuclear palsy, corticobasal degenera-
tion, Parkinson’s disease, and Huntington’s disease are characterized by peculiar
“forgetfulness,” psychomotor retardation, and mood changes. Progression of
cognitive impairment with visual disturbance is a characteristic clinical feature of
Creutzfeldt–Jakob disease, which causes a diffuse cortical degeneration with
accentuation of occipital change. Progressive supranuclear palsy is clinically char-
acterized by dementia, supranuclear ophthalmoplegia, and pseudobulbar palsy.
Clinical characteristics of “subcortical dementia,” namely, forgetfulness with
psychomotor retardation, difficulty with complex problem solving and concept
formation, and a relative absence of “cortical” features, including aphasia, apraxia,
and agnosia, was originally described in this disorder.
Corticobasal degeneration is characterized clinically by cognitive impairment,
rigidity, clumsiness, and “alien limb” phenomena.
14 K. Miyoshi and Y. Morimura
Conclusions
References
Abstract Thyroid hormones are important for the development and maturation of
the brain as well as for the functioning of the mature brain. Most thyroid hormone-
responsive genes are sensitive to thyroid hormones only during distinct periods of
brain development, but some are also sensitive in the mature brain. A variety of factors
influence the effects of thyroid hormones in the brain: availability of iodine; thyroid
diseases and dysfunction; genetic variations that affect thyroid axis-related proteins,
such as deiodinases, thyroid hormone transporters, and receptors; and timing of events.
Interaction of these factors contributes to the development of the brain as well as to
presentation of psychiatric symptoms and disorders in the mature brain. Clinical and
subclinical thyroid dysfunction, thyroid autoimmunity, as well as individual genetic
variations and mutations of thyroid axis-related proteins, may contribute not only to the
presentation of psychiatric symptoms and disorders but also to response to psychiatric
treatments. Better understanding of genomic and nongenomic mechanisms related to
thyroid hormone metabolism in the brain opens new venues for finding new markers,
new targets, and new agents for the treatment of mental disorders.
R. Bunevičius (*)
Institute of Psychophysiology and Rehabilitation,
Kaunas University of Medicine, Vyduno 4, 00135 Palanga, Lithuania
e-mail: [email protected]
A.J. Prange, Jr.
Department of Psychiatry, University of North Carolina at Chapel Hill,
Campus Box #7160, Chapel Hill, NC 27599-7160, USA
e-mail: [email protected]
Introduction
Thyroid hormones are important for the development and maturation of the brain
as well as for functioning of the mature brain. Thyroid hormones influence brain
gene expression. The majority of these genes are sensitive to thyroid hormones only
during distinct periods of brain development, but some of them are also sensitive in
the mature brain. Thyroid hormone deficiency during fetal and early postnatal life
results in severe mental retardation and cretinism, while thyroid dysfunction in
adult life results in mental symptoms, such as mood disorders and cognitive dys-
function. Although most psychiatric patients do not display overt thyroid dysfunc-
tion, many patients display lesser abnormalities. A variety of factors influence the
effects of thyroid hormones in the brain: availability of iodine; thyroid diseases and
dysfunction; genetic variations that affect proteins, such as deiodinases, thyroid
hormone transporters, and receptors; and timing of events. Consideration of inter-
action of these factors contributes to better understanding of the presentation of
psychiatric disorders as well as of the response to psychiatric treatments [1].
Thyroid hormones penetrate cell membranes and are responsible for the majority
of genomic and nongenomic effects of thyroid hormones. Until recently it was
presumed that cellular entry by free thyroid hormones was mediated via passive
diffusion because of their lipophylic nature. Now it is recognized that thyroid hor-
mones enter target cells using an energy-dependent transport mechanism that is
mediated by the monocarboxylate transporter-8 (MCT8) and by other transporters
such as the organic anion transporter protein 1c1 (OATP1c1). To enter the brain, thyroid
hormones must cross the blood–brain barrier or the choroid-plexus–cerebrospinal
fluid (CSF) barrier. OATP1c1 is a T4-specific transporter; MCT8 may transport T3
as well as T4. In the brain, T4 enters astrocytes, where it is converted to T3 by local
D2. T3 generated in the astrocytes as well as T3 from the general circulation is
transported into neurons via MCT8, where, after completion of its action, it is
degraded by D3 [5, 6] (Fig. 1).
The genomic action of T3 is mediated via nuclear thyroid hormone receptors (TRs),
which are members of the steroid/thyroid family. Human TRs are encoded by two
genes and have alpha- and beta-receptor isoforms. TRs bind T3 and, as ligand-
inducible transcription factors, regulate expression of T3-responsive target genes.
TR alpha-1, beta-1, and beta-2 isoforms can be occupied by T3, but the TR alpha-2
isoform cannot be occupied by T3. TRs, whether or not occupied by T3, bind to the
DNA response elements, producing different effects on gene expression. Binding
of T3-occupied TRs leads to activation of the responsive gene; binding of T3-
unocupied TRs leads to suppression [7].
TRs are expressed in the brain before fetal thyroid hormone secretion.
TR alpha-1 is the major isoform expressed in the brain during fetal development.
22 R. Bunevičius and A.J. Prange, Jr.
Neuron
Glial Cell
Tanycyte
DNA
CSF
TR
Nucleus
mRNA
Protein
T3 T4 MCT8 D2 ? MCT8 T3 D3 T2
? T4 T3
?
OATP1c1
OATP1c1
MCT8
T4 T3 T4 T3 Circulation
Fig. 1 Thyroid hormone delivery and metabolism in the brain. Thyroxine (T4) is transported across
the blood–brain barrier (BBB) via organic anion transporter protein 1c1 (OATP1c1) located in
endothelium cells. Triiodothyronine (T3) is transported via an unknown mechanism. From the cere-
brospinal fluid (CSF), thyroid hormones are transported via monocarboxylate transporter-8 (MCT8)
located in tanycytes. Choroid plexus (ChP) expresses both transporters OATP1c1 and MCT8. T4
enters the glial cell via an unknown mechanism and is converted to T3 by type II deiodinase (D2).
T3 exits the glial cell via an unknown mechanism and enters the neuron via MCT8. In the neuron,
T3 binds to nuclear thyroid receptor (TR) and acts as a transcription factor initiating protein synthe-
sis, or is converted to inactive diiodothyronine (T2) by type III deiodinase (D3)
has not been started demonstrate symptoms of hypothyroidism and growth retardation
together with mental retardation, spasticity, and speech and language deficits.
Neurological deficits are less severe than in endemic cretinism [13]. Immediate thyroid
hormone replacement prevents most neurological symptoms in neonatal hypothyroid-
ism, although mild deficits in cognitive functioning persist [13].
Endemic cretinism may be prevented if administration of iodine is started in the
first trimester. Despite knowledge of this simple remedy, iodine deficiency remains
the most prevalent preventable cause of mental retardation worldwide [11].
Findings in iodine-deficient areas in China revealed a positive association of D2
polymorphisms with mental retardation [14]. This finding suggests an important
interaction between environmental iodine deficiency and genetic variation,
diminishing the enzymatic conversion of T4 to T3 in the brain.
In most parts of the world, dietary iodine is sufficient. Nevertheless, maternal
hypothyroidism [15] or hypothyroxinemia [16], especially in early pregnancy, is
associated with delay in infant neurodevelopment. Women who are unable to
increase their production of T4 early in pregnancy may constitute a population at
risk for producing children with neurological disabilities [17]. On the other hand,
maternal hyperthyroidism [18] may affect reactivity to stress in offspring [19].
It has been recognized recently that mutations in MCT8 affect T3 transport into the
neuron, causing isolated brain hypothyroidism with elevated serum T3 concentra-
tions [5, 6]. Clinical features of MCT8 mutations include severe mental retardation,
axial hypotonia, absence of speech, and resemblance to patients with Allan–
Herndon–Dudley syndrome (AHDS). In fact, MCT8 mutations were found in all
families with AHDS, providing a molecular basis for this syndrome [20].
Apparently, when hypothyroidism is confined to the brain, the effects on brain
development resemble those caused by systematic hypothyroidism. Although there
are no effective treatments for patients with MCT8 mutation, the detection of this
mutation is important for genetic counseling.
Treatment of Hypothyroidism
Treatment of Hyperthyroidism
It may be that it is not thyroid gland dysfunction per se, but rather thyroid
autoimmune processes, that frequently cause thyroid gland dysfunction [43], is
responsible for comorbidity with mood disorders. Involvement of AIT in brain
functioning was found in several neuroimaging studies. Euthyroid patients with
autoimmune thyroiditis were compared to euthyroid patients without autoimmune
thyroiditis. The former group showed more brain perfusion abnormalities, anxiety,
and depression [44]. Neuroimaging abnormalities were similar to those observed in
Hashimoto’s encephalopathy, an infrequent but life-threatening acute brain syn-
drome caused by AITD. These findings suggest a higher than expected involvement
of the brain in AITD [45].
Thyroid Hormones
Thyrotropin-Releasing Hormone
Some clinical reports, although not all, show that a single intravenous injection
of TRH produces a prompt, albeit partial and brief, relief of symptoms in depressed
patients. Consistent with the state-dependent concept is the finding that TRH also
may cause improvement in manic patients [60]. TRH has yet to find broad accep-
tance as a treatment modality. However, one TRH congener, taltirelin, is available
in Japan for the treatment of spinocerebellar degeneration. Also in Japan an intra-
venous formulation of TRH is available for the treatment of head injury and distur-
bances of consciousness. The recent finding that there are at least two types of TRH
receptors [59] may give added impetus to the search for other TRH-related drugs.
Conclusion
Acknowledgments The authors thank the Hope for Depression Research Foundation (HDRF)
and the Institute of the Study of Affective Neuroscience (ISAN) for research support.
References
1. Bunevicius R (2009) Thyroid disorders in mental patients. Curr Opin Psychiatry 22:391–395
2. Weetman AP (2004) Autoimmune thyroid disease. Autoimmunity 37:337–340
3. Pedersen IB, Knudsen N, Jorgensen T et al (2003) Thyroid peroxidase and thyroglobulin
autoantibodies in a large survey of populations with mild and moderate iodine deficiency. Clin
Endocrinol (Oxf) 58:36–42
4. Peeters RP, van der Deure WM et al (2006) Genetic variation in thyroid hormone pathway
genes; polymorphisms in the TSH receptor and the iodothyronine deiodinases. Eur J Endocrinol
155:655–662
5. Williams GR (2008) Neurodevelopmental and neurophysiological actions of thyroid hormone.
J Neuroendocrinol 20:784–794
6. Suzuki T, Abe T (2008) Thyroid hormone transporters in the brain. Cerebellum 7:75–83
7. Yen PM (2005) Genomic and nongenomic actions of thyroid hormones. In: Braverman
LE, Utiger RD (eds) Warner and Ingbar’s the thyroid: a fundamental and clinical text, 9th edn.
Lippincott Williams & Wilkins, Philadelphia
8. Lifschytz T, Segman R, Shalom G et al (2006) Basic mechanisms of augmentation of
antidepressant effects with thyroid hormone. Curr Drug Targets 7:203–210
30 R. Bunevičius and A.J. Prange, Jr.
30. Mason GA, Bondy SC, Nemeroff CB et al (1987) The effects of thyroid state on beta-adrenergic
and serotonergic receptors in rat brain. Psychoneuroendocrinology 12:261–270
31. Grabe HJ, Volzke H, Ludemann J et al (2005) Mental and physical complaints in thyroid
disorders in the general population. Acta Psychiatr Scand 112:286–293
32. Samuels MH, Schuff KG, Carlson NE et al (2008) Health status, mood, and cognition in
experimentally induced subclinical thyrotoxicosis. J Clin Endocrinol Metab 93:1730–1736
33. Bunevicius R, Velickiene D, Prange AJ Jr (2005) Mood and anxiety disorders in women with
treated hyperthyroidism and ophthalmopathy caused by Graves’ disease. Gen Hosp Psychiatry
27:133–139
34. Trzepacz PT, McCue M, Klein I et al (1988) Psychiatric and neuropsychological response to
propranolol in Graves’ disease. Biol Psychiatry 23:678–688
35. Bunevicius R, Prange AJ Jr (2006) Psychiatric manifestations of Graves’ hyperthyroidism:
pathophysiology and treatment options. CNS Drugs 20:897–909
36. Bunevicius R (2009) Low triiodothyronine syndrome and depression in patients with chronic
heart failure. In: Iervasi G, Pingitore A (eds) Thyroid and heart failure: from pathophysiology
to clinics. Springer, Milan
37. Mafrica F, Fodale V (2008) Thyroid function, Alzheimer’s disease and postoperative cognitive
dysfunction: a tale of dangerous liaisons? J Alzheimers Dis 14:95–105
38. Premachandra BN, Kabir MA, Williams IK (2006) Low T3 syndrome in psychiatric depression.
J Endocrinol Invest 29:568–572
39. Yazici K, Yazici AE, Taneli B (2002) Different neuroendocrine profiles of remitted and non-
remitted schizophrenic patients. Prog Neuropsychopharmacol Biol Psychiatry 26:579–584
40. Engum A, Bjoro T, Mykletun A et al (2002) An association between depression, anxiety and
thyroid function: a clinical fact or an artefact? Acta Psychiatr Scand 106:27–34
41. Bunevicius R, Peceliuniene J, Mickuviene N et al (2007) Mood and thyroid immunity
assessed by ultrasonographic imaging in a primary health care. J Affect Disord 97:85–90
42. Pop VJ, Wijnen HA, Lapkiene L et al (2006) The relation between gestational thyroid parameters
and depression: a reflection of the downregulation of the immune system during pregnancy?
Thyroid 16:485–492
43. Bunevicius R, Lasas L, Kazanavicius G et al (1996) Pituitary responses to thyrotropin releasing
hormone stimulation in depressed women with thyroid gland disorders. Psychoneuroendo-
crinology 21:631–639
44. Zettinig G, Asenbaum S, Fueger BJ et al (2003) Increased prevalence of subclinical brain
perfusion abnormalities in patients with autoimmune thyroiditis: evidence of Hashimoto’s
encephalitis? Clin Endocrinol (Oxf) 59:637–643
45. Chong JY, Rowland LP, Utiger RD (2003) Hashimoto encephalopathy: syndrome or myth?
Arch Neurol 60:164–171
46. Kirkegaard C, Faber J (1991) Free thyroxine and 3, 3’, 5’-triiodothyronine levels in cerebrospinal
fluid in patients with endogenous depression. Acta Endocrinol (Copenh) 124:166–172
47. Baumgartner A (2000) Thyroxine and the treatment of affective disorders: an overview of the
results of basic and clinical research. Int J Neuropsychopharmacol 3:149–165
48. DeLisi LE, Boccio AM, Riordan H et al (1991) Familial thyroid disease and delayed language
development in first admission patients with schizophrenia. Psychiatry Res 38:39–50
49. Philibert RA, Sandhu HK, Hutton AM et al (2001) Population-based association analyses of
the HOPA12bp polymorphism for schizophrenia and hypothyroidism. Am J Med Genet
105:130–134
50. Baumgartner A, Pietzcker A, Gaebel W (2000) The hypothalamic-pituitary-thyroid axis in
patients with schizophrenia. Schizophr Res 44:233–243
51. Jick H, Kaye JA, Jick SS (2004) Antidepressants and the risk of suicidal behaviors. JAMA
292:338–343
52. Altshuler LL, Bauer M, Frye MA et al (2001) Does thyroid supplementation accelerate tricyclic
antidepressant response? A review and meta-analysis of the literature. Am J Psychiatry
158:1617–1622
32 R. Bunevičius and A.J. Prange, Jr.
53. Aronson R, Offman HJ, Joffe RT et al (1996) Triiodothyronine augmentation in the treatment
of refractory depression. A meta-analysis Arch Gen Psychiatry 53:842–848
54. Papakostas GI, Cooper-Kazaz R, Appelhof BC et al (2009) Simultaneous initiation (coinitiation)
of pharmacotherapy with triiodothyronine and a selective serotonin reuptake inhibitor for
major depressive disorder: a quantitative synthesis of double-blind studies. Int Clin
Psychopharmacol 24:19–25
55. Cooper-Kazaz R, van der Deure WM, Medici M et al (2009) Preliminary evidence that a
functional polymorphism in type 1 deiodinase is associated with enhanced potentiation of the
antidepressant effect of sertraline by triiodothyronine. J Affect Disord 116:113–116
56. Iosifescu DV, Bolo NR, Nierenberg AA et al (2008) Brain bioenergetics and response to
triiodothyronine augmentation in major depressive disorder. Biol Psychiatry 63:1127–1134
57. Joffe RT, Singer W (1990) A comparison of triiodothyronine and thyroxine in the potentiation
of tricyclic antidepressants. Psychiatry Res 32:241–251
58. Gary KA, Sevarino KA, Yarbrough GG et al (2003) The thyrotropin-releasing hormone
(TRH) hypothesis of homeostatic regulation: Implications for TRH-based therapeutics.
J Pharmacol Exp Ther 305:410–416
59. Kamath J, Yarbrough GG, Prange AJ Jr et al (2009) The thyrotropin-releasing hormone
(TRH)-immune system homeostatic hypothesis. Pharmacol Ther 121:20–28
60. Yarbrough GG, Kamath J, Winokur A et al (2007) Thyrotropin-releasing hormone (TRH) in
the neuroaxis: therapeutic effects reflect physiological functions and molecular actions. Med
Hypotheses 69:1249–1256
Lack of Insight and Awareness in Schizophrenia
and Neuropsychiatric Disorders
Introduction
The terms “anosognosia,” “insight,” “lack of awareness,” and even “denial” are often
used synonymously to describe a collection of attitudes and behaviors directed at
one’s illness. Anosognosia is generally used to convey lack of awareness of
specific functions seen after brain injury, such as hemiplegia. In contrast, insight
and lack of awareness are typically used to describe the phenomena in psychiatric
disorders, such as schizophrenia, and in neurological conditions, such as
Alzheimer’s disease (AD), where the awareness in question refers to that
of being ill in general and, more specifically, the capacity to judge impairment of
memory, or finally, the content of symptoms, such as delusions and hallucinations,
as not being real [1, 2]. These terms, therefore, differ regarding the object(s) of
insight to which they refer [3]. This distinction is crucial because awareness of an
objective obvious deficit such as hemiplegia would seem to be different in kind
from that of an objectively verifiable but invisible deficit such as amnesia, which
is different again from a subjective experience such as a hallucination.
Within the schizophrenia field there has been an attempt to fractionate insight
into different components or dimensions of awareness, which may, to some degree,
be independent (e.g. [1, 4–6]). David [1] proposed three dimensions: recognition of
having a mental illness, compliance with treatment, and the ability to label unusual
events as pathological. Amador et al. [6] split insight into five components: four
relate to (un)awareness of having a mental disorder, of the effects of medication, of
consequences of illness, and of specific symptoms, and the fifth or final component
is the attribution of symptoms to illness. Popular measures of insight include the
Schedule for the Assessment of Insight – Expanded Version (SAI-E) [7, 8]; the
semistructured interview: Scale to Assess Unawareness of Mental Disorder
(SUMD) [6]; the simple clinician-rated Insight and Treatment Attitudes
Questionnaire (ITAQ) [9]; and the Birchwood self-report Insight Scale (IS) [10].
All these scales have reasonably good intercorrelation [8]. Many authors have used
a single item from general psychopathology schedules, and others, particularly in
the dementia and brain injury fields, have made use of patient–carer discrepancy
questionnaires such as the Patient Competency Rating Scale (PCRS) [11] and the
Dysexecutive Questionnaire (DEX) [12].
Before the early nineteenth century, it would have been seen as a logical contradiction
in terms to talk of “insight” into psychiatric symptoms such as delusions, yet at
around this time, early French “alienists” began to acknowledge the concept of
“partial” insanity, wherein “madness” could be accompanied by lucidity, indicating
that some aspects of mental function could be “deranged” while others were
preserved [3, 13]. It seems intuitive that awareness would be associated with severity
of psychopathology, such that increased severity of delusions and hallucinations
would necessarily by their very nature leave the sufferer unaware that their experi-
ences were not real. Several studies have shown a relationship between awareness
and global psychopathology in schizophrenia [4, 14–20], whereas others have
found associations only with positive symptoms [21–27], or only with negative
symptoms [28–31], or with both positive and negative symptoms (e.g., [32]). Other
studies find awareness to be associated with specific symptoms such as “formal
thought disorder” [33, 34], “degree of grandiosity” [35], or with degree of “unusual
thought content” (equivalent to delusions), as measured by the Brief Psychiatric
Rating Scale [20, 36].
In an effort to clarify the relationship of awareness and psychopathology, Mintz
et al. [32] conducted a meta-analysis of 40 relevant studies and found small nega-
tive associations between awareness and global, positive, and negative symptoms,
accounting for 7.2%, 6.3%, and 5.2% of the variance in awareness, respectively.
This finding would strongly suggest that symptomatology plays only a small part
in the degree of awareness displayed by schizophrenia patients. It must therefore be
concluded that insight is related to psychopathology: as one increases, the other
tends to decrease. Nevertheless the association is weak cross sectionally and variable
longitudinally. In other words, lack of awareness or poor insight is more than “just
psychopathology” [37].
Several studies have shown that schizophrenia patients present with less aware-
ness than patients with other diagnoses such as bipolar disorder and major
depressive disorder [17, 38] and schizo-affective disorder and mood disorder with
and without psychosis ([39]; but see [40, 41]), or with similar levels of awareness
as bipolar patients but less awareness than patients with unipolar affective disorder
[42, 43]. However, others have found no significant differences between different
patient groups [4, 30, 34, 44, 45].
Age has been found to associate with awareness in only a few studies [6, 22, 23, 42].
A meta-analysis reported that age at onset of the disorder moderated the relation-
ship between awareness and symptom clusters [32], such that acute patient status
was also found to act as a moderator variable between awareness and positive
symptoms; acutely ill patients were least aware.
Neuroimaging
As interest in awareness has grown, so has the use of magnetic resonance imaging
(MRI) as an investigative tool. The findings to date suggest an association between
poor insight and reduced total brain volume [53, 54], frontal lobe atrophy [31],
reduced frontal lobe volume [55, 56], reduced cingulate gyrus and temporal lobe
grey matter volume [56], and ventricular enlargement [14]. There is, however, some
inconsistency in these findings, much study variation in the location of brain–
insight correlates, and in some instances a failure to identify any brain abnormalities
associated with poor insight [57]. One explanation for this inconsistency could
Lack of Insight and Awareness in Schizophrenia and Neuropsychiatric Disorders 37
Mood
One of the more reliable findings in the literature is the positive correlation
between awareness and low mood or depression (and between elevated mood
and lack of awareness [41]), which has been shown across different patient
groups [3, 63]. Although findings are variable, many studies have reported that
increased awareness in schizophrenia is associated with greater depressive
symptoms [25, 34, 47, 64–69], including a meta-analysis [32]. In this way, low
awareness of symptoms and illness is conceptualized as a form of denial to main-
tain self-esteem and preclude the psychological consequences of acknowledging
one has a mental illness.
A critical question is this: Does a depressive mood ensue from awareness of
illness, or does a depressive mood foster a more self-critical attitude? The frame-
work of “depressive realism” may help explain this association (see [70]). On the
other hand, Rathod et al. [71] reported that patients undergoing cognitive-behavioral
therapy (CBT) intervention to improve awareness became depressed subsequent to
gaining awareness, but this result was not found in an earlier study [72].
even at the earliest stages [74]. Understanding awareness in dementia has important
implications [63, 75], as it has been found to be associated with greater perceived
burden of care by their carers [76], delayed diagnosis [77], and poorer outcome after
rehabilitative treatment [78].
Awareness has been shown to worsen with disease progression [79] and has
been found to be associated more frequently with damage to certain anatomical
sites, for example, right frontal and parietal lobes [80–82], which are also often
found to be associated with anosognosia following brain injury (see [80]). Other
studies have found awareness to worsen with disease severity [83], although
some have not [84, 85]. Similarly, although some studies have found an association
between “frontal”/executive performance and awareness [86, 87], others have
not [88, 89].
Less commonly, unawareness is conceived as being a defense mechanism against
the knowledge and consequences of illness. As already noted with respect to schizo-
phrenia, defensive denial is proposed to serve to defend Alzheimer’s patients from
depression, and indeed patients with less insight show less depressive symptomatology
[82, 83, 90, 91], but again other studies have not shown this [77, 84, 92], and the
direction of causality of this effect is debatable.
Most studies of awareness investigate a single patient population. Is there a case for
comparing different patient groups on the same measures [100, 101]? As well as
perhaps revealing more about the nature of awareness per se, doing so also allows
us to observe how awareness may differ in these different groups where pathology
is a known factor. By contrasting different patient groups, it may be possible to better
elucidate which mechanisms subserve awareness, or alternatively it may reveal that
patients from different clinical groups show lack of awareness for different reasons.
Just as patients with schizophrenia have, in addition to their core symptoms, cognitive
impairments and behavioral and social deficits, so patients with AD and brain injury
may have a range of psychopathologies about which they may or may not have
degrees of awareness. Comparison within – as well as between – groups may have
important theoretical implications. For example, if patient groups with widely divergent
pathologies (e.g., Alzheimer and schizophrenia patients) both have memory deficits,
then we can ask whether the same factors associated with awareness of these are
consistent across the groups (e.g., [102, 103] for awareness of cognitive deficits in
schizophrenia). If the answer is broadly affirmative, it suggests that a common cognitive
structure of awareness pertains, and this in turn can prompt overarching cognitive
models that need not be overly concerned with diagnosis. Furthermore, if, say, awareness
of psychopathology is relatively independent of awareness into cognitive deficits (the
correlation was nonsignificant in one recent study [102], regardless of diagnosis), it
points to modularity in awareness. Modularity of “awarenesses” is perhaps the most
likely pattern from the neurological literature [79] (Fig. 1). Such modularity has seldom
� � �
Insight into… Insight into… Insight into… Insight into… Insight into…
modulation
Mood +/–
Fig. 1 Model of multiple, modality-specific awareness systems (modularity), each of which may
or may not be impaired, but with general modulation by factors such as mood. In this theoretical
example, insight is preserved into social difficulties, psychiatric symptoms, and physical disability
but not cognitive problems and poor activities of daily living (ADLs)
40 J. Gilleen et al.
been tested using broad domains such as psychopathology and behavioral problems
(alongside neuropsychological impairments). Where it has, as in the large
pan-European brain injury study [104], considerable within-diagnosis heterogeneity
was found. Similarly, in the dementia field different levels of awareness have been
noted by contrasting behavior with cognition [63, 73, 75, 105].
Participants
Methods
Results
Between-Group Contrasts
Across the groups, patient and informant ratings of behavioral problems, as mea-
sured by the total DEX score, were highly discrepant in the brain injury and AD
groups (see Table 1; Fig. 2), representing low awareness of behavioral impairments,
but this was much less so in the schizophrenia group, suggesting that patients
exhibit different levels of unawareness of behavioral deficits. Importantly, ratings
made by either patients or informants in any patient group were not grossly at
ceiling or floor, but varied somewhat, suggesting that these scales provided
sensitivity in measuring behavioral impairment and, in turn, awareness.
42 J. Gilleen et al.
Fig. 2 Graphs show mean Dysexecutive Questionnaire (DEX) self and informant subfactor
scores (per individual item) paneled according to patient group. Zero scores reflect the absence of
problems. The lower panel shows the resultant DEX discrepancy scores for the three groups. For
ease of presentation, the sign of the discrepancy score has been reversed so that a more positive
value indicates greater discrepancy (i.e., less patient awareness). BI brain injury; ALZ Alzheimer’s;
SCZ schizophrenia
functioning, but their ratings were concordant with the informants’ view across all
subdomains; this reflects good awareness of behavioral problems. The lower panel
shows that this results in lowest awareness for the Alzheimer group, and greatest
awareness for the schizophrenia group, but also that there is no consistent profile
across patients, failing to support the notion of a hierarchy of awareness.
The entire patient sample was pooled (n = 84), and Pearson correlational analyses
were performed between the awareness and other clinical and symptoms ratings.
The DEX discrepancy score correlated with clinician-rated insight (SAI-E) at 0.321
(P < 0.001) and self-rated depression on the Beck Depression Inventory at r = 0.562
(P < 0.001) (Fig. 3); SAI-E also correlated with BDI at r = 0.239 (P < 0.05). That is
to say, as the discrepancy score became closer to zero or positive, indicating good
awareness, so did depression scores increase. DEX scores were correlated signifi-
cantly negatively with overall psychopathology as measured by the BPRS
(r = –0.570, P < 0.001); that is, as the discrepancy became smaller (i.e., awareness
increased), so depressive symptoms became worse.
Fig. 3 Scatterplot shows the correlation between DEX discrepancy scores (DEX-D) and total
Beck Depression scores (BDtot) for the three patient groups: schizophrenia (SCZ), Alzheimer’s
(ALZ), and brain injury (BI). The regression line shows that increasing awareness is associated
with increasing depression scores
44 J. Gilleen et al.
Acknowledgments James Gilleen was supported by an MRC Ph.D. Studentship. We are grateful
to Prof. Simon Lovestone for facilitating access to his Alzheimer cohort; Laura Bach helped with
recruitment of brain injury patients, and Sandra Randell provided secretarial support. We are
grateful to all the patients and carers who generously took part in our studies.
References
9. McEvoy JP, Apperson LJ, Appelbaum PS et al (1989) Insight in schizophrenia. Its relationship
to acute psychopathology. J Nerv Ment Dis 177:43–47
10. Birchwood M et al (1994) A self-report Insight Scale for psychosis: reliability, validity and
sensitivity to change. Acta Psychiatr Scand 89:62–67
11. Prigatano GP, Fordyce DJ (1986) Cognitive dysfunction and psychosocial adjustment after
brain injury. In: Prigatano GP, Fordyce DJ (eds) Neuropsychological rehabilitation after brain
injury. John Hopkins University Press, Baltimore, pp 96–118
12. Burgess PW, Alderman N, Evans J et al (1998) The ecological validity of tests of executive
function. J Int Neuropsychol Soc 4:547–558
13. Berrios GE (2004) Insight: a conceptual history. In: Amador XF, David AS (eds) Insight and
psychosis: awareness of illness in schizophrenia and related disorders, 2nd edn. Oxford
University Press, Oxford
14. Takai A, Uematsu M, Ueki H et al (1992) Insight and its related factors in chronic schizo-
phrenic patients: a preliminary study. Eur J Psychiatry 6:159–170
15. Young DA, Davila R, Scher H (1993) Unawareness of illness and neuropsychological
performance in chronic schizophrenia. Schizophr Res 10:117–124
16. Kemp RA, Lambert TJ (1995) Insight in schizophrenia and its relationship to psychopathology.
Schizophr Res 18:21–28
17. Fennig S, Everett E, Bromet EJ et al (1996) Insight in first-admission psychotic patients.
Schizophr Res 22:257–263
18. Young DA, Zakzanis KK, Bailey C et al (1998) Further parameters of insight and neuropsy-
chological deficit in schizophrenia and other chronic mental disease. J Nerv Ment Dis
186:44–50
19. Rossell SL, Coakes J, Shapleske J et al (2003) Insight: its relationship with cognitive function,
brain volume and symptoms in schizophrenia. Psychol Med 33:111–119
20. Keshavan MS, Rabinowitz J, DeSmedt G et al (2004) Correlates of insight in first episode
psychosis. Schizophr Res 70:187–194
21. Almeida OP, Levy R, Howard RJ et al (1996) Insight and paranoid disorders in late life (late
paraphrenia). Int J Geriatr Psychiatry 11:653–658
22. Kim Y, Sakamoto K, Kamo T et al (1997) Insight and clinical correlates in schizophrenia.
Compr Psychiatry 38:117–123
23. Lysaker PH, Bell MD, Bryson G et al (1998) Neurocognitive function and insight in schizo-
phrenia: support for an association with impairments in executive function but not with
impairments in global function. Acta Psychiatr Scand 97:297–301
24. Schwartz RC (1998) Symptomatology and insight in schizophrenia. Psychol Rep
82:227–233
25. Carroll A, Fattah S, Clyde Z et al (1999) Correlates of insight and insight change in schizo-
phrenia. Schizophr Res 35:247–253
26. McCabe R, Quayle E, Beirne AD et al (2002) Insight, global neuropsychological functioning,
and symptomatology in chronic schizophrenia. J Nerv Ment Dis 190:519–525
27. Olfson M, Marcus SC, Wilk J et al (2006) Awareness of illness and nonadherence to
antipsychotic medications among persons with schizophrenia. Psychiatr Serv 57:205–211
28. Macpherson R, Jerrom B, Hughes A (1996) Relationship between insight, educational
background and cognition in schizophrenia. Br J Psychiatry 168:718
29. Collins AA, Remington GJ, Coulter K et al (1997) Insight, neurocognitive function and symp-
tom clusters in chronic schizophrenia. Schizophr Res 27:37–44
30. Cuesta MJ, Peralta V, Zarzuela A (2000) Reappraising insight in psychosis. Multi-scale
longitudinal study. Br J Psychiatry 177:233–240
31. Laroi F, Fannemel M, Ronneberg U et al (2000) Unawareness of illness in chronic schizophre-
nia and its relationship to structural brain measures and neuropsychological tests. Psychiatry
Res 100:49–58
32. Mintz AR, Dobson KS, Romney DM (2003) Insight in schizophrenia: a meta-analysis.
Schizophr Res 61:75–88
46 J. Gilleen et al.
33. Baier M, DeShay E, Owens K et al (2000) The relationship between insight and clinical fac-
tors for persons with schizophrenia. Arch Psychiatr Nurs 14:259–265
34. Smith TE, Hull JW, Israel LM et al (2000) Insight, symptoms, and neurocognition in schizo-
phrenia and schizoaffective disorder. Schizophr Bull 26:193–200
35. Heinrichs DW, Cohen BP, Carpenter WT Jr (1985) Early insight and the management of
schizophrenic decompensation. J Nerv Ment Dis 173:133–138
36. Overall JE, Gorham D (1962) The Brief Psychiatric Rating Scale. Psychol Rep 10:799–812
37. David AS (2004) The clinical importance of insight. In: Amador XF, David AS (eds) Insight
and psychosis: awareness of illness in schizophrenia and related disorders. Oxford University
Press, Oxford
38. Michalakeas A, Skoutas C, Charalambous A et al (1994) Insight in schizophrenia and mood
disorders and its relation to psychopathology. Acta Psychiatr Scand 90:46–49
39. Amador XF et al (1994) Awareness of illness in schizophrenia and schizoaffective and mood
disorders. Arch Gen Psychiatry 51:826–836
40. Varga M, Magnusson A, Flekkoy K et al (2007) Clinical and neuropsychological correlates of
insight in schizophrenia and bipolar I disorder: does diagnosis matter? Compr Psychiatry
28:583–591
41. Ghaemi SN, Rosenquist KJ (2004) Is insight in mania state-dependent?: a meta-analysis.
J Nerv Ment Dis 192:771–775
42. Weiler MA, Fleisher MH, Arthur-Campbell D (2000) Insight and symptom change in schizo-
phrenia and other disorders. Schizophr Res 45:29–36
43. Pini S, Cassano GB, Dell’Osso L et al (2001) Insight into illness in schizophrenia, schizoaf-
fective disorder, and mood disorders with psychotic features. Am J Psychiatry 158:122–125
44. David A, van Os J, Jones P et al (1995) Insight and psychotic illness. Cross-sectional and
longitudinal associations. Br J Psychiatry 167:621–628
45. Arduini L, Kalyvoka A, Stratta P et al (2003) Insight and neuropsychological function in
patients with schizophrenia and bipolar disorder with psychotic features. Can J Psychiatry
48:338–341
46. Marks KA, Fastenau PS, Lysaker PH et al (2000) Self-Appraisal of Illness Questionnaire
(SAIQ): relationship to researcher-rated insight and neuropsychological function in schizo-
phrenia. Schizophr Res 45:203–211
47. Pyne JM, Bean D, Sullivan G (2001) Characteristics of patients with schizophrenia who do
not believe they are mentally ill. J Nerv Ment Dis 189:146–153
48. David AS (1999) To see ourselves as others see us. Aubrey Lewis’s insight. Br J Psychiatry
174:210–216
49. Cooke MA, Peters ER, Kuipers E et al (2005) Disease, deficit or denial? Models of poor
insight in psychosis. Acta Psychiatr Scand 112:4–17
50. Lysaker P, Bell M (1994) Insight and cognitive impairment in schizophrenia. Performance
on repeated administrations of the Wisconsin Card Sorting Test. J Nerv Ment Dis
182:656–660
51. Morgan KD, David AS (2004) Neuropsychological studies of insight in patients with psy-
chotic disorders. In: Insight and psychosis: awareness of illness in schizophrenia and related
disorders. Oxford University Press, Oxford
52. Aleman A, Agrawal N, Morgan KD et al (2006) Insight in psychosis and neuropsychological
function: meta-analysis. Br J Psychiatry 189:204–212
53. Flashman LA, McAllister TW, Andreasen NC et al (2000) Smaller brain size associated with
unawareness of illness in patients with schizophrenia. Am J Psychiatry 157:1167–1169
54. McEvoy JP, Johnson J, Perkins D et al (2006) Insight in first-episode psychosis. Psychol Med
36:1385–1393
55. Flashman LA, McAllister TW, Johnson SC et al (2001) Specific frontal lobe subregions cor-
related with unawareness of illness in schizophrenia: a preliminary study. J Neuropsychiatry
Clin Neurosci 13:255–257
56. Sapara A, Cooke M, Fannon D et al (2007) Prefrontal cortex and insight in schizophrenia: a
volumetric MRI study. Schizophr Res 89:22–34
Lack of Insight and Awareness in Schizophrenia and Neuropsychiatric Disorders 47
57. Bassitt DP, Neto MRL, de Castro CC et al (2007) Insight and regional brain volumes in
schizophrenia. Eur Arch Psychiatry Clin Neurosci 257:58–62
58. Ha TH, Youna T, Ha K et al (2004) Grey matter abnormalities in paranoid schizophrenia and
their clinical correlations. Psychiatry Res Neuroimaging 132:251–260
59. Shad MU, Muddasani S, Keshavan MS (2006) Prefrontal subregions and dimensions of
insight in first-episode schizophrenia: a pilot study. Psychiatry Res 146:35–42
60. Cooke MA, Fannon D, Kuipers E et al (2008) Neurological basis of poor insight in psychosis:
a voxel-based MRI study. Schizophr Res 103:40–51
61. Schmitz TW, Johnson SC (2007) Relevance to self: a brief review and framework of neural
systems underlying appraisal. Neurosci Biobehav Rev 31:585–596
62. Berti A, Bottini G, Gandola M et al (2005) Shared cortical anatomy for motor awareness and
motor control. Science 309:488–491
63. Clare L (2004) Awareness in early-stage Alzheimer’s disease: a review of methods and evi-
dence. Br J Clin Psychol 43:177–196
64. Smith TE, Hull JW, Santos L (1998) The relationship between symptoms and insight in
schizophrenia: a longitudinal perspective. Schizophr Res 33:63–67
65. Moore O, Cassidy E, Carr A et al (1999) Unawareness of illness and its relationship with
depression and self-deception in schizophrenia. Eur Psychiatry 14:264–269
66. Iqbal Z, Birchwood M, Chadwick P et al (2000) Cognitive approach to depression and suicidal
thinking in psychosis. 2. Testing the validity of a social ranking model. Br J Psychiatry
177:522–528
67. White R, Bebbington P, Pearson J et al (2000) The social context of insight in schizophrenia.
Soc Psychiatry Psychiatr Epidemiol 35:500–507
68. Schwartz RC (2001) Self-awareness in schizophrenia: its relationship to depressive
symptomatology and broad psychiatric impairments. J Nerv Ment Dis 189:401–403
69. Mutsatsa SH, Joyce EM, Hutton SB et al (2006) Relationship between insight, cognitive func-
tion, social function and symptomatology in schizophrenia: the West London first episode
study. Eur Arch Psychiatry Clin Neurosci 256:356–363
70. Ghaemi SN (2007) Feeling and time: the phenomenology of mood disorders, depressive
realism, and existential psychotherapy. Schizophr Bull 33:122–130
71. Rathod S, Kingdon D, Smith P et al (2005) Insight into schizophrenia: the effects of cognitive
behavioural therapy on the components of insight and association with sociodemographics:
data on a previously published randomised controlled trial. Schizophr Res 74:211–219
72. Kemp R, Kirov G, Everitt B et al (1998) Randomised controlled trial of compliance therapy:
18-month follow-up. Br J Psychiatry 172:413–419
73. Vasterling JJ, Seltzer B, Foss JW et al (1995) Unawareness of deficit in Alzheimer’s disease.
Domain-specific differences and disease correlates. Neuropsychiatry Neuropsychol Behav
Neurol 8:26–32
74. Starkstein SE, Chemerinski E, Sabe L et al (1997) Prospective longitudinal study of depression
and anosognosia in Alzheimer’s disease. Br J Psychiatry 171:47–52
75. Clare L (2004) The construction of awareness in early-stage Alzheimer’s disease: a review of
concepts and models. Br J Clin Psychol 43:155–175
76. Seltzer B, Vasterling JJ, Hale MA et al (1995) Unawareness of memory deficit in Alzheimer’s
disease relation to mood and other disease variables. Neuropsychiatry Neuropsychol Behav
Neurol 8:176–181
77. Derouesne C, Thibault S, Lagha-Pierucci S et al (1999) Decreased awareness of cognitive deficits
in patients with mild dementia of the Alzheimer type. Int J Geriatr Psychiatry 14:1019–1030
78. Koltai DC, Welsh-Bohmer KA, Schmechel DE (2001) Influence of anosognosia on treatment
outcome among dementia patients. Cogn Rehabil Dement 3:455–475
79. McGlynn SM, Schacter DL (1989) Unawareness of deficits in neuropsychological syndromes.
J Clin Exp Neuropsychol 11:143–205
80. Laroi F, Barr WB, Keefe RSE (2004) The neuropsychology of insight in psychiatric and neu-
rological disorders. In: Amador X, David A (eds) Insight and psychosis: awareness of illness
in schizophrenia and related disorders. Oxford University Press, Oxford
48 J. Gilleen et al.
81. Vogel A, Hasselbalch SG, Gade A et al (2005) Cognitive and functional neuroimaging
correlate for anosognosia in mild cognitive impairment and Alzheimer’s disease. Int J
Geriatr Psychiatry 20:238–246
82. Harwood DG, Sultzer DL, Wheatley MV (2000) Impaired insight in Alzheimer disease:
association with cognitive deficits, psychiatric symptoms, and behavioral disturbances.
Neuropsychiatry Neuropsychol Behav Neurol 13:83–88
83. Feher EP, Mahurin RK, Inbody SB et al (1991) Anosognosia in Alzheimer’s disease.
Neuropsychiatry Neuropsychol Behav Neurol 4:136–146
84. DeBettignies BH, Mahurin RK, Pirozzolo FJ (1990) Insight for impairment in independent liv-
ing skills in Alzheimer’s disease and multi-infarct dementia. J Clin Exp Neuropsychol
12:355–363
85. Michon A, Deweer B, Pillon B et al (1994) Relation of anosognosia to frontal lobe dysfunc-
tion in Alzheimer’s disease. J Neurol Neurosurg Psychiatry 57:805–809
86. Auchus AP, Goldstein FC, Green J et al (1994) Unawareness of cognitive impairments in
Alzheimer’s disease. Neuropsychiatry Neuropsychol Behav Neurol 7:25–29
87. Lopez OL, Becker JT, Somsak D et al (1994) Awareness of cognitive deficits and anosognosia
in probable Alzheimer’s disease. Eur Neurol 34:277–282
88. Dalla Barba G, Parlato V, Iavarone A et al (1995) Anosognosia, intrusions and frontal functions
in Alzheimer’s disease and depression. Neuropsychologia 33:247–259
89. Migliorelli R, Teson A, Sabe L et al (1995) Prevalence and correlates of dysthymia and major
depression among patients with Alzheimer’s disease. Am J Psychiatry 152:37–44
90. Sevush S (1999) Relationship between denial of memory deficit and dementia severity in
Alzheimer disease. Neuropsychiatry Neuropsychol Behav Neurol 12:88–94
91. Feher EP, Larrabee GJ, Sudilovsky A et al (1994) Memory self-report in Alzheimer’s disease
and in age-associated memory impairment. J Geriatr Psychiatry Neurol 7:58–65
92. Zanetti O, Vallotti B, Frisoni GB et al (1999) Insight in dementia: when does it occur?
Evidence for a nonlinear relationship between insight and cognitive status. J Gerontol B
Psychol Sci Soc Sci 54:100–106
93. Prigatano GP, Altman IM (1990) Impaired awareness of behavioral limitations after trau-
matic brain injury. Arch Phys Med Rehabil 71:1058–1064
94. Sherer M, Boake C, Levin E (1998) Characteristics of impaired awareness after traumatic
brain injury. J Int Neuropsychol Soc 4:380–387
95. Trudel TM, Tryon WW, Purdum CM (1998) Awareness of disability and long-term outcome
after traumatic brain injury. Rehabil Psychol 43:267–281
96. Sherer M, Hart T, Nick TG et al (2003) Early impaired self-awareness after traumatic brain
injury. Arch Phys Med Rehabil 84:168–176
97. Wise K, Ownsworth T, Fleming J (2005) Convergent validity of self-awareness measures and
their association with employment outcome in adults following acquired brain injury. Brain
Inj 19:765–775
98. Bergquist TF, Jacket MP (1993) Awareness and goal setting with the traumatically -brain
injured. Brain Inj 7:275–282
99. Prigatano GP (2005) Disturbances of self-awareness and rehabilitation of patients with trau-
matic brain injury: a 20-year perspective. J Head Trauma Rehabil 20:19–29
100. Banks SJ, Weintraub S (2009) Generalized and symptom-specific insight in behavioral variant
frontotemporal dementia and primary progressive aphasia. J Neuropsychiatry Clin Neurosci
21:299–306
101. O’Keeffe FM, Murray B, Coen RF et al (2007) Loss of insight in frontotemporal dementia,
corticobasal degeneration and progressive supranuclear palsy. Brain 130:753–764
102. Medalia A, Lim RW (2004) Self-awareness of cognitive functioning in schizophrenia.
Schizophr Res 71:331–338
103. Kircher TTJ, Koch K, Stottmeister F et al (2007) Metacognition and reflexivity in patients
with schizophrenia. Psychopathology 40:254–260
104. Teasdale TW, Christensen AL, Willmes K et al (1997) Subjective experience in brain injured
patients and their close relatives: a European Brain Injury Questionnaire study. Brain Inj
11:543–564
Lack of Insight and Awareness in Schizophrenia and Neuropsychiatric Disorders 49
105. Vasterling JJ, Seltzer B, Watrous WE (1997) Longitudinal assessment of deficit unawareness
in Alzheimer’s disease. Neuropsychiatry Neuropsychol Behav Neurol 10:197–202
106. Burgess PW, Alderman N, Emslie H, Evans JJ, Wilson BA (1996) The dysexecutive
questionnaire. In: Wilson BA, Alderman N, Burgess PW, Emslie H, Evans JJ (eds)
Behavioural assessment of the Dysexecutive Syndrome. Thames Valley Test Company, Bury
St. Edmunds, UK
107. Bayard S, Capdevielle D, Boulenger JP et al (2009) Dissociating self-reported cognitive
complaint from clinical insight in schizophrenia. Eur Psychiatry 24:251–258
108. Flashman LA, Roth RM, McAllister TW et al (2007) Dissociation of awareness of different
components of illness in schizophrenia. J Neuropsychiatry Clin Neurosci 19:221–222
109. Morgan KD, Dazzan P, Morgan C et al (2010). Insight, grey matter and cognitive function
in first-onset psychosis, Brit J Psychiat (in press).
Visual Hallucinations in Neurodegenerative
Disorders
Introduction
Neurodegenerative diseases are characterized by progressive decline in the
structure, activity, and function of the central nervous system. Neuronal dropout
usually results in disruption of multiple pathways, regional neurotransmitter
Epidemiology
Dementia with Lewy bodies (DLB) is frequently recognized as the second most
common degenerative dementia [1]. Although extensive epidemiological literature
is lacking, a recent review indicates that prevalence estimates for DLB range from
0.1% to 5.0% in the general population and from 1.7% and 30.5% in dementia cases
[1]. In accordance with current consensus criteria, recurrent VH are among the core
Visual Hallucinations in Neurodegenerative Disorders 53
features indicative of DLB [2]. Approximately two-thirds of patients with DLB present
with VH [3, 4]. Patients with DLB commonly present with hallucinations, and
nearly half have hallucinations within 2 years after the first clinical symptom [5].
A recent community-based autopsy study found that cases with VH were more
likely than cases without VH to have Lewy-related pathology (78% versus 45%)
[6]. Overall cortical Lewy body (LB) burden is greater in cases with early VH (at
onset or within the first 2 years of the disease course), which relates to more LBs
in the inferior temporal cortices [5]. Additional neuropathological evidence indi-
cates that VH are associated with more LB pathology in the parahippocampus and
amygdala [5].
Neuroimaging technologies are also used to elucidate factors associated with VH.
Although most effort focuses on identifying imaging markers that differentiate DLB
from other dementing disorders, some investigations examine potential mechanisms
related specifically to VH in DLB. 18F-Fluorodeoxyglucose-positron emission
tomography (18F-FDG-PET) evaluations show that people living with DLB and VH
have relative metabolic reductions in the right temporo-occipital junction and the
right middle frontal gyrus compared to those with DLB without VH [7].
Certain clinical characteristics are also associated with VH in DLB. The
Consortium on DLB [8] reports that VH may particularly present during periods of
diminished consciousness. Visual impairment exacerbates VH; this is likely a con-
sequence of selective sensory deprivation, and an increase in environmental stimu-
lation may provide temporary relief [8]. People living with DLB and VH perform
worse on overlapping figure identification compared to those without VH [9].
Additional evidence shows that DLB and demented PD patients with recurrent VH
are significantly more impaired in visual discrimination, space-motion perception,
and object-form perception than patients without VH [10].
While people living with DLB have hallucinatory experiences that encompass a
variety of modalities, VH are most common [4]. These VH are frequently complex,
occur at least once a day, last for minutes, and consist of a single, colored, station-
ary object in the central visual field [11]. The experiences are often benign and not
perceived as threatening. However, there are exceptions. These characteristics are
largely similar to those evidenced in PD dementia. Specific phenomenological
characteristics of VH in DLB include anonymous people/soldiers; body parts;
animals; friends and family; children/babies; and machines [11]. VH in DLB tend
to persist and are stable over time [12].
54 S. Papapetropoulos and B.K. Scanlon
Parkinson’s Disease
Epidemiology
Worldwide prevalence rates of Parkinson’s disease (PD) tend to vary widely [18].
Estimates of the number of people living with PD in the world’s ten most populous
nations, along with Western Europe’s five most populous nations, range between
4.1 and 4.6 million [19]. This number is expected to double to somewhere between
8.7 and 9.3 million by 2030. Annual incidence ranges between 4.9 and 26 per
100,000 [20]. Hallucinations occur in 20% to 40% of PD patients receiving
symptomatic therapy [21], although as many as 75% of patients develop
hallucinatory phenomena of a visual nature [22]. VH in PD are often considered
treatment related because prevalence rates in the pre-levodopa era were as low as
5% [23, 24].
Clinical Characteristics of VH in PD
The VH most often reported in PD are drug induced, complex, and consist of ani-
mate or inanimate objects or persons [31]. However, perceptual phenomena that are
more transient and less clear can also occur [28]. VH typically appear several years
after disease onset and usually contain five or fewer images, which are sometimes
meaningful to the patient. The typical “hallucinator’s experience” [32] occurs while
alert and with eyes open, in dim surroundings. The hallucination is present for a few
seconds and then suddenly vanishes. Initially, hallucinations are “friendly.” Patients
often see fragmented figures of beloved familiar persons or animals. As reality test-
ing and insight further decrease, the content of the hallucinations may change to
frightening images (e.g., insects, rats), inducing anxiety and panic attacks [33].
Hallucinations may become malignant, disabling, and intermingled with paranoid
patterns, including suspiciousness, negativism, and sexual accusations. Ideas of
persecution, fearfulness, agitation, aggression, confusion, and delirium become
commonplace. It is at this point that the situation at home often becomes unbear-
able, and the patient is frequently placed in a nursing home [34]. Although VH are
predominant, auditory hallucinations (mixed with visual) are reported to affect
about 10% of patients [28].
VH in Tauopathies
Alzheimer’s Disease
Epidemiology
AD is the most common degenerative dementia. There were 4.5 million people
living with AD in the United States in 2000; this number is expected to nearly triple
by 2050 [49]. A population-based European study estimates the age-standardized
prevalence of AD to be 4.4%, with prevalence increasing with age [50]. Nearly
19% of patients with AD experience VH [51]. Data on persistence of psychotic
symptoms, including VH, in AD are mixed, and methodological differences likely
contribute to disparate findings.
Visual Hallucinations in Neurodegenerative Disorders 57
Clinical Characteristics of VH in AD
supranuclear gaze palsy, and postural instability [57]. Prevalence estimates range
from 1.39/100,000 to 6.5/100,000 [58–60], and incidence is estimated to be 5.3
new cases per 100,000 person-years in people 50 to 99 years of age [61].
Estimates of VH prevalence in PSP range from 9.1% to 13.4% [62, 63]. Several
studies report phenomenological characteristics of VH in PSP [62, 64–66]. VH
are frequently whole people, animals, or fragmented faces. They are familiar
sights and occur during twilight or nighttime more often than not. VH can present
with an auditory component and may become frightening as complexity increases
and disease progresses. The relationship between VH in PSP and antiparkinso-
nian treatment appears to be equivocal. Nonpharmacological interventions are the
first-line treatment, and pharmacotherapy may be useful if the VH become threat-
ening and severely impair quality of life. Antipsychotics show limited utility in
treating VH in PSP. However, before implementation, the risk of severe adverse
events must be strongly considered [62].
Frontotemporal Dementia
may be useful. Assuming neuroleptics are not needed for the control of involuntary
movements, newer agents such as risperidone, olanzepine, or quetiapine may be best
as these have less chance of extrapyramidal side effects [80]. Systematic review
deems risperidone “possibly useful” for the treatment of psychosis in HD [81].
Creutzfeldt–Jakob disease (CJD), although quite rare, is the most common prion
disease affecting humans. Several forms of the disease are currently recognized,
including sporadic, familial, iatrogenic, and variant. Rapidly progressive dementia
and myoclonus are the cardinal features of sporadic CJD (sCJD). One study
describing VH in CJD reports patients hallucinate birds, people, and other visual
phenomena [82]. Cases of sCJD are frequently referred to as the Heidenhain variant
if visual disturbances, including VH, occur within the first week after onset [83].
VH are present in approximately 13% of Heidenhain variant cases and 2.3% of all
sCJD cases [83]. However, other studies show hallucinations of unspecified modal-
ity occur in nearly 25% of cases [84].
Although VH occur in other neurodegenerative disorders, this review is unable
to address them all. As the neuropsychiatric features of neurodegenerative disorders
receive more attention in the literature, a more developed understanding of symp-
tom presentation and treatment options will likely ensue.
Conclusions
References
52. Lin SH, Yu CY, Pai MC (2006) The occipital white matter lesions in Alzheimer’s disease
patients with visual hallucinations. Clin Imaging 30(6):388–393
53. Wilson RS, Krueger KR, Kamenetsky JM et al (2005) Hallucinations and mortality in
Alzheimer disease. Am J Geriatr Psychiatry 13(11):984–990
54. Bassiony MM, Lyketsos CG (2003) Delusions and hallucinations in Alzheimer’s disease:
review of the brain decade. Psychosomatics 44(5):388–401
55. Scarmeas N, Brandt J, Albert M et al (2005) Delusions and hallucinations are associated with
worse outcome in Alzheimer disease. Arch Neurol 62(10):1601–1608
56. Ballard C, Waite J (2006) The effectiveness of atypical antipsychotics for the treatment of
aggression and psychosis in Alzheimer’s disease. Cochrane Database Syst Rev 1:CD003476
57. Williams DR, Lees AJ (2009) Progressive supranuclear palsy: clinicopathological concepts
and diagnostic challenges. Lancet Neurol 8(3):270–279
58. Golbe LI, Davis PH, Schoenberg BS et al (1988) Prevalence and natural history of progressive
supranuclear palsy. Neurology 38(7):1031–1034
59. Schrag A, Ben-Shlomo Y, Quinn NP (1999) Prevalence of progressive supranuclear palsy and
multiple system atrophy: a cross-sectional study. Lancet 354(9192):1771–1775
60. Nath U, Ben-Shlomo Y, Thomson RG et al (2001) The prevalence of progressive supranuclear
palsy (Steele-Richardson-Olszewski syndrome) in the UK. Brain 124(Pt 7):1438–1449
61. Bower JH, Maraganore DM, McDonnell SK et al (1997) Incidence of progressive supranu-
clear palsy and multiple system atrophy in Olmsted County, Minnesota, 1976 to 1990.
Neurology 49(5):1284–1288
62. Papapetropoulos S, Mash DC (2005) Visual hallucinations in progressive supranuclear palsy.
Eur Neurol 54(4):217–219
63. Williams DR, Warren JD, Lees AJ (2008) Using the presence of visual hallucinations to dif-
ferentiate Parkinson’s disease from atypical parkinsonism. J Neurol Neurosurg Psychiatry
79(6):652–655
64. Compta Y, Marti MJ, Rey MJ et al (2009) Parkinsonism, dysautonomia, REM behaviour
disorder and visual hallucinations mimicking synucleinopathy in a patient with progressive
supranuclear palsy. J Neurol Neurosurg Psychiatry 80(5):578–579
65. Cooper AD, Josephs KA (2009) Photophobia, visual hallucinations, and REM sleep behavior
disorder in progressive supranuclear palsy and corticobasal degeneration: a prospective study.
Parkinsonism Relat Disord 15(1):59–61
66. Diederich NJ, Leurgans S, Fan W et al (2008) Visual hallucinations and symptoms of REM
sleep behavior disorder in Parkinsonian tauopathies. Int J Geriatr Psychiatry 23(6):598–603
67. Rosso SM, Donker Kaat L, Baks T et al (2003) Frontotemporal dementia in The Netherlands:
patient characteristics and prevalence estimates from a population-based study. Brain 126(Pt
9):2016–2022
68. Ratnavalli E, Brayne C, Dawson K et al (2002) The prevalence of frontotemporal dementia.
Neurology 58(11):1615–1621
69. Mercy L, Hodges JR, Dawson K et al (2008) Incidence of early-onset dementias in
Cambridgeshire, United Kingdom. Neurology 71(19):1496–1499
70. Neary D, Snowden JS, Gustafson L et al (1998) Frontotemporal lobar degeneration: a consen-
sus on clinical diagnostic criteria. Neurology 51(6):1546–1554
71. Le Ber I, Guedj E, Gabelle A et al (2006) Demographic, neurological and behavioural char-
acteristics and brain perfusion SPECT in frontal variant of frontotemporal dementia. Brain
129(Pt 11):3051–3065
72. Mourik JC, Rosso SM, Niermeijer MF et al (2004) Frontotemporal dementia: behavioral
symptoms and caregiver distress. Dement Geriatr Cogn Disord 18(3–4):299–306
73. Liu W, Miller BL, Kramer JH et al (2004) Behavioral disorders in the frontal and temporal
variants of frontotemporal dementia. Neurology 62(5):742–748
74. Reischle E, Sturm K, Schuierer G et al (2003) A case of schizophreniform disorder in fronto-
temporal dementia (FTD). Psychiatr Prax 30(suppl 2):S78–S82
Visual Hallucinations in Neurodegenerative Disorders 63
75. Mendez MF, Lauterbach EC, Sampson SM et al (2008) An evidence-based review of the
psychopathology of frontotemporal dementia: a report of the ANPA Committee on Research.
J Neuropsychiatry Clin Neurosci 20(2):130–149
76. Mendez MF, Lipton A (2001) Emergent neuroleptic hypersensitivity as a herald of presenile
dementia. J Neuropsychiatry Clin Neurosci 13(3):347–356
77. van Duijn E, Kingma EM, van der Mast RC (2007) Psychopathology in verified Huntington’s
disease gene carriers. J Neuropsychiatry Clin Neurosci 19(4):441–448
78. Paulsen JS, Ready RE, Hamilton JM et al (2001) Neuropsychiatric aspects of Huntington’s
disease. J Neurol Neurosurg Psychiatry 71(3):310–314
79. Zappacosta B, Monza D, Meoni C et al (1996) Psychiatric symptoms do not correlate with
cognitive decline, motor symptoms, or CAG repeat length in Huntington’s disease. Arch
Neurol 53(6):493–497
80. Rosenblatt A, Ranen N, Nance M, et al (1999) A physician’s guide to the management of
Huntington’s disease. http://www.hdsa.org/images/content/1/1/11289.pdf. Accessed 1 Sept
2009
81. Bonelli RM, Wenning GK (2006) Pharmacological management of Huntington’s disease: an
evidence-based review. Curr Pharm Des 12(21):2701–2720
82. Proulx AA, Strong MJ, Nicolle DA (2008) Creutzfeldt-Jakob disease presenting with visual
manifestations. Can J Ophthalmol 43(5):591–595
83. Appleby BS, Appleby KK, Crain BJ et al (2009) Characteristics of established and proposed
sporadic Creutzfeldt-Jakob disease variants. Arch Neurol 66(2):208–215
84. Lundberg PO (1998) Creutzfeldt-Jakob disease in Sweden. J Neurol Neurosurg Psychiatry
65(6):836–841
Organic Delusional Syndrome: Tentative
Neuropsychological Mechanism of Delusions
Y. Ohigashi
International Center, Graduate School of Human and Environmental Studies,
Kyoto University, Kyoto, Japan
e-mail: [email protected]
M. Yamada (*)
Molecular Imaging Center,
NationalInstituteofRadiologicalSciences,Chiba,Japan
e-mail: [email protected]
Introduction
Case SA was a 23-year-old female ambidextrous college student. She had a traffic
accident at age 16 years and had been unconscious for about 10 days. She received
a craniotomy to remove a left parietal hematoma. Her lesions were detected in the
right temporal pole and the left parietal lobe (Fig. 1). When she was discharged from
hospital after 3 months, she showed mildly impaired memory retention, reduced
calculation ability, articulation disorder, and mild right hemiparesis. After the acci-
dent, she was noticed by her family and friends to behave and talk like a child. She
complained of her difficulty in memory retention and concentration; however, she
had spent a quasi-normal daily life without many serious problems.
About 4 years later she began to experience hallucinations and delusions. She
complained: “my father looks like a different person,” “I feel as if TV is speaking
of me,” and “I’m very scared and always feel as if I’m chased by someone.” She
also experienced functional auditory hallucinations, such as “I hear the sound of
typing computer keyboard as human voice.”
Case DR was a 36-year-old right-handed woman. She received a college educa-
tion. A traffic accident at age 14 resulted in skull fracture and intracranial hemorrhage
with a loss of consciousness for 3 days. Her major complaint was headache, insom-
nia, and irritability. After 5 years after the accident, she began to develop persecutory
delusion, such as “I hear some noise that speaks evil of me” and “my vulgar idea is
detected by other people.” She was first diagnosed as “schizophrenia” in a psychiatric
department but was now rediagnosed as PDFTBI. Computed tomography (CT) and
single-photon emission computerized tomography (SPECT) indicated apparent
lesions in the left anterior temporal lobe and the left posterior frontal cortex (Fig. 2).
The ability of both patients to evaluate emotional intensity from others’ faces was
investigated using the Emotional Intensity Scale (EIS [5]) and compared with that of
healthy subjects. In this task, subjects were asked to rate the perceived intensity of a
given emotion word (six basic emotions in total) for each facial expression of six basic
emotions (see [5]fordetailedprocedure).ResultsindicatedthatcaseSAperceiveda
sad facial expressions not only as “sadness” itself but also as high intensities of
68 Y. Ohigashi and M. Yamada
Fig. 2 CTfindingsofcaseDR
5 5
**
Happiness intensity
Surprise intensity
Mean Rating of
Mean Rating of
4 4
3 3
*
2 2
**
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
5 5
** **
4 4
**
Mean Rating of
Mean Rating of
Anger intensity
Fear intensity
3 3
2 2
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
5 5
4 4
Sadness intensity
Mean Rating of
Disgust intensity
Mean Rating of
3 3
2 2
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
Fig. 3 Results of Emotional Intensity Scale in case SA. Horizonta axis, evaluated facial
expressions; vertical axis, intensity of an emotion word
“surprise” and “fear” (Fig. 3). This patient showed a tendency to perceive facial expressions
in heightened emotions of “surprise,” “disgust,” and “happiness” compared to
normalsubjects.CaseDRperceivedemotionsoffear,anger,andsadnessinalmostall
facial expressions except for happiness (Fig. 4).
OrganicDelusionalSyndrome:TentativeNeuropsychologicalMechanismofDelusions 69
Happiness intensity 5 5 **
Surprise intensity
** 4
Mean Rating of
4
Mean Rating of
3 3
**
2 2
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
5 5
** ** **
4 ** ** 4 **
Mean Rating of
Mean Rating of
Anger intensity
**
Fear intensity
3 3
2 2
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
5 5
** ** ** **
**
Sadness intensity
4 ** 4
Disgust intensity
Mean Rating of
Mean Rating of
3 3
2 2
1 1
0 0
happiness surprise fear anger disgust sadness happiness surprise fear anger disgust sadness
Facial Expression Facial Expression
Fig. 4 ResultsofEmotionalIntensityScale(EIS)incaseDR
Several common features between the two cases can be pointed out. (1) Their
common lesion site was the temporal pole. (2) Psychotic states appeared 4 to 5 years
after head injuries. (3) Persecutory delusion with hallucination was the predominant
clinicalpictureinbothcases.(4)ResultsofEISsuggestedtheconfusedjudgmentsof
facial expressions, characterized by a negatively biased-emotion estimation.
The temporal pole is known to play a crucial role in high-level recognition
because it is anatomically located at the endpoint of the auditory and visual “what”
stream. A general function of the temporal pole is supposed to be to couple emotional
responses to highly processed sensory stimuli through a tight connection with the
amygdala [6]. Thus, the temporal pole is regarded as a relay point that links the
final “what” perception to emotional responses in the amygdala. The lesion sites
in our cases suggest that the route from the temporal pole to the amygdala could be
disrupted, and that the functions between these two sites might be isolated. As
indicated by EIS results, the patients overestimated negative emotions regardless of
the actual emotional signals of the facial expression. This overestimation of negative
emotion has been observed in the case of ictal fear [7], which implied that the
70 Y. Ohigashi and M. Yamada
amygdala was not dysfunctional but rather hypersensitive. Consistent with this
amygdala hypersensitivity account, a negatively biased perception in our cases
could have persisted through limbic kindling, which might have created a trait-like
misinterpretation of others’ minds, or persecutory delusions.
In sum, the temporal pole lesions in patients with PDFTBI may segregate the function
of the amygdala from visual information processing. This isolation may cause the
amygdala to respond to emotional stimuli overly intensively and unselectively. We regard
this as one possible cause of delusional perception and persecutory delusion in PDFTBI.
Visual Input
Structural encoding
A B
C
Name Name Person identity Affect response Skin conductance
production Retrieval nodes To familiar stimuli response
Fig. 5 Model of face recognition and Capgras delusion, formulated by Ellis and Young [10]
(figure is a reprint from [11], modified for this publication). Disconnection in the overt recognition
route (a) yields prosopagnosia, whereas that in the covert recognition route (b) produces Capgras
syndrome
been identified. (5) The model explains the face recognition processing but ignores
the occasional coexistence of misidentification of objects or places. (6) The model
does not account for patients’ resistance to modify their delusional belief in the
presence of very strong evidence against it.
Among different explanations for DMS provided by other researchers, a two-
factor model, proposed by Coltheart and his colleagues [15], takes into consider-
ation the foregoing issue (see “6,” above). In their model, although the first factor
is composed of the failure of autonomic responsiveness to familiar faces, the second
abnormal factor is the patient’s resistance to revise the delusional belief, which is
speculated to arise from a disrupted belief evaluation system associated with the
right frontal cortex. In accord with their idea, neuroimaging studies revealed that
the right prefrontal cortex was involved in mediating the ability to detect or resolve
conflicts in thinking [16, 17]. In this sense, we also hypothesize that the delusion
in Capgras syndrome as well as other DMSs might represent a dysfunction of the
cognitive-conflict resolution mechanism, which is supported by the right frontal
lobe. Further support for this view is observed from evidence that right frontotemporal
lesions are predominant in patients with Capgras syndrome in the setting of focal
72 Y. Ohigashi and M. Yamada
Anosognosia for left hemiplegia was first described by Babinski [1] as the denial
of left hemiplegia. Somatoparaphrenia was reported by Gerstmann [2] as delu-
sional beliefs concerning a contralesional side of body (the left side in most
cases), which is characterized by a pathological alteration of the ownership of the
limbs. In a famous monograph, The Parietal Lobes [23], Critchley classified the
content of distortion of the body image as follows: (1) unilateral neglect, (2) lack
of concern (anosodiaphoria), (3) unawareness of hemiparesis (anosognosia), (4)
defective appreciation of the existence of hemiparesis, (5) denial of hemiparesis,
(6) denial of hemiparesis with confabulation, (7) loss of awareness of one body-
half (asomatognosia), (8) undue heaviness, deadness, or lifelessness of one half,
(9) phantom third limb, (10) personification of paralyzed limb, and (11) misople-
gia (the last two categories were included in 1955 and 1974, respectively). In his
categorization, anosognosia and various somatoparaphrenia were not sharply
demarcated but closely related. In 1972, Hécaen [24] distinguished hemi-
somatognosic disorder into three categories: (1) anosognosia for the left hemiple-
gia, (2) hemi-asomatognosia raging from simple neglect to amnesia, unawareness
of one side of the body, and (3) feeling of absence of one’s body part or one side
of the body, including disownership and phantom limbs. Apparently, these disor-
ders still lie on a continuum.
Here we attempt to provide a neuropsychological mechanism of anosognosia
(unawareness of left hemiparesis) and a feeling of disownership of the left side of
the body, based on a novel framework of consciousness proposed by Edelman
[25] and on our refined definitions of “body schema” and “body consciousness.”
Body schema represents a symbolic semiotic or linguistic body concept, possibly
OrganicDelusionalSyndrome:TentativeNeuropsychologicalMechanismofDelusions 73
SELF NONSELF
Internal homeostatic systems World signals
and proprioception
Conceptual
categorization
Reentrant loop
Connection value-category
Special
Value-category
Memory in frontal, temporal.
Parietal areas
supported by the left parietal lobe. Phylogenetically, new “body schema,” which
would have coincided with the genesis of language, could be attributed to the
higher-order consciousness within Edelman’s reentry hypothesis for the genesis
of consciousness [25] (Fig. 6). The close relationship between body schema and
language is supported by the evidence that impaired body schema can provoke,
for instance, bilateral finger agnosia or autotopagnosia. In contrast, body con-
sciousness represents a basic and immediate body feeling that would be phyloge-
netically older than body schema. We would attribute body consciousness to the
primary consciousness in Edelman’s model, which may be supported predomi-
nantly by the right hemisphere.
74 Y. Ohigashi and M. Yamada
Conclusion
References
1. Babinski MJ (1914) Contribution a l’értude des troubles mentaux dans l’hemiplegie organique
cérébrale.RevNeurol1:845–848
2. Gerstmann J (1942) Problem of imperception of disease and of impaired body territories with
organiclesions.Relationtobodyschemeanditsdisorders.ArchNeurolPsychiatry48:890–913
3. Fujii DE, Ahmed I (2002) Psychotic disorder following traumatic brain injury: a conceptual
framework.CognNeuropsychiatry7:41–62
4. Fujii DE, Ahmed I, Hishimura E (2004) A neuropsychological comparison of psychotic
disorder following traumatic brain injury, traumatic brain injury without psychotic disorder,
andschizophrenia.JNeuropsychiatryClinNeurosci16:306–314
5. Adolphs R, Tranel D, Damasio H et al (1994) Impaired recognition of emotion in facial
expressionsfollowingbilateraldamagetothehumanamygdala.Nature(Lond)372:669–672
6. OlsonIR,PlotzkerA,EzzyatbY(2007)Theenigmatictemporalpole:areviewoffindingson
social and emotional processing. Brain 130:1718–1731
7. Yamada M, Murai T, Sato W et al (2005) Emotion recognition from facial expressions in a
temporallobeepilepticpatientwithictalfear.Neuropsychologia43:434–441
8. Capgras J, Reboul-Lachaux J (1923) L’illusion des ‘soises’ dans un délire systématisé
chronique. Bull Soc Clin Med Ment 11:6–16. In: Enoch MD, Trethowan WH (eds)
Uncommon psychiatric syndromes. Wright and Sons, Bristol, 1979
9. Forstl H, Almeida OP, Owen AM et al (1991) Psychiatric, neurological and medical aspects
of misidentification syndromes: a review of 260 cases. Psychol Med 21:905–910
10. Ellis HD, Young AW (1990) Accounting for delusional misidentification. Br J Psychiatry 157:239–248
11. Ellis HD (2007) Delusions: a suitable case for imaging? Int J Psychophysiol 62:146–151
12. HirsteinW,RamachandranVS(1997)Capgrassyndrome:anovelprobeforunderstandingthe
neural representation of the identity and familiarity of persons. Proc Biol Sci 264:437–444
13. BreenN,CaineD,ColtheartM(2000)Modelsoffacerecognitionanddelusionalmisidenti-
fication:acriticalreview.CognNeuropsychol17:55–71
14. Hohwy J (2004) Top-down and bottom-up in delusion formation. Philos Psychiatr Psychol
11:65–70
15. Coltheart M, Langdon R, McKay R (2007) Schizophrenia and monothematic delusions.
Schizophr Bull 33:642–647
16. FinkGR,MarshallJC,HalliganPWetal(1999)Theneuralconsequencesofconflictbetween
intention and the senses. Brain 122:497–512
17. Goel V, Buchel C, Frith C et al (2000) Dissociation of mechanisms underlying syllogistic
reasoning.Neuroimage12:504–514
18. Alexander MP, Stuss DT, Benson DF (1979) Capgras syndrome: a reduplicative phenomenon.
Neurology29:334–339
19. Staton RD, Brumback RA, Wilson H (1982) Reduplicative paramnesia: a disconnection
syndrome of memory. Cortex 18:23–35
20. Moser DJ, Cohen RA, Malloy PF et al (1998) Reduplicative paramnesia: longitudinal
neurobehavioralandneuroimaginganalysis.JGeriatrPsychiatryNeurol11:174–180
21. Papageorgiou P, Ventouras E, Lykouras L et al (2003) Psychophysiological evidence for
alteredinformationindelusionalmisidentificationsyndromes.ProgNeuropsychopharmacol
Biol Psychiatry 27:365–372
22. Young G (2008) Capgras delusion: an interactionist model. Conscious Cogn 17:863–876
23. CritchleyM(1953)Theparietallobes.Hafner,NewYork
24. Hécaen H (1972) Introduction à la neuropsychologie. Larousse, Paris
25. Edelman GM (2004) Wider than the sky: the phenomenal gift of consciousness. Yale
UniversityPress,NewHaven
76 Y. Ohigashi and M. Yamada
26. CeredaC,GhikaJ,MaederPetal(2002)Strokesrestrictedtotheinsularcortex.Neurology
59:1950–1955
27. Karnath HO, Baier B, Nagele T (2005) Awareness of the functioning of one’s own limbs
mediatedbytheinsularcortex?JNeurosci25:7134–7138
28. Baier B, Karnath H-O (2008) Tight link between our sense of limb ownership and self-
awareness of actions. Stroke 39:486–488
29. Halligan PW, Marshall JC (1995) Supernumerary phantom limb after right hemispheric
stroke.JNeurolNeurosurgPsychiatry59:341–342
Neurological and Psychological Forms
of Amnesia
Michael D. Kopelman
Introduction
onset of an illness or injury. Retrograde amnesia (RA) involves the loss of memory
for episodes or facts that occurred before the onset of an illness or injury.
Primary or working memory (“short-term memory” in psychological parlance)
consists of memories lasting a few seconds or as long as they are actively rehearsed
and held in consciousness – what Williams James called the “specious present” [1].
This type of memory is characteristically preserved in the amnesic syndrome.
Autobiographical memory involves a person’s recollection of past incidents or
events, which occurred at a specific time and place, such that the person can “travel
back mentally in time.” The term autobiographical memory is sometimes used
interchangeably with “episodic memory,” although the latter also includes a person’s
performance on standard learning tasks that may have a differing neurobiological
basis [2]. By definition, autobiographical or episodic memory is severely impaired in
amnesia. Semantic memory refers to a person’s knowledge of language, concepts, and
facts that do not have a specific location in time or place; this is variably affected in
amnesia. Implicit memory refers to learning without awareness, and this incorporates
both procedural (perceptuo-motor) skill learning and the response to priming. Implicit
memory is characteristically spared in amnesia, as is discussed below. In principle,
episodic, semantic, and implicit memory can each have an anterograde and a retro-
grade component. Figure 1 shows that they can be differentially affected in memory
disorders: the amnesic syndrome principally affects episodic (autobiographical)
memory, sometimes exclusively so; semantic dementia principally involves semantic
AMNESIC STATES
memory; and Alzheimer dementia usually affects both the episodic and semantic
components of memory, and eventually implicit skills may also be affected.
Transient epileptic amnesia (TEA) refers to the minority of patients with TGA in
whom epilepsy appears to be the underlying cause of the syndrome [3]. The term was
originally coined by Kapur [9]. The main predictive factors for an underlying epileptic
etiology in TGA are brief episodes of memory loss, lasting an hour or less, and a
multiplicity of attacks [3]. It is important to note that standard electroencephalography
(EEG) and computed tomography (CT) findings are often normal. However, an epilep-
tic basis to the disorder may be revealed on a sleep-deprived EEG recording [3, 10].
TEA patients may show residual memory deficits between their attacks, pre-
sumably related to their underlying neuropathology. Kopelman et al. found a
moderate degree of residual anterograde memory impairment, which was probably
related to small foci of MRI signal alteration and bilateral medial temporal hypo-
metabolism on PET in the medial temporal lobes bilaterally [10]. Such patients
quite commonly also report “gaps” in their past personal memories [11]. Manes
et al. and Butler and Zeman have reported abnormal long-term forgetting of newly
learned material, which they attributed to hippocampal dysfunction [11, 12].
However, forgetting rates are extremely difficult to measure across long intervals,
and these studies have been confounded by “ceiling” and “floor” effects, such that
the apparently fast forgetting may have reflected impaired initial acquisition of the
material. Similarly, disproportionate RA has been claimed, but whether a patient’s
reported gaps in past memories result from faulty initial encoding of those memo-
ries (because of subclinical ictal activity), impaired memory consolidation, or defi-
cits in memory retrieval remains highly controversial and difficult to disentangle.
Epilepsy can, of course, give rise to automatisms or postictal confusional states.
Such automatisms appear always to be associated with bilateral involvement of those
medial temporal brain structures involved in memory formation. Hence, amnesia for
the period of automatic behavior is always present and is usually complete; claims
of automatism in the absence of significant amnesia should be rejected.
Neurological and Psychological Forms of Amnesia 81
The amnesic syndrome can be defined as “an abnormal mental state in which
memory and learning are affected out of all proportion to other cognitive functions
in an otherwise alert and responsive patient” [13]. Please note that there is no
reference to short-term memory in this definition and no reference to confabulation.
Moreover, many patients may suffer from variable degrees of specific memory
impairment (in the absence of generalized cognitive impairment), but not so
severely as to constitute an amnesic syndrome; such patients are poorly categorized
within the International Classification of Diseases.
The critical lesions that give rise to AA involve pathology in the medial temporal
lobes (hippocampi, parahippocampal gyri), thalami, mammillary bodies, the mam-
millo-thalamic tract, the retrosplenium, and the fornix. Through their interconnections
with these limbic circuits, pathology in the basal forebrain (particularly the septal
nucleus) and elsewhere within the frontal lobes can also give rise to anterograde
memory impairment. The neuropathological basis of RA is much more controversial,
some theorists arguing that damage to the hippocampi (and their connections) is
critical [14, 15], whereas others have argued that it is damage beyond the medial
temporal lobes which is critical for RA [16, 17].
The diseases or disorders that give rise to an amnesic syndrome are those which
implicate these critical brain structures. Hence, bilateral medial temporal lobec-
tomy in the patient HM gave rise to profound amnesia [18], and bilateral damage
is now a well-recognized reason for avoiding temporal lobe surgery in epilepsy.
Herpes encephalitis or voltage-gated potassium channel pathology gives rise to
structural damage in the medial temporal lobes and resulting memory disorder [19];
and profound hypoxia or hypoxic/ischemic insufficiency can also give rise to hip-
pocampal damage and amnesia [20]. By contrast, thiamine deficiency (for example,
in the alcoholic Korsakoff syndrome) particularly affects the thalami, the mammillary
bodies, and the mammillo-thalamic tract. Vascular infarction in branches of the pos-
terior cerebral artery can affect either the thalami, the hippocampi, or both, and
subarachnoid hemorrhage sometimes produces profound amnesia by damage to the
septal nucleus. Head injury, deep midline tumors, and TB meningitis can all
produce amnesia by effects upon the medial temporal, diencephalic, or frontal
structures of the brain.
The term Korsakoff syndrome should nowadays be reserved for cases of the
amnesic syndrome resulting from thiamine deficiency. These cases are almost
invariably a consequence of alcohol misuse [21].
There is a profound AA, and a RA that extends back at least 20–25 years and
involves autographical, personal semantic, and more general knowledge of public
82 M.D. Kopelman
information [22]. Korsakoff noted these patients had severe disorientation in time,
in particular, difficulty in remembering the temporal sequence of events [23]. He also
noted his patients’ preserved reasoning and other skills. Confabulation is not
necessarily present, and it often consists of the inappropriate retrieval of fragments
of real memory, jumbled up and confused in temporal context, as various others
have also noted [13].
Many cases of the Korsakoff syndrome are diagnosed following an acute
Wernicke encephalopathy, involving confusion, ataxia, nystagmus, and opthal-
moplegia. Usually, not all these features are present, and the opthalmoplegia in
particular responds rapidly to treatment with high-dose vitamins. These features
are often also associated with a peripheral neuropathy. However, the Korsakoff
syndrome can alternatively have an insidious onset, and such slower-onset
cases are more likely to come to the attention of psychiatrists or clinical
psychologists. In such cases, there may be either no known history of Wernicke
features, or only a transient history of such signs. Moreover, reports from
Scandinavia and Australia indicate that the characteristic Wernicke–Korsakoff
neuropathology (see following) is found much more commonly at autopsy in
alcoholics than the diagnosis is made in life, implying that many cases are
missed [24, 25].
Victor et al. reported that 25% of patients with the Korsakoff’s syndrome
“recover,” 50% show improvement through time, and 25% remain unchanged [13].
Although it is unlikely that any established patient shows complete recovery, the
present author’s experience is that a substantial improvement does occur over a
matter of years if the patient remains abstinent. In such cases, it is probably correct
to say that 75% of these patients show a variable degree of improvement while 25%
show no change [21].
The characteristic neuropathology in the Wernicke–Korsakoff syndrome consists
of neuronal loss, microhemorrhages, and gliosis in the paraventricular and the
periaqueductal grey matter [13]. There has been considerable debate concerning
which particular lesions are critical for the presence of chronic memory disorder
(Korsakoff syndrome). Victor et al. argued that damage in the medial dorsal nucleus
of the thalamus was critical in that patients who lacked pathology within this
specific site showed only a transient Wernicke state [13]. However, Mair et al. and
Mayes et al. argued that it was pathology within the mammillary bodies, the
midline and anterior portions of the thalamus, but not the medial-dorsal nuclei,
which was critical for memory disorder, and they based this observation on careful
ante mortem neuropsychological and post mortem neuropathological assessments
[26, 27]. More recently, Harding et al. compared eight patients who had suffered a
persistent Korsakoff syndrome with five patients who experienced only a transient
Wernicke episode: they argued that pathology in the anterior principal thalamic
nuclei was the critical difference between these two groups of patients [28]. Taken
together, these various findings suggest that the connections between the mammillary
bodies, the mammillo-thalamic tract, and the anterior thalamus may be more
important to memory dysfunction than pathology within the medial dorsal nucleus
of the thalamus.
Neurological and Psychological Forms of Amnesia 83
Herpes Encephalitis
Herpes encephalitis (HSE) can give rise to a particularly severe form of the amnesic
syndrome as a result of devastating damage to the medial aspects of the temporal
lobes [32, 33].
Many cases are said to be primary infections, although others may involve a
reactivation of the virus after it has lain dormant in neural tissue. Characteristically,
the disorder commences with a fairly abrupt onset of acute fever, headache, and
nausea. There may be accompanying behavioral changes, and seizures can occur.
Later, there is neck rigidity, vomiting, and motor and sensory deficits. However,
some cases commence more insidiously with behavioral change or psychiatric
phenomena, the confusion and neurological features becoming evident only much
later. These latter cases are those most likely to be missed, which is now important
because antiviral agents, such as acyclovir, are most effective when administered
within the first 48 h of symptoms [34, 35]. Diagnosis is by polymerase chain reac-
tion (PCR) test or a raised titer of antibodies in the cerebrospinal fluid (CSF), but a
presumptive diagnosis sometimes has to be made on the basis of the clinical picture
as well as severe signal alteration and subsequent atrophy in the medial temporal
lobes on MRI.
Neuropathological and neuroimaging studies usually show extensive bilateral
temporal lobe damage (signal alteration, volume loss, and hemorrhage), although
occasionally the changes are surprisingly unilateral [29, 36]. There may be frontal
changes, often in the orbitofrontal regions, and there may be focal changes else-
where as well as a variable degree of general cortical atrophy. The medial temporal
lobes are usually particularly severely affected, resulting in a profound AA, usually
accompanied by an extensive retrograde memory loss. Autobiographical and epi-
sodic memory are particularly severely affected. Widespread damage to the left
temporal lobes characteristically affects aspects of semantic memory, the patients
often showing severe impairment in naming and word-finding, reading (a so-called
surface dyslexia, in which the reading of irregularly spelled words is particularly
affected), and impairments in comprehension. Right temporal lobe damage can
result in particularly severe impairment in face recognition memory knowledge of
people and/or remote autobiographical memory loss [37].
Wilson et al. have described a particularly severe case of amnesia following
herpes encephalitis [33]. In this case, there was a long delay before the prescription
84 M.D. Kopelman
of an antiviral agent, resulting in extensive left temporal lobe and right medial
temporal lobe damage, evident on a quantified, structural MRI brain scan. An
interesting feature of this patient was his relatively preserved skill as a musician
(procedural/implicit memory). Despite this, he lacked any memory of having used
his musical skills recently (episodic memory); when he was shown a video of him-
self playing the piano, he stated that he had not been “conscious” at the time that
the video-clip was recorded. Indeed, for many years, he stated several times a day
that he had “just woken up,” and he also wrote this frequently in his diary.
Confabulation
Psychogenic Fugue
from just one particularly happy period of his life. This patient recovered his
memories after watching a funeral on television a few days after the onset. Glisky
et al. described a German who lost his memories in Tucson, Arizona, following
offenses in Germany and being thrown out of accommodation in Tucson [53]. He
appeared not to retain any memory of his past, or of the German language. However,
he did show implicit knowledge of German on a paired-associate test, and he also
manifested increased galvanic skin responses to personal information of which he
denied “explicit” knowledge. A functional MRI (fMRI) task involving German
words indicated relatively diminished frontal lobe activation.
Situation-Specific Amnesia
been malingering their amnesia. Of the other cases who had claimed amnesia, 33%
reported complete return of their memories at 3 years post conviction, 26% had
partial recovery of their memories, and 41% reported no change in their amnesia.
Conclusions
References
11. Butler CR, Zeman A (2008) Recent insights into the impairment of memory in epilepsy: a
systematic review of transient epileptic amnesia, accelerated long-term forgetting and remote
memory impairment. Brain 131:2243–2263
12. Manes F, Graham KS, Zeman A et al (2005) Autobiographical amnesia and accelerated for-
getting in transient epileptic amnesia. J Neurol Neurosurg Psychiatry 76:1387–1391
13. Victor M, Adams RD, Collins GH (1971) The Wernicke-Korsakoff syndrome. Davis,
Philadelphia
14. Nadel L, Moscovitch M (1997) Memory consolidation, retrograde amnesia and the hippocam-
pal complex. Curr Opin Neurobiol 7:217–227
15. Moscovitch M, Nadel L, Winocur G et al (2006) The cognitive neuroscience of remote epi-
sodic, semantic and spatial memory. Curr Opin Neurobiol 16:179–190
16. Bayley PJ, Gold JJ, Hopkins RO et al (2005) The neuroanatomy of remote memory. Neuron
46:799–810
17. Bright P, Buckman J, Fradera A et al (2006) Retrograde amnesia in patients with hippocam-
pal, medial temporal, temporal lobe, or frontal pathology. Learn Mem 13:545–557
18. Scoville WB, Milner B (1957) Loss of recent memory after bilateral hippocampal lesions. J
Neurol Neurosurg Psychiatry 20:11–21
19. Vincent A, Buckley C, Schott JM et al (2004) Potassium channel antibody-associated enceph-
alopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain
127:701–712
20. Caine D, Watson JD (2000) Neuropsychological and neuropathological sequelae of cerebral
anoxia: a critical review. J Int Neuropsychol Soc 6:86–99
21. Kopelman MD, Thomson A, Guerrini I et al (2009) The Korsakoff syndrome: clinical aspects,
psychology and treatment. Alcohol Alcohol 44:148–154
22. Kopelman MD (1989) Remote and autobiographical memory, temporal context memory, and
frontal atrophy in Korsakoff and Alzheimer patients. Neuropsychologia 27:437–460
23. Korsakoff SS (1889) Psychic disorder in conjunction with peripheral neuritis (translated and
republished: Victor M, Yakovlev PI (1955)) Neurology 5:394–406
24. Harper C (2009) The neuropathology of alcohol-related brain damage. Alcohol Alcohol
44:136–140
25. Torvik A, Lindboe C, Rogde S (1982) Brain lesions in alcoholics: a neuropathological study.
J Neurol Sci 56:233–248
26. Mair WGP, Warrington EK, Weiskrantz L (1979) Memory disorder in Korsakoff’s psychosis;
a neuropathological and neuropsychological investigation of two cases. Brain 102:749–783
27. Mayes AR, Meudell PR, Mann D et al (1988) Location of lesions in Korsakoff’s syndrome:
neuropsychological and neuropathological data on two patients. Cortex 24:367–388
28. Harding A, Halliday G, Caine D et al (2000) Degeneration of anterior thalamic nuclei dif-
ferentiates alcoholics with amnesia. Brain 123:141–154
29. Colchester A, Kingsley D, Lasserson D et al (2001) Structural MRI volumetric analysis in
patients with organic amnesia: 1. Methods and comparative findings across diagnostic groups.
J Neurol Neurosurg Psychiatry 71:13–22
30. Sullivan EV, Pfefferbaum A (2009) Neuroimaging of the Wernicke–Korsakoff syndrome.
Alcohol Alcohol 44:155–165
31. Reed LJ, Lasserson D, Marsden P et al (2003) 18FDG–PET findings in the Wernicke–Korsakoff
syndrome. Cortex 39:1027–1045
32. Hokkanen L, Launes J (2007) Neuropsychological sequelae of acute-onset sporadic viral
encephalitis. Neuropsychol Rehabil 17:429–449
33. Wilson BA, Kopelman M, Kapur N (2008) Prominent and persistent loss of past awareness in
amnesia: delusion, impaired consciousness or coping strategy? Neuropsychol Rehabil 18:527–540
34. McGrath N, Anderson NE, Croxson MC et al (1997) Herpes simplex encephalitis treated with
acyclovir: diagnosis and long term outcome. J Neurol Neurosurg Psychiatry 63:321–326
35. Raschilas F, Wolff M, Delatour F et al (2002) Outcome of and prognostic factors for herpes
simplex encephalitis in adult patients: results of a multicenter study. Clin Infect Dis
35:254–260
90 M.D. Kopelman
36. Reed LJ, Lasserson D, Marsden P et al (2005) Correlations of regional cerebral metabolism
with memory performance and executive function in patients with herpes encephalitis or
frontal lobe lesions. Neuropsychology 19:555–565
37. Kopelman MD (2002) Disorders of memory. Brain 125:2152–2190
38. Kopelman MD (1987) Two types of confabulation. J Neurol Neurosurg Psychiatry
50:1482–1487
39. Schnider A (2003) Spontaneous confabulation and the adaptation of thought to ongoing real-
ity. Nat Rev Neurosci 4:662–671
40. Gilboa A, Alain C, Stuss DT et al (2006) Mechanisms of spontaneous confabulations: a stra-
tegic retrieval account. Brain 129:1399–1414
41. Turner MS, Cipolotti T, Yousry TA et al (2008) Confabulation: damage to a specific interior
medial prefrontal system. Cortex 44:637–648
42. Kopelman MD, Ng N, Van den Brouke O (1997) Confabulation extending across episodic
memory, personal and general semantic memory. Cognit Neuropsychol 14:683–712
43. Schnider A, von Däniken C, Gutbrod K (1996) The mechanisms of spontaneous and provoked
confabulations. Brain 119:1365–1375
44. Burgess PW, Shallice T (1996) Confabulation and the control of recollection. Memory
4:359–411
45. Schacter DL, Norman KA, Koustaal W (1998) The cognitive neuroscience of constructive
memory. Annu Rev Psychol 49:289–318
46. Fotopoulou A, Solms M, Turnbull O (2004) Wishful reality distortions in confabulations: a
case study. Neuropsychologia 42:727–744
47. Fotopoulou A, Conway MA, Solms M et al (2008) Self-serving confabulation in prose recall.
Neuropsychologia 46:1429–1441
48. Dalla Barba G (1993) Confabulation: knowledge and recollective experience. Cognit
Neuropsychol 10:1–20
49. Dalla Barba G, Cappelletti YJ, Signorini M et al (1997) Confabulation: remembering
“another” past, planning “another” future. Neurocase 3:425–436
50. Berrington WP, Liddell DW, Foulds GA (1956) A re-evaluation of the fugue. J Ment Sci
102:280–286
51. Stengel E (1941) On the aetiology of the fugue states. J Ment Sci 87:572–599
52. Schacter DL, Wang PL, Tulving E et al (1982) Functional retrograde amnesia: a quantitative
case study. Neuropsychologia 5:523–532
53. Glisky EL, Ryan L, Reminger S et al (2004) A case of psychogenic fugue: I understand, aber
ich verstehe nichts. Neuropsychologia 42:1132–1147
54. Mechanic M, Resick PA, Griffin MG (1998) A comparison of normal forgetting, psychopa-
thology, and information-processing models of reported amnesia of recent sexual trauma.
J Consult Clin Psychol 66:948–957
55. Andrews B, Brewin CR, Rose S et al (2000) Predicting PTSD symptoms in victims of violent
crime: the role of shame, anger, and childhood abuse. J Abnorm Psychol 109:69–73
56. Taylor PJ, Kopelman MD (1984) Amnesia for criminal offences. Psychol Med 14:581–588
57. Kopelman MD (2002) Psychogenic amnesia. In: Baddeley AD, Kopelman MD, Wilson BA
(eds) The handbook of memory disorders, 2nd edn. Wiley, Chichester
58. Pyszora NM, Barker AF, Kopelman MD (2003) Amnesia for criminal offences: a study of life
sentence prisoners. J Forens Psychiatry Psychol 14:475–490
59. Christianson SA, Merckelbach H, Kopelman MD (2006) Crime-related amnesia. In: Heaton-
Armstrong A, Shepherd E, Gudjonsson G et al (eds) Witness testimony: psychological, inves-
tigative and evidential perspectives. Oxford University Press, Oxford, pp 105–126
60. Gudjonsson GH, MacKeith JAC (1983) A specific recognition deficit in case of homicide.
Med Sci Law 23:37–40
61. Kopelman MD (2000) Focal retrograde amnesia and the attribution of causality: an exception-
ally critical review. Cognit Neuropsychol 17:585–621
Neurological and Psychological Forms of Amnesia 91
62. Kapur N (2000) Focal retrograde amnesia and the attribution of causality: an exceptionally
benign commentary. Cognit Neuropsychol 17:623–637
63. Kapur N (1993) Focal retrograde amnesia in neurological disease: a critical review. Cortex
29:217–234
64. Anderson MC, Green C (2001) Suppressing unwanted memories by executive control. Nature
(Lond) 410:366–369
65. Anderson MC, Ochsner KN, Kuhl B et al (2004) Neural systems underlying the suppression
of unwanted memories. Science 303:232–235
The Neuropsychological Aspect of Epilepsy
Yoshio Morita
Introduction
Instudiesofhigherbrainfunction,clinicalneuropsychologyandclinicalepileptology
are closely related. Both sciences aim at the elucidation of local brain functions:
clinical neuropsychology presents syndromes of functional loss and stimulation
associated with local injuries of the human brain, whereas various syndromes of
human epileptic seizures are observed in clinical epileptology.
Studies attempt to elucidate the association between changes in brain function
induced in the cerebral disease process and the psychological process employing
methods supplementing each other. Clinical brain wave measurement including
evoked responses and psychological measurement are employed in this study.
Paroxysmal neuropsychological symptoms; visuo-spatial disorders and body-scheme
Y. Morita (*)
DepartmentofNeuropsychiatry,HyogoCollegeofMedicine,
1-1,Mukogawa-cho,Nishinomiya,663-8501,Hyogo,Japan
e-mail: [email protected]
AccordingtoW.G.Lennox,epilepsyisdefinedasrecurrentself-sustainedparoxys-
mal disorders of brain function.
Epilepsy encompasses paroxysmal disorders, and the international classification
categorizes epileptic seizures as follows [1]:
1. Localization-related(focal,local,andpartial)epilepsiesandsyndromes
(a) Idiopathic(withage-relatedonset)
(b) Symptomatic
(c) Cryptogenic
2. Generalizedepilepsiesandsyndromes
3. Epilepsiesandsyndromesundeterminedastowhetherfocalorgeneralized
4. Specialsyndromes
Temporal lobe epilepsy, one of the types of complex partial seizures, is classified as
symptomatic epilepsy in this classification [1]. The paroxysmal symptoms of this
type of epilepsy are closely related to anatomical localization of the lesion in the
temporal lobe.
In addition to paroxysmal symptoms, accompanying signs and symptoms at
onset are important indices of the origin of paroxysmal discharge. On the other
hand, classical clinical neuropsychology searches for associations between func-
tional changes in brain disorders and psychological processes.
Neurologicalsymptoms;essentialsensationandmovementdisorders,aswellas
neuropsychological symptoms; aphasia, apraxia, and agnosia, are common symp-
toms in epileptic patients. On the other hand, neuropsychiatric symptoms, such as
hallucination, disturbance of memory, spontaneity disorder, personality disorder
are less frequently noted in epilepsy.
Fifty-sevenepisodesofaurasensationsin18patientswithtemporallobeepilepsy
aretabulatedinTable1and2.
The main symptoms of temporal lobe syndrome in temporal lobe epilepsies
(TLEs)aresimpleandcomplexpartialseizures,secondarilygeneralizedseizures,
or a combination of these symptoms.
Complex partial seizure can be categorized into P0–P4 based on the phasal
developmentofseizures;P0aura-sensations,P1representsthetonic-lapsephase,
P2 oral automatism, P3 behavioral automatism, and P4 secondarily generalized
tonic-clonic seizures. Aura-sensations of visuo-spatial distortions were noted in the
P0stageof18patientswithtemporallobeepilepsy.
TheNeuropsychologicalAspectofEpilepsy 97
which69patientsunderwentelectricstimulationduringbrainsurgery.Itshowed
another characteristic in that it concomitantly occurred with other visuo-spatial
distortions [4].
Penfield and Perot [5] reported the case of patient with heautoscopy. Their case
wasa37-year-oldhousewife,whobegantohaveseizuresattheageof20,consist-
ingofthefollowingpattern:(1)thoracicsensationandpalpitation,(2)experiential
hallucination,visualtype,and(3)occasionalmajorseizures,mainlyrightsided.
At the beginning of an attack, she would mentally visualize scenes that she some-
times had difficulty recognizing or remembering, but they were often from her
own past. Frequently, the scene consisted of herself during childbirth and also in
a picture. Often, as soon as she recognized the scene it would disappear. On sur-
gery, the left temporal lobe was exposed, and an area of gliosis identified.
StimulationatpointN(Brodmann21)causedthepatienttosaythatshesuddenly
TheNeuropsychologicalAspectofEpilepsy 99
Table 4 Interpretiveresponsesandillusions(fromPenieldandPerot[5])
Auditory illusions: Sounds heard seemed louder or clearer, fainter or more distinct, nearer or
farther
Visual illusions: Things seen seemed clearer or blurred; nearer or farther; larger or smaller;
fatter or thinner
Illusionsofrecognition:Presentexperienceseemedfamiliar(déjàvu),strange,altered,orunreal
Illusionalemotions:Feelingsoffear,loneliness,sorrow,ordisgust
Table 5 Ictalautoscopyhallucination[6]
Author Focus
1 Nouet 1923 r. temporal
2 Ehrenwald 1931 r. central
3 Lunn 1948 r. central
4 Lunn 1948 l. central
5 Hecaen and Ajuriaguerra 1952 l. centro-parieto occipital
6 Ionasescu 1960 l. temporal-occipital
7 Ionasescu 1960 r. temporal
8 Ohashi 1965 r. temporal
9 Hamanaka 1974 bil. diffuse
10 Morita 1978 r. temporal
11 Sengoku 1981 r. temporal
12 Kamiya and Okamoto 1982 r. temporal
13 Kamiya and Okamoto 1982 l. temporal
14 Kamiya and Okamoto 1982 r. temporal
saw herself in childbirth and she felt as if she were reliving the experience [5]
(Table 4).
Here the brief history of our patient with heautoscopy is presented.
Thefemalepatientof26yearsoldwhosufferedwithaura-sensationofcomplex
partial seizure of temporal lobe epilepsy complaint the frequent episodes of heau-
toscopicexperiencesandinvertedvision.Inter-ictalscalp-recordelectroencepha-
logram showed the spike and slow wave complex at left anterior temporal lobe of
thispatient[6](Table5).
Ictalautoscopyhallucinationisobservedfrequentlyinthepatientswithtemporal
lobe epilepsy. This peculiar experience is considered one of the feeling of alteration of
body scheme and asomatognosia.
epilepsy [2,3]. And, occasionally the symptoms are accompanied by the state of
double consciousness [7]. Neuropsychological symptoms, like aphasia, heautos-
copy[8],andinvertedvision,distortionofthetimesense[9]arealsoencountered
in the complex partial seizures.
Heautoscopy is regarded as one of the disorder of body scheme (asomatognosia)
[8](Table6).Paroxysmalepisodesofthepeculiarexperienceofseeingone’sself
or “double” self (heautoscopy) may occur in the patients with temporal lobe epilepsy.
In the brain stimulation study reported by Penfield and Perot [5], heautoscopy
was reproducedin4of69cases,andthefocioftheseizureswerepresentinthe
temporal lobe, similarly to our patients.
As we see in the present chapter, neuropsychological symptoms commonly occur
in paroxysmal seizures of the complex partial epilepsy, especially temporal lobe
epilepsy. Therefore, collaborative studies in the fields of epileptology and neuropsy-
chology are important to understand the complexity of the psychiatric symptoms in
temporal lobe epilepsy.
Although the complete understanding of human brain function might not be pos-
sible at this moment, it should be a goal of the clinical neurosciences in the future.
There is no doubt, however, neuropsychological studies of epileptic patients con-
tribute to understand the brain function, as the encourage words of Penfield and
Jackson:“Hewhoisfaithfullyanalyzingmanydifferentcasesofepilepsyisdoing
farmorethanstudyingepilepsy”(WilderPenfieldandHughlingeJackson).
References
7. KamiyaS,OkamotoS(1982)Doubleconsciousnessinepileptics:aclinicalpictureandminor
hemisphericspecialization.Advancesinepileptology.Raven,NewYork
8. OhhashiH,KawaiI,HamanakaT(1969)Aclinicalandelectroencephalographicstudyonthe
paroxysmaldisordersofbodyimage.PsychiatrNeurolJpn71:435–449
9. KawaiI,KawagoeT,HatadaKetal(1983)Ictaldistortionoftimesenseinepilepticpatients.
JJpnEpilepsySoc1:17–22
The Interictal Dysphoric Disorder of Epilepsy
Marco Mula
It has been known for a long time that there are some associations between epilepsy
and depression. The Greek physician Hippocrates, around 400 b.c., observed that
“melancholics ordinarily become epileptics, and epileptics, melancholics: what
M. Mula (*)
Division of Neurology, Maggiore Hospital, Amedeo Avogadro University,
C.so Mazzini 18, 28100, Novara, Italy
e-mail: [email protected]
determines the preference is the direction the malady takes; if it bears upon the
body, epilepsy, if upon the intelligence, melancholy” [1].
There are several reasons why these two conditions may be closely linked, and
suchreasonsarebothbiologicalandpsychosocial.Epilepsyisachronicdisorder,
which still brings about much social discrimination, and as with any chronic disor-
der, epilepsy might be expected to be linked to demoralization and a negative
perspective toward life. On the other hand, the biological contribution to the asso-
ciation, based on neuroanatomical and neurochemical principles, needs to be taken
intoaccount.Epilepsyisfrequentlyassociatedwithdamagesinthelimbicsystem,
which plays a key role in the processing of emotions. Also, patients with epilepsy
are chronically exposed to antiepileptic drugs that are potent psychoactive com-
pounds with a number of effects on mood and behavior. For all these reasons it is
expected that depression is more frequent among patients with epilepsy when com-
pared to the general population, and data coming from community studies support
such an impression. The General Practice UK study reported a 22% prevalence of
depression in unselected samples of patients with epilepsy [2]. A Canadian
Community Health Survey noted very similar lifetime prevalence rates for depression
(around 22%), much higher than those reported in the general population (around
12%) [3]. A U.S. survey pointed out that depression is more frequent in epilepsy
(36.5%) in comparison to people with asthma (27.8%) and healthy controls [4], sug-
gesting that depression is more frequently reported in epilepsy than in other chronic
medical conditions. Another relevant point relates to seizure control being such
comorbidity greater in those patients with higher seizure frequencies and with con-
tinuing seizures compared to seizure-free patients. In fact, several authors have
noted a correlation with seizure frequency that most likely reflects in a large part
on the intractability of the seizure disorders. Jacoby et al. [5] reported depression
in 4% of seizure-free patients, 10% in patients with less than one seizure per month,
and 21% in patients with higher seizure frequency. O’Donoghue et al. [6] pointed
out that patients with epilepsy with continuing seizures are significantly more likely
to suffer from depression than those in remission (33% vs. 6%).
The association between depression and epilepsy has relevant implications in
terms of prognosis and treatment; in fact, it seems quite established that depressive
symptoms are the most important predictor of quality of life of patients with epilepsy,
an even more powerful predictor than the actual seizure frequency [7–9].
An important finding that has been replicated by several studies is that the link
between depression and epilepsy is not necessarily unidirectional; that is, patients
with such a comorbidity always present with the seizure disorder before the emer-
gence of the depression, but it has been noted that having a prior mood disorder can
be associated with an increased risk of epilepsy [10, 11]. Although it is reasonable to
hypothesize that the development of the epilepsy may be related to suicidal attempts,
or to drug abuse, or follow some other kinds of trauma such as head trauma, it
tempting to speculate that such a finding may reflect an underlying common patho-
genesis, which may relate to some as yet unknown genetic factor, or some link with
neurotransmitter function (for example, related to transmitters that are known to play
aroleinbothepilepsyanddepressionsuchasserotoninorGABA)[12, 13].
TheInterictalDysphoricDisorderofEpilepsy 105
The issue of phenomenology of depression in epilepsy has been and still is very
much a matter of debate. Some authors have emphasized the endogenous features
[21] while others commented on the reactive nature of depression [22]. In general
terms, the spectrum of manifestations is likely to be large, and it is reasonable to
hypothesize that patients with epilepsy can experience forms of mood disorders
identical to those of patients without epilepsy. However, it is equally reasonable to
assume that the underlying brain pathology may influence the final phenomenology
of mood disorder symptoms, emphasizing some aspects or attenuating others.
Data favoring the existence of an epilepsy-specific mood disorder come from the
clinical observation that the psychopathology of patients with epilepsy often has
unique manifestations that are poorly reflected by conventional classification
systems such as Diagnostic and Statistical Manual of Mental Disorders (DSM)-IV
106 M. Mula
Fig. 1 Major DSM-IV Axis I diagnoses overlapping with the interictal dysphoric disorder
The features of the so-called interictal dysphoric disorder overlap with a variety
of affective disorders seen in clinical psychiatric practice (Fig. 1); however, data
presented suggest that such a syndrome represents a frequent comorbidity in
patients with epilepsy and its diagnosis can be challenging even for expert clini-
cians because of the pleomorphic phenomenology of the syndrome.
Conclusions
References
1. TemkinO(1971)Thefallingsickness.JohnHopkins,Baltimore
2. EdehJ,TooneB(1987)Relationshipbetweeninterictalpsychopathologyandtypeofepilepsy.
Resultsofasurveyingeneralpractice.BrJPsychiatry151:95–101
3. Tellez-Zenteno JF, Patten SB, Jetté N et al (2007) Psychiatric comorbidity in epilepsy: a
population-basedanalysis.Epilepsia48:2336–2344
4. EttingerA,ReedM,CramerJetal(2004)Depressionandcomorbidityincommunity-based
patients with epilepsy or asthma. Neurology 63:1008–1014
5. JacobyA,BakerGA,SteenNetal(1996)Theclinicalcourseofepilepsyanditspsychosocial
correlates:findingsfromaUKcommunitystudy.Epilepsia37:148–161
6. O’Donoghue MF, Goodridge DM, Redhead K et al (1999) Assessing the psychosocial conse-
quencesofepilepsy:acommunity-basedstudy.BrJGenPract49:211–214
7. PerrineK,HermannBP,MeadorKJetal(1995)Therelationshipofneuropsychologicalfunc-
tioning to quality of life in epilepsy. Arch Neurol 52:997–1003
8. GilliamF,KuznieckyR,FaughtEetal(1997)Patientvalidatedcontentofepilepsyspecific
qualityoflifemeasurement.Epilepsia38:233–236
9. BoylanLS,FlintLA,LabovitzDLetal(2004)Depressionbutnotseizurefrequencypredicts
quality of life in treatment-resistant epilepsy. Neurology 62:258–261
10. Forsgren L, Nystrom L (1990) An incident case-referent study of epileptic seizures in adults.
EpilepsyRes6:66–81
11. Hesdorffer DC, Hauser WA, Annegers JF et al (2000) Major depression is a risk factor for
seizures in older patients. Ann Neurol 47:246–249
12. Kanner AM, Balabanov A (2002) Depression and epilepsy: how closely related are they?
Neurology 58(8 suppl 5):S27–S39
13. Kanner AM (2006) Depression and epilepsy: a new perspective on two closely related disor-
ders.EpilepsyCurr6:141–146
14. Quiske A, Helmstaedter C, Lux S et al (2000) Depression in patients with temporal lobe
epilepsyisrelatedtomesialtemporalsclerosis.EpilepsyRes39:121–125
TheInterictalDysphoricDisorderofEpilepsy 111
15. SwinkelsWA,vanEmdeBW,KuykJetal(2006)Interictaldepression,anxiety,personality
traits,andpsychologicaldissociationinpatientswithtemporallobeepilepsy(TLE)andextra-
TLE.Epilepsia47:2092–2103
16. BremnerJD,NarayanM,AndersonERetal(2000)Hippocampalvolumereductioninmajor
depression. Am J Psychiatry 157:115–118
17. FrodlT,MeisenzahlEM,ZetzscheTetal(2002)Hippocampalchangesinpatientswithafirst
episode of major depression. Am J Psychiatry 159:1112–1118
18. HermannBP,SeidenbergM,HaltinerAetal(1991)Moodstateinunilateraltemporallobe
epilepsy.BiolPsychiatry30:1205–1218
19. SchmitzEB,MoriartyJ,CostaDCetal(1997)Psychiatricprofilesandpatternsofcerebral
blood flow in focal epilepsy: interactions between depression, obsessionality, and perfusion
related to the laterality of the epilepsy. J Neurol Neurosurg Psychiatry 62:458–463
20. Bromfield E, Altschuler L, Leiderman D (1990) Cerebral metabolism and depression in
patientswithcomplexpartialseizures.Epilepsia31:625
21. BettsTA(1974)Afollowupstudyofacohortofpatientswithepilepsyadmittedtopsychiat-
riccareinanEnglishcity.In:HarrisP,MawdsleyC(eds)Epilepsy:ProceedingsoftheHans
BergerCentenarySymposium.ChurchillLivingstone,Edinburgh,pp326–338
22. Mulder DW, Daly D (1952) Psychiatric symptoms associated with lesions of the temporal
lobe. JAMA 141:173–176
23. Krishnamoorthy ES, Trimble MR, Blumer D (2007) The classification of neuropsychiatric
disorders in epilepsy: a proposal by the ILAE Commission on Psychobiology of Epilepsy.
EpilepsyBehav10:349–353
24. Mendez MF, Cummings JL, Benson DF (1986) Depression in epilepsy. Significance and
phenomenology. Arch Neurol 43:766–770
25. Kanner AM, Kozac AM, Frey M (2000) The use of sertraline in patients with epilepsy: is it
safe?EpilepsyBehav1:100–105
26. Kanner AM, Soto A, Gross-Kanner H (2004) Prevalence and clinical characteristics of pos-
tictal psychiatric symptoms in partial epilepsy. Neurology 62:708–713
27. Kanner AM (2008) Do peri-ictal psychiatric symptoms account for the differences between
depressivedisordersinpatientswithandwithoutepilepsy?In:KannerAM,SchachterS(eds)
Psychiatriccontroversiesinepilepsy.Elsevier,NewYork,pp201–209
28. KraepelinE(1923)Psychiatrie.Band3.JohannAmbrosiusBarth,Leipzig
29. BleulerE(1949)LehrbuchderPsychiatrie,8thedn.Springer-Verlag,Berlin
30. GastautH,MorinG,LesevreN(1955)Etudeducomportementdesepileptiquespsychomo-
teurs dans l’intervalle de leurs crises: les troubles de l’activitè globale et de la sociabilitè. Ann
Med Psychol (Paris) 113:1–27
31. BlumerD(2000)Dysphoricdisordersandparoxysmalaffects:recognitionandtreatmentof
epilepsy-related psychiatric disorders. Harv Rev Psychiatry 8:8–17
32. HimmelhochJM(1984)Majormooddisordersrelatedtoepilepticchanges.In:BlumerD(ed)
Psychiatric aspects of epilepsy. American Psychiatric, Washington, DC, pp 271–94
33. Kanner AM (2003) Depression in epilepsy: a frequently neglected multifaceted disorder.
EpilepsyBehav4:S11–S19
34. Blumer D, Montouris G, Davies K (2004) The interictal dysphoric disorder: recognition,
pathogenesis, and treatment of the major psychiatric disorder of epilepsy. Epilepsy Behav
5:826–840
35. Mula M, Jauch R, Cavanna A et al (2008) Clinical and psychopathological definition of the
interictaldysphoricdisorderofepilepsy.Epilepsia49:650–656
36. MulaM,SchmitzB,JauchRetal(2008)Ontheprevalenceofbipolardisorderinepilepsy.
EpilepsyBehav13:658–61
37. BlumerD,HeilbronnM,HimmelhochJ(1988)Indicationsforcarbamazepineinmentalill-
ness: atypical psychiatric disorder or temporal lobe syndrome? Compr Psychiatry
29:108–122
38. Möller HJ, Curtis VA (2004) The bipolar spectrum: diagnostic and pharmacologic consider-
ations.ExpertRevNeurother4(6suppl2):S3–8
112 M. Mula
39. BenazziF(2007)Isthereacontinuitybetweenbipolaranddepressivedisorders?Psychother
Psychosom 76:70–6
40. Hirschfeld RM, Holzer C, Calabrese JR et al (2003) Validity of the mood disorder question-
naire: a general population study. Am J Psychiatry 160:178–180
41. Akiskal HS, Pinto O (1999) The evolving bipolar spectrum. Prototypes I, II, III, and IV.
Psychiatr Clin North Am 22:517–34
42. Judd L, Akiskal HS (2003) The prevalence and disability of bipolar spectrum disorders in the
U.S.population:re-analysisoftheECAdatabasetakingintoaccountsubthresholdcases.
J Affect Disord 73:123–131
43. KrishnamoorthyES(2006)Theevaluationofbehavioraldisturbancesinepilepsy.Epilepsia
47(suppl 2):3–8
44. Mintzer S, Lopez F (2002) Comorbidity of ictal fear and panic disorder. Epilepsy Behav
3:330–337
45. Krishnamoorthy ES (2005). Epidemiology and assessment of psychiatric disorders of epi-
lepsy. Ph.D. thesis, University College London
46. JonesJE,HermannBP,WoodardJLetal(2005)Screeningformajordepressioninepilepsy
withcommonself-reportdepressioninventories.Epilepsia46:731–735
47. GilliamFG,BarryJJ,HermannBPetal(2006)Rapiddetectionofmajordepressioninepi-
lepsy: a multicentre study. Lancet Neurol 5:399–405
48. BlumerD,MontourisG,DaviesKetal(2002)Suicideinepilepsy:psychopathology,patho-
genesis,andprevention.EpilepsyBehav3:232–241
49. MulaM,TrimbleMR(2008)Whatdoweknowaboutmooddisordersinepilepsy?In:Kanner
AM,SchachterS(eds)Psychiatriccontroversiesinepilepsy.Elsevier,NewYork,pp49–66
Neuropsychiatric Symptoms of Seizure
Disorders with Special Reference
to the Amygdala
Michael Trimble
Abstract In this chapter, the anatomy and function of the amygdala in relationship
to emotional processing are explored. It is now known that the amygdala can be
activated by certain emotional stimuli, and the fearful faces paradigm has been well
tested experimentally. Studies with epilepsy reveal that amygdala sclerosis often
accompanies hippocampal sclerosis. Studies of patients with amygdala sclerosis
show that this affects the activity to emotional stimuli of the amygdala but also the
fusiform and related cortical areas distant from the amygdala. Studies of patients who
have undergone temporal lobectomy show the influence of the amygdala in relation
to both preoperative and postoperative affective states, especially of the nondominant
hemisphere. It is concluded that further studies of the amygdala in epilepsy and other
neurological disorders are valuable in studying the functions of the limbic system.
Introduction
The clinical associations between epilepsy and psychiatric syndromes have now
been widely accepted, and the term comorbidity is often used to express such a
relationship. In the past, psychological and psychosocial explanations were widely
used to explain links between such syndromes as anxiety and depression in epi-
lepsy, although the more severe syndromes such as psychoses or the controversial
personality disorders were less likely to be accounted for without considering the
underlying biology of the epilepsy [1].
Part of this confusion was the result of the almost exclusive psychological
approach to neuropsychiatric disorders taken by psychiatrists, and the relative
neglect by neurologists of behavior problems that could be associated with
M. Trimble (*)
Institute of Neurology, Queen Square, London, WC1N3BG, UK
e-mail: [email protected]
neurological disorders. Further, in the field of epilepsy there was confusion over the
distinction between seizures and epilepsy. Many people equated the two, but seizures
as signs and causing symptoms are quite distinct from the underlying pathological
processes of the epilepsy. The ongoing interictal electrophysiological disturbances,
which presumably reflect underlying electrochemical aberrations within the brain,
which are easily identifiable by various imaging techniques, may be expected to lead
to continuing interictal disturbances of cerebral function, and if these occur in areas
of the central nervous system that have an impact on emotion and behavior, then
psychiatric disturbances may be the expected outcome for at least some people with
epilepsy, depending upon the site and type of the underlying epileptic discharge.
There has been a growing literature that emphasizes not only the possible neuro-
logical underpinnings of psychiatric disorders generally, but also the psychiatric com-
plications of neurological disorders, including epilepsy. From a purely anatomical
point of view, the unravelling of the concept of the limbic system, from the earlier
circuitry proposed by Papez, to the later more sophisticated elaborations of people
such as MacLean, has emphasized that within the brain there were neuronal structures
and circuits which have specifically to do with modulation of emotion. This was a new
idea, as before the development of the limbic system concept there was no clear cere-
bral framework for an understanding of how the brain felt and expressed emotion.
It was crucial to elaborating on the link between epilepsy and emotion to realize that
two key limbic structures, the hippocampus and the amygdala, were frequently
involved in the underlying pathology of epilepsy, particularly in the localization-
related epilepsies, and newly developed techniques of recording from sites within the
brain revealed that between seizures, interictal abnormalities were recorded from such
structures. More recently, the uncovering and elaboration of the direct associations
between medial temporal structures and limbic forebrain structures, and the unraveling
of the neuroanatomy of the limbic forebrain by authors such as Heimer and colleagues,
have given clear neurological underpinnings for an understanding of the behavioral
consequences of neurological disorders, epilepsy being no exception [2].
The Amygdala
That the amygdala plays a central role in animal behaviour has been known for many
years, although its precise role, and its relative importance across different species,
have yet to be fully determined. Until recently several lines of evidence have pointed
to the role of the amygdala in human behavior, and there is considerable evidence that
this structure is closely involved with emotional responsivity in humans. Using func-
tional magnetic resonance imaging (fMRI), it has become commonplace to image the
amygdala in vivo, and there are case histories of patients who have had either damage
to the amygdala or degeneration of the amygdala, revealing behavioral problems [3].
The amygdala is located at the anterior part of the temporal lobes, in front of and
above the temporal horn of the lateral ventricle. It abuts the rostral part of the
hippocampus and is a composite of several neuronal aggregates. There are two main
components, a larger basolateral complex, which has extensive connections with the
Neuropsychiatric Symptoms of Seizure Disorders with Special Reference to the Amygdala 115
Fig. 1 The amygdala has widespread influence on cortical and subcortical structures. L lateral;
CE central; M medial; B basal; AB accesory basal nucleus; PAC periamygdaloid cortex. (Reproduced
with permission from Asla Pitkanen)
used to explore the brains of patients with epilepsy, and data from both structural
volumetric analyses and functional data using fMRI have drawn attention to the
importance of the amygdala for regulating behavior in patients with epilepsy.
Structural Changes
van Elst et al. 2000 [7, 8] carried out a series of studies examining the volume of the
amydala in different groups of patients with epilepsy. In the first investigation, they
examined a group of patients with intermittent explosive disorder (DSM4) associated
with epilepsy, comparing their data to patients with epilepsy without such behaviors.
They reported that the aggression group contained a subgroup of patients with very
Neuropsychiatric Symptoms of Seizure Disorders with Special Reference to the Amygdala 117
small amygdala, and there was a higher prevalence of amygdala sclerosis in the
aggressive patients. Twenty percent of their sample had severe amygdala atrophy in
the context of a history of encephalitis. Left-sided lesions were overrepresented.
In further studies, they looked at the volumes of the amygdala in patients with
epilepsy and psychosis. Two patient groups were examined: the first were patients
with epilepsy and interictal psychosis (n = 11), and the second had post-ictal
psychosis (n = 15). Two control groups consisted of 20 healthy volunteers and 20
randomly selected cases of temporal lobe epilepsy who had not displayed any
psychopathology; these were matched for age, sex, duration of epilepsy, and anti-
epileptic medication to the study group. The psychotic episodes were defined
according to ICD10 criteria for the paranoid subtype of schizophrenia; the mini-
mum requirement for the diagnosis of psychosis was the presence of delusions or
hallucinations. The relationship of the psychopathology to the seizures was also
determined, and the groups therefore divided into those with post-ictal and those
with interictal psychoses. Further, patients with other first-axis psychiatric disor-
ders were excluded from the study except for those with minor affective symptoms
that are common in patients with temporal lobe epilepsy.
The results from the study are shown in Fig. 2. When adjusted for cerebral size
(patients with psychoses of epilepsy had significantly smaller total brain volumes
Results
MRI-Volumetry:Amygdalavolumes
R F = 8.211; P=0.001
L ANOVA
F = 9.079; P<0.000
***
cm3 ***
**
**
2,2
2,1
1,9
1,8
1,7
1,6
1,749 1,756 1,798 1,826 2,056 2,067
CON TLE-CON TLE&POE
= right amygdala = left amygdala
Fig. 2 Results of amygdala volumetry in epileptic psychoses. MRI magnetic resonance imaging;
CON control; TLE temporal lobe epilepsy; POE psychoses of epilepsy. (From van Elst et al. [8])
118 M. Trimble
Functional Studies
they found a significant relationship between the amount of sclerosis and reduced
emotional activation, especially in the posterior fusiform and left occipital areas.
These data reveal that the visual responsiveness of the occipital cortical areas was
intact, but the enhanced emotional responses did not occur.
These data are complemented by neurophysiological studies, and also studies of
patients who have had a temporal lobe removed to treat intractable epilepsy. Vuilleumier
and Driver [12], using a similar experimental paradigm, reported that amygdala damage
reduced or completely eliminated the enhancement of the P1 component of the visual
evoked potential for fearful faces, relative to neutral faces. These findings supported the
view of the distance effects of amygdala lesions on emotional processing.
Bonelli et al. [13] examined patients with anterior temporal lobe resections hav-
ing assessed the preoperative amygdala activation of these patients using the fearful
face paradigm. The responses of patients with refractory epilepsy were compared
with healthy controls, and measures of anxiety and depression were taken preopera-
tively and 4 months postoperatively, being assessed with standardized anxiety and
depression rating scales.
Preoperatively, the patients with left temporal lobe epilepsy had significantly
reduced activation of the amygdala bilaterally, whereas patients with right temporal
lobe epilepsy showed bilateral amygdala activation. Patients with right temporal
lobe epilepsy revealed left and right amygdala activation that was correlated to the
preoperative anxiety and depression levels, and the preoperative right amygdala
activation was correlated significantly with the postoperative increases of both
anxiety and depression scores. Similar correlations were not found for patients with
left-sided temporal lobe epilepsy.
These data, in accordance with the other findings reported above, not only sup-
port the view that the amygdala is of considerable importance in relationship to the
affective state of patients with epilepsy, but they also contributed to the studies of
laterality. Thus, these data suggest that the right amygdala is of more importance in
relationship to affective disorders, in keeping with the known role of the nondomi-
nant hemisphere in relationship to control of affect, and predict that resection of the
right amygdala with patients who have surgery for epilepsy is more likely to lead
to emotional disturbances.
Conclusions
Studies of temporal lobe epilepsy have until recently concentrated much more on
the hippocampus than on the amygdala. However, particularly since the advent of
modern brain imaging, with the ability to examine not only the structure, but also
the function, of the amygdala, the role of the amygdala in emotional processing in
a variety of settings has become clear. Further, there are studies that show that
alteration of the function of the amygdala, either from structural lesions or from
epilepsy, have an influence on the processing of emotional stimuli in patients with
such pathologies.
120 M. Trimble
References
David B. Arciniegas
Abstract Traumatic brain injury (TBI) is a common and costly problem worldwide.
In the United States, 1.5 million people sustain TBI each year, and approximately
230,000 of these require hospitalization for management of their injury. The majority
of TBI resulting in hospitalization are moderate to severe in nature and produce
significant mortality and morbidity. Among those surviving their injuries, most
will develop cognitive, emotional, and behavioral (collectively referred to here as
neuropsychiatric) disturbances in the acute postinjury period and will require acute
rehabilitation management. These posttraumatic neuropsychiatric sequelae present
substantial clinical management challenges that the consulting neuropsychiatrist is
well suited to evaluate and manage. In the service of offering the consulting neuro-
psychiatrist with information that may be of use in the care of persons with TBI
receiving care in the acute neurorehabilitation setting, this chapter first defines and
describes TBI and reviews the neuroanatomical and neurobehavioral consequences
of TBI relevant to understanding posttraumatic neuropsychiatric disturbances.
These disturbances are organized under the framework of posttraumatic enceph-
alopathy, and the characteristic forms and stages of recovery of this condition are
discussed. Finally, a neuropsychiatric approach to the evaluation of persons with
TBI in the acute inpatient neurorehabilitation setting is described.
Keywords Cognitiveimpairment•Encephalophathy•Frontalassessmentbattery
(FAB)•Mini-mentalstateexam(MMSE)•Traumaticbraininjury(TBI)
*ThisworkwassupportedinpartbyHealthONESpaldingRehabilitationHospital
D.B. Arciniegas (*)
BrainInjuryRehabilitationUnit,HealthONESpaldingRehabilitationHospital,
Aurora, CO, USA
and
Neurobehavioral Disorders Program, Department of Psychiatry,
SchoolofMedicine,UniversityofColoradoDenver,13001East17thPlace,
Campus Box F546, Aurora, CO, 80045, USA
e-mail: [email protected]
Introduction
Defining TBI
Table 1 Clinical case definition of traumatic brain injury (adapted from the Center for Disease
Control Guidelines for the Surveillance of Central Nervous System Injury, 2002)
Clinical phenomena Description
Objective neurological Focal motor, sensory, or reflex abnormalities
abnormality Seizures (focal or generalized)
Alteration of consciousness Loss of consciousness (LOC)
Impairmentofwakefulness(arousal)and/orawareness
Amnesia Loss or impairment of peri-event memory
Retrograde amnesia (RGA): impaired memory for events
immediately preceding the injury
Anterograde amnesia (AGA): impaired memory for the
injury or the events that follow it
Posttraumatic amnesia (PTA): the period of dense
impairment in new learning following TBI; inclusive of
both AGA and RGA
Objective neuropsychological Standardized neuropsychological examination (in the
abnormality immediate postinjury period) reveals impairment of
cognition (e.g., disorientation, confusion), disturbances
of behavior (e.g., agitation), or other abnormalities in
neuropsychiatric status (e.g., personality change)
Diagnosed intracranial lesion On computed tomography (CT), magnetic resonance imaging
(MRI),oranotherneurodiagnostic(i.e.,neuroimaging)
study,thereisevidenceofdiffuseaxonalinjury,and/
or epidural, subdural, subarachnoid, or intracerebral
hematoma,and/orcerebralcontusionorlaceration,and/or
penetration of brain by foreign body (e.g., gunshot wound)
128 D.B. Arciniegas
TBI severity is generally divided into three categories: mild, moderate, and severe.
Among hospitalized patients, the Glasgow Coma Scale (GCS) [20] is the most com-
monlyusedmetricfordeterminingTBIseverity.Mild,moderate,andsevereTBIare
definedbyGCSscoresof13–15,9–12,and3–8,respectively,reflectingperformance
on three relatively elementary assessments of verbal, eye opening, and motor responses.
While the GCS is an excellent research and clinical measure when administered in a
consistent and timely manner [6, 21, 22], its use in many hospitals is inconsistent, at
best, and the data it yields are frequently confounded by other non-TBI factors [23].
In the absence of GCS scores, or as a supplement or complement to them, determi-
nation of the duration of PTA is also a useful gauge of TBI severity [24]. PTA describes
the period of dense impairment in the ability to learn new information (with or without
some degree of retrograde amnesia). Although PTA is most accurately understood as
one of the later stages within the larger condition of posttraumatic encephalopathy
(PTE;discussedlaterinthischapter)[25], the conventional assessment of PTA dura-
tion encompasses the entire period between injury and the recovery of reasonably
continuous and accurate memory for daily events. There are several measures with
which to formally assess PTA severity and duration, the most commonly used of which
are the Galveston Orientation and Amnesia Test (GOAT) [26] and the Orientation Log
(O-Log) [27, 28]; among these, we prefer the O-Log given its ease of administration
and interpretation and its excellent statistical comparison to the GOAT [29].
When neither GCS nor prospective PTA data are available, TBI severity may be
characterized retrospectively using the American Congress of Rehabilitation
Medicine(ACRM)definitionofmildTBI[30, 31]. These criteria define TBI as a
physiological disruption in brain function resulting from the application of an exter-
nalphysical(includingacceleration/deceleration)force,asevidencedbyanyone
(or more) of the following: LOC, PTA, altered mental state (“dazed and confused”),
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 129
and/orafocalneurologicaldeficitthatmayormaynotbetransient.Toremainin
the mild category, LOC must be less than 30 min, after which GCS scores are in
the13–15range,and/orPTAdurationisnolongerthan24h.Injuriesthatproduce
LOC of 30 or more min, GCS scores less than 13 at 30 min (or later) post injury,
and/orPTAlongerthan24hareclassifiedasmoderatetosevere.McMillanand
colleaguesin1996[31] demonstrated that retrospective interview-based estimates
PTA of duration among hospitalized individuals with TBI correlate highly with
prospective, GOAT-determined duration of PTA. Among patients whose PTA dura-
tion is longer than 24 h, practical classification of injuries as moderate or severe
may be made according to PTA duration (whether prospectively or retrospectively
determined)of1–7daysandmorethan7days,respectively.
For the consulting neuropsychiatrist, the usefulness of measured or estimated
PTA duration and PTA-based severity classifications lies in the prognostic utility of
this value: PTA duration (as a continuous variable) is a robust predictor of functional
independence [29, 34] and disability [34] at the end of acute inpatient rehabilitation,
Glasgow Outcome Scale [35] scores at 6 and 12 months post injury [36, 37], long-
term cognitive recovery [38–40], productivity [41], employment [42], and commu-
nity reintegration [43]. As with the GCS, noninjury factors (e.g., sedating medications,
severe communication impairments) must be considered when estimating PTA
duration, particularly among subjects with more severe general physical injuries
and complications. Nonetheless, in our clinical experience time between injury and
emergence from PTA – regardless of the TBI and concurrent factors contributing to
the duration of that period – define usefully the severity of injury and offer valuable
prognostic information that the consulting neuropsychiatrist can use when commu-
nicating with patients and their families as well as other healthcare providers about
the patient’s prognosis and likely posthospital treatment and resource needs.
AnimportantqualifieronACRM-basedTBIseverityclassificationofwhichthe
consulting neuropsychiatrist should be aware is “complicated mild TBI” [32, 33].
ThissubtypeofmildTBIisusedtodenoteindividualswhomeetACRMcriteria
for mild TBI but whose computed tomography (CT) or magnetic resonance imaging
(MRI) of the brain demonstrates abnormalities consistent with TBI. The impor-
tance of noting this subtype is that the outcome of subjects with complicated mild
TBI more closely resembles that of persons with moderate TBI than those with
uncomplicated (i.e., “simple”) mild TBI; awareness of this issue will allow the
consulting neuropsychiatrist to interpret the patient’s clinical presentation more
accurately and to offer more fully informed education, counseling, and prognostic
information to patients and their families.
Neurobiology of TBI
destructionofneuronsand/ortheiraxonaltermini.Excitatoryaminoacidexcesses
also overdrive glucose utilization, and oxidative metabolism, and produce poten-
tially toxic accumulations of lactate. Concurrent excess of acetylcholine also appear
to be excitotoxic and may amplify the destructive effects of excitatory amino acid
excesses and be particularly injurious to brain areas where these neurotransmitters
are densely colocated (i.e., hippocampus, frontal cortices; see Phillips and Reeves
[55], Arciniegas [56], and Arciniegas and Silver [57] for review). The effects, benign
or malignant, of acute cerebral monoaminergic (i.e., dopamine, norepinephrine, and
serotonin) excesses that are part of the cytotoxic cascade remain uncertain. All these
neurotransmitter excesses appear to wane over the first several weeks following TBI
[58, 59], although the exact time course over which these excesses abate is not char-
acterized fully. It is clear, however, that by several weeks post-TBI there is a relative
cholinergic deficit resulting from injury to ventral forebrain cholinergic nuclei and
their cortical projections [56, 57]. It is possible that TBI also results in primary or
secondary disturbances in cerebral monoaminergic, and particularly noradrenergic
and dopaminergic, systems [60], the effects of which may be modified by geneti-
cally mediated variations in catecholamine metabolism. These issues are particularly
important for the consulting neuropsychiatrist, as the types and timings of post-
traumatic neurotransmitter disturbances carry implications for pharmacotherapies
directed at cognitive, emotional, and behavioral problems during the acute rehabili-
tation period and thereafter.
In addition to biomechanical and cytotoxic injury processes, persons with TBI
who require hospitalization often experience other secondary neurological and
systemic problems, whether as a consequence of TBI or as a comorbid process, that
may complicate or exacerbate TBI. Such problems include traumatic intracerebral
hematomas, focal or diffuse cerebral edema, elevated intracranial pressure (ICP),
obstructive hydrocephalus, hypoxic-ischemic injury, intracranial/intracerebral
infection, and subfalcine or transtentorial herniation. These latter problems may be
fatal or, if not fatal, may compromise vascular supply in the areas of brain compres-
sion and thereby superimpose acute ischemic stroke on TBI. Additionally, systemic
medicalcomplicationssuchasvolumedepletion/bloodloss,hypoperfusion,hypo-
thermia or hyperthermia, hypoxia, infection, and related problems may further
complicate TBI. Aggressive treatment directed at these problems during acute care
and, when necessary, in the acute rehabilitation period, therefore is essential.
As noted in the prior section of this chapter, TBI disproportionately affects the
anterior and ventral aspects of the frontal and temporal lobes, medial frontal and
temporal areas, ventral forebrain, the diencephalon (thalamus, hypothalamus), the
rostral and ventral areas of the upper brainstem, and the white matter within and
between these areas [44, 53, 54, 61, 62]. Injury to these neuropsychiatrically salient
areas produces typical, but not invariate, patterns of posttraumatic neuropsychiatric
132 D.B. Arciniegas
disturbances in the acute and subacute postinjury periods (Table 2). These patterns
of neuropsychiatric disturbances evolve in the days to weeks following TBI and are
subsumedundertheheadingofPTE[25].
Posttraumatic Encephalopathy
Posttraumaticencephalopathy(PTE)ischaracterizedbyfivestages:posttraumatic
coma, posttraumatic delirium, PTA, posttraumatic dysexecutive syndrome, and
recovery (Fig. 1). These stages are named according to the most salient (although
clearly not the only) neurobehavioral feature of the clinical presentation. During
posttraumatic coma, the most salient feature of the patient’s presentation is complete
impairment of arousal. The transition from posttraumatic coma to posttraumatic
delirium is marked by the return of wakefulness (arousal), albeit sometimes in a
fluctuating manner, and marked impairments in selective, sustained, and other aspects
of attention; in the parlance of the American Psychiatric Association’s Diagnostic
andStatisticManual,FourthEdition,TextRevised[63], “reduced clarity of aware-
ness of the environment.” Although posttraumatic delirium also entails other distur-
bances in cognition, emotion, and behavior, profound inattention is the most salient
andcharacteristicfeatureofthisstageofPTE.
TBI
Brief
LOC, Posttraumatic Dysexecutive Syndrome to recovery...
PTA,
or Mild TBI
altered Posttraumatic Dysexecutive Syndrome to recovery...
mentation
Moderate TBI
Severe TBI
Posttraumatic Amnesia
Posttraumatic
Dysexecutive
Syndrome to
recovery...
Posttraumatic Delirium
Functional Cognition
Posttraumatic Amnesia
Posttraumatic
Coma
Posttraumatic Delirium
Posttraumatic Coma
Time
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation
Fig. 1 Typicalcourseofrecoverythroughthestagesofposttraumaticencephalopathy(PTE)followingmild,moderate,andseveretraumaticbraininjury.TBI
traumatic brain injury; LOC loss of consciousness; PTA posttraumatic amnesia
133
134 D.B. Arciniegas
As the patient emerges from this stage, basic selective and sustained attention
improve markedly; as a consequence, the patient’s striking and dense impairment in
declarative new learning becomes the most salient feature of the clinical presenta-
tion, and marks the patient’s transition into PTA. As with posttraumatic delirium,
the period of PTA also entails impairments in other aspects of cognition, especially
executive function and executive control of attention, language, memory, and
praxis. The patient is regarded as “in PTA” until his or her declarative new learning
improves to the point that he or she is able to construct a reasonably continuous
narrative of daily events from that point forward (formal criteria for emergence
from PTA using the GOAT or O-Log are described in the next section of this chap-
ter). The interval between onset of injury and subsequent recovery of declarative
new learning defines the duration of PTA.
Unfortunately, the measured period of PTA duration is sometimes misunder-
stood as suggesting that the entire period measured is first and foremost character-
ized by impaired declarative new learning (i.e., amnesia); this clearly is not the
case. It is important to be clear that although the formally measured duration of
PTA necessarily encompasses posttraumatic coma, posttraumatic delirium, and
(by some accounts) any preinjury period of retrograde amnesia, neither the con-
cept of PTA nor its clinical manifestations are synonymous with posttraumatic
delirium or, more obviously, posttraumatic coma. It is true that declarative new
learningisimpairedintheseearlierstagesofPTE;however,eachoftheseentails
other, and more salient, cognitive impairments. Nonetheless, duration of PTA as
defined and used in the TBI literature is useful for both TBI severity characteriza-
tion as well as a predictor of TBI outcomes and is, in this author’s view, the most
usefuloftheearlystagesofPTEtomeasureassiduouslywhenofferingprognoses
to patients, their families, and other rehabilitation clinicians.
Upon emergence from PTA, impairments of executive function are the most
salientfeatureoftheclinicalpresentation;accordingly,thisstageofPTEisreferred
to as the posttraumatic dysexecutive syndrome. Common clinical features of this
syndrome include impairments of intrinsic executive function (e.g., abstraction,
problem solving, the ability to generate, shift, and alter cognitive sets independent
of environmental contingencies, judgment, and insight) as well as impaired execu-
tive control of other, more basic, cognitive functions. As illustrated in Fig. 1, some
patientsemergefromthisstageofPTEintocompletecognitiverecoverywhereas
for others this becomes a chronic posttraumatic neurocognitive disorder.
TheimpairmentsthatcompriseeachstageofPTEoccuronacontinuumclini-
callyandtemporally:patientsatthetransitionbetweenstagesofPTEmayvacillate
fordays(orlonger)betweenthosestages.Nonetheless,identifyingthestageofPTE
that best describes that patient is useful in that it facilitates the development of a
treatment plan which is appropriate to the patient’s current clinical status and also
allows clinicians and the patient’s family members to anticipate the course of
continued recovery. By extension, this approach to PTE also helps clinicians to
identify deviations from the expected course of recovery after TBI and therefore the
need to evaluate the patient for conditions that explain such deviations.
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 135
Standardized assessments appropriate to the phase of PTE in which the patient
presents facilitate accurate diagnosis and guide prognostic and therapeutic formula-
tions. Data derived from these measures may be used to gauge not only the extent
and rate of recovery but also responses to treatment.
Although it is uncommon in the United States for patients to present to rehabili-
tation settings in posttraumatic coma, presentation of patients in this and other
states of impaired arousal and awareness (i.e., with disorders of consciousness) is
not uncommon in other parts of the world. The Coma/Near-Coma Scale [64] is
particularly useful as an assessment of coma severity and recovery.
Upon emergence from posttraumatic coma, patients enter the period of post-
traumatic delirium; at this point, it is very common for patients to be admitted to an
acute inpatient rehabilitation hospital. The Delirium Rating Scale-Revised-98
(DRS-R-98)[65] or the Confusion Assessment Protocol (CAP) [66] are appropriate
and useful measures for the consulting neuropsychiatrist to apply to the evaluation
and monitoring of patients in posttraumatic delirium. The period of posttraumatic
delirium corresponds to levels II–V of the Rancho Los Amigos Scale (RLAS) [67].
This scale and the descriptors of these lower levels of recovery after TBI are used
frequently by rehabilitation physicians and therapists; the consulting neuropsychiatrist
136 D.B. Arciniegas
will be well served to be familiar with this scale to ensure proper interdisciplinary
communication when discussing patients in posttraumatic delirium.
Consistent with the description of RLAS levels II–V, posttraumatic delirium
generally begins as a state of impaired arousal and profound inattention (RLAS
level II). As arousal improves, inattention and behavioral disturbances (agitation)
become prominent features of the clinical presentation (RLAS levels III–IV).
As agitation remits and attentional disturbances become less problematic (RLAS
level V), patients transition into a state in which impaired declarative new learning
is the most salient clinical problem, or PTA. Identifying the point of transition
between these states may be facilitated by the use of appropriate cutoff scores from
deliriumontheDRS-R-98ortheCAPandcontinuedimpairmentonmeasuresof
PTA such as the O-Log or the GOAT [26].
With this in mind, it is useful to begin the assessment of PTA using the O-Log
or the GOAT early during the course of posttraumatic delirium. Assessment using
the O-Log of the GOAT continues until the patient meets criteria for emergence
from PTA (on two consecutive days, O-Log scores ³25 or GOAT scores ³76).
Although the validity of assessing orientation and memory function in the severely
delirious patient is dubious, early and daily assessment with these measures ensures
that the end of PTA will be captured accurately; as noted earlier, and as demon-
strated by our own work [29], duration of PTA offers short- and long-term prognostic
information that is useful to clinicians and also to patients and their families.
Neuropsychiatric assessment is most often undertaken when patients are in
posttraumatic delirium or PTA. Our service, which provides Behavioral Neurology
and Neuropsychiatry consultations on an acute inpatient neurorehabilitation unit,
integrates neurological and neuropsychiatric assessments to offer diagnostic and
treatment recommendations as well as guidance on rehabilitation prognosis and the
types of support and caregiving resources that are likely to be needed during these
periodsofPTEaswellasduringandaftersubsequentrehabilitationcare.Inaddi-
tion to elementary neurological and general mental status examinations, our
consultations include detailed examination for subtle neurological signs (SNS)
and a thorough bedside cognitive examination.
The assessment for SNS focuses on paratonia (mitgehen and/or gegenhalten)
and also several primitive reflexes (also known as “frontal release signs”): gla-
bellar response, snout response, suck reflex, palmomental response (left and right),
grasp response (left and right), and rooting response. These simple additions to
the neurological examination, from which we have developed a preliminary metric
referred to as the SNS score [68], yield important information regarding neurobehav-
ioral status and rehabilitation outcome: SNS score predicts raw and Z-transformed
mini-mental state examination (MMSE) and frontal assessment battery (FAB)
scores (all P<0.003),FIMscoresatconsultation(allP < 0.04), and rehabilitation
discharge (all P < 0.03), and RLOS (P < 0.0002). Accordingly, inclusion of these
items in the neuropsychiatric assessment of persons with TBI during the acute
rehabilitation period is recommended.
The bedside cognitive examination used on our service includes, among other
items,theMMSE[69] and FAB [70]. Because performance on these measures is
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 137
Neurodiagnostic Methods
SymbolDigitModalitiesTest,groovedpegboard,phonemicandcategoricalword
generation tasks, Wechsler Test of Adult Reading, and Wisconsin Card Sorting
Test-64 may be a useful and practical brief neuropsychological assessment battery
in the inpatient neurorehabilitation setting. Additionally, this battery – and, more
specifically, its Weschler Test of Adult Reading and Trail Making Test-Part B
components – are significant predictors of 1-year outcome after TBI as measured
by the Disability Rating Scale, Supervision Rating Scale, and Glasgow Outcome
Scale-Extended [81]. When it is feasible to obtain neuropsychological testing of
this type in the acute neurorehabilitation setting, it is advisable to do so as a comple-
ment to other data obtained during the neuropsychiatric assessment.
Structural neuroimaging is an integral component of the neuropsychiatric assess-
ment of patients receiving acute neurorehabilitation after TBI. In many (perhaps
most) cases, CT of the brain will be performed in the acute care setting; unfortunately,
CT is sensitive to gross abnormalities (i.e., skull fracture, acute hemorrhage or hem-
orrhagic contusion, severe DAI) but its value is generally limited to cases in which
verysevereinjuriesweresustained.MRIofthebrainisfrequentlymoreusefulasa
neuroimaging guide to the severity of TBI; as a tool with which to determine the cor-
respondence between bedside examination-identified neurological/neuropsychiatric
problems and neuroimaging abnormalities; and as a guide to prognosis and treatment
planning. For example, ventral prefrontal cortical and white matter injury is a rela-
tively common consequence of severe TBI (see Table 2) and frequently is associated
with impulsive, disinhibited, and/or aggressive behavior. MR evidence of overtly
destructive damage (i.e., traumatic ablation) to these structures influences the selec-
tion of pharmacologic agents directed at these behaviors: selective serotonin reuptake
inhibitors, anticonvulsants, or atypical antipsychotics suppressing brain (limbic) areas
driving these behaviors is likely to be more useful than are agents (e.g., stimulants or
cholinesterase inhibitors) intended to augment the function of the ventral prefrontal-
subcortical circuit. Accordingly, we recommend obtaining (or reviewing one previ-
ously obtained) an MRI of the brain in all neurorehabilitation inpatients receiving
neuropsychiatricassessmentafterTBI.WhenMRIisperformed,T1, fluid-attenuated
inversion recovery (FLAIR), T2* gradient echo, susceptibility-weighted, and diffu-
sion-weighted sequences are the most useful sequences to obtain [82].
Electroencephalography (EEG), including evoked potentials, event-related
potentials,andquantitativeEEG(qEEG),doesnotusuallycontributeusefullytothe
neuropsychiatric assessment of patients undergoing acute neurorehabilitation after
TBI [83]. When clinical history suggests the possibility of seizures (particularly
complex partial seizures with post-ictal confusion or behavioral disturbances), then
itisappropriatetoobtainEEGtoidentifypotentiallyepileptiformabnormalities.
However,itisimportanttoremainmindfulthatinterictalEEGisrelativelyinsensi-
tive to epileptiform abnormalities and that the decision to treat patients for post-
traumatic seizures rests on the event semiology and not on the presence or absence
of electroencephalographic abnormalities.
The literature guiding the selection of laboratory assessments relevant to the neurop-
sychiatric assessment in the acute neurorehabilitation setting is underdeveloped.
Itisreasonableandappropriatetoreviewand/orobtainlaboratorydata(including
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 139
serum and urine studies) that may inform on contributors to, or alternate explana-
tions for, delirium and cognitive impairments experienced by persons with TBI.
Recent reviews suggest that neuroendocrine disturbances are common and under-
diagnosed in this population [84]; other than assessment of thyroid-stimulating
hormone and thyroid hormone levels, however, the optimal methods for assessing
and treating other posttraumatic neuroendocrine disturbances remains uncertain.
and aggression) [95] and as agents with which to improve compliance with mechanical
ventilation [96]. Agents that attenuate noradrenergic function, including clonidine,
propranolol, and other antihypertensives, are commonly used for medical and/or
behavioral purposes as well. However, dopaminergic and noradrenergic antagonists
delay neuronal recovery and impair neuronal plasticity [97–103]. Among persons
with TBI, typical antipsychotics exacerbate cognitive impairments [104] and prolong
the period of PTA [105]. Benzodiazepines are well known to impair memory and
other aspects of cognition [106] in healthy adults and among persons with TBI [107].
In light of these findings, use of agents with potent antidopaminergic, antinoradren-
ergic, and/or GABA-ergic properties is best avoided during the neurorehabilitative
care of persons with TBI. When evaluating neuropsychiatrically patients receiving
these agents, considering their potential effects on neurological and cognitive func-
tion before making definitive diagnostic or prognostic statements is prudent.
With respect to the effects of other agents on posttraumatic neuropsychiatric func-
tion, medications possessing potent in vivo anticholinergic properties are of concern as
well. These medications are commonly prescribed for posttraumatic dizziness and
urinary incontinence. Antidepressants with potent anticholinergic properties (e.g.,
tricyclic and tetracyclic antidepressants and also paroxetine [108]) are often prescribed
by rehabilitation physicians for pain, headaches, and emotional disturbances. However,
TBI produces acute and chronic disturbances of cerebral cholinergic function [56], and
cholinergic deficits become prominent and functionally significant in many patients
over the first several weeks after TBI. As a result of these deficits, patients with TBI
are vulnerable to the adverse cognitive and behavioral effects of agents with anti-
cholinergic properties. In general, prescription of agents with potent anticholinergic
properties for persons with TBI should be avoided whenever possible.
Finally, pain and spasticity are common problems among persons with TBI,
most often as a result of injury to the head or other orthopedic or soft tissue injuries
that are sustained concurrently with TBI. Among the agents used for these prob-
lems, opiate analgesics are particularly likely to adversely affect cognitive and
neuropsychiatric function. At typical analgesic doses, these agents produce impair-
ments in memory among persons without TBI of severities comparable to those
encountered among persons in PTA [109]. These agents may exacerbate, prolong,
or mimic posttraumatic coma, posttraumatic delirium, PTA, and the posttraumatic
dysexecutive syndrome. Using the minimum necessary dose of any of these agents
for as brief a time as is feasible clinically, or, better, avoiding or eliminating these
whenever possible, is recommended.
Conclusion
areas. Damage to these structures explains, in large part, the anatomic contributions
to the neurobehavioral sequelae of TBI, including alterations of arousal, attention,
processing speed, memory, functional communication, executive function, emotional
regulation, and behavior.
As discussed in this chapter, the period of neuropsychiatric disturbance that imme-
diatelyfollowsTBIisunderstoodusefullyasposttraumaticencephalopathy(PTE),a
condition with several stages through which patients proceed during recovery from
TBI. These stages include, in typical sequence, posttraumatic coma, posttraumatic
delirium, posttraumatic amnesia (PTA), posttraumatic dysexecutive syndrome, and full
recovery. Among these, duration of PTA is particularly useful to measure accurately:
duration of PTA is strongly predictive of short- and long-term neurorehabilitation
outcomes. Additionally, data developed on our neuropsychiatric consultation service
suggest that several elements of the neuropsychiatric assessment, including the number
of SNS (paratonia, primitive reflexes) and also a brief bedside neurobehavioral status
examination(normativelyinterpretedMMSEandFAB),yielddatathatallowneurop-
sychiatrists to assess functional status, functional prognosis, and rehabilitation lengths
of stay among persons with TBI receiving inpatient neurorehabilitative care.
Treatment approaches based on data yielded by the neuropsychiatric assessment
vary widely among institutions, both nationally and internationally. These treatment
issues are beyond the scope of this chapter but are well described elsewhere [57,
110–117]. Independent of specific treatment recommendations, the principles of
neuropsychiatric assessment of TBI in the neurorehabilitation setting described in this
chapter apply broadly. Well informed and equipped with this information, neuropsy-
chiatrists will contribute importantly and effectively to multidisciplinary teams work-
ing to improve the neuropsychiatric and functional outcomes of persons with TBI.
References
1. Thurman D, Guerrero J (1999) Trends in hospitalization associated with traumatic brain
injury.JAMA282:954–957
2. ThurmanDJ,AlversonC,DunnKAetal(1999)TraumaticbraininjuryintheUnitedStates:
a public health perspective. J Head Trauma Rehabil 14:602–615
3. SelassieAW,ZaloshnjaE,LangloisJAetal(2008)Incidenceoflong-termdisabilityfollow-
ing traumatic brain injury hospitalization, United States, 2003. J Head Trauma Rehabil
23:123–131
4. LevinHS(1995)Neurobehavioraloutcomeofclosedheadinjury:implicationsforclinical
trials. J Neurotrauma 12:601–610
5. severeheadinjury.ExperienceoftheTraumaticComaDataBank.JNeurosurg73:699–709
6. RapoportM,McCauleyS,LevinHetal(2002)Theroleofinjuryseverityinneurobehavioral
outcome 3 months after traumatic brain injury. Neuropsychiatry Neuropsychol Behav Neurol
15:123–132
7. HartT,WhyteJ,PolanskyMetal(2003)Concordanceofpatientandfamilyreportofneurobe-
havioralsymptomsat1yearaftertraumaticbraininjury.ArchPhysMedRehabil84:204–213
8. KaufmanHH,LevinHS,HighWMJretal(1985)Neurobehavioraloutcomeaftergunshot
woundstotheheadinadultciviliansandchildren.Neurosurgery(Baltim)16:754–758
9. DikmenS,MachamerJ,MillerBetal(2001)Functionalstatusexamination:anewinstru-
mentforassessingoutcomeintraumaticbraininjury.JNeurotrauma18:127–140
142 D.B. Arciniegas
10. DikmenSS,MachamerJE,PowellJMetal(2003)Outcome3to5yearsaftermoderateto
severetraumaticbraininjury.ArchPhysMedRehabil84:1449–1457
11. HallKM,BushnikT,Lakisic-KazazicBetal(2001)Assessingtraumaticbraininjuryout-
come measures for long-term follow-up of community-based individuals. Arch Phys Med
Rehabil82:367–374
12. Doctor JN, Castro J, Temkin NR et al (2005) Workers’ risk of unemployment after traumatic
braininjury:anormedcomparison.JIntNeuropsycholSoc11:747–752
13. GuilmetteTJ,PagliaMF(2004)Thepublic’smisconceptionabouttraumaticbraininjury:a
followupsurvey.ArchClinNeuropsychol19:183–189
14. SwiftTL,WilsonSL(2001)Misconceptionsaboutbraininjuryamongthegeneralpublic
andnon-experthealthprofessionals:anexploratorystudy.BrainInj15:149–165
15. MarrAL,CoronadoVG(2002)CentralNervousSystemInjurySurveillanceDataSubmission
Standards – 2002
16. KrausJF,RiceTM,Peek-AsaCetal(2003)Facialtraumaandtheriskofintracranialinjury
inmotorcycleriders.AnnEmergMed41:18–26
17. JohnstonJJ(2007)TheGalaskoreportimplemented:theroleofemergencymedicineinthe
managementofheadinjuries.EurJEmergMed14:130–133
18. PalchakMJ,HolmesJF,VanceCWetal(2003)Adecisionruleforidentifyingchildrenat
lowriskforbraininjuriesafterbluntheadtrauma.AnnEmergMed42:492–506
19. Borg J, Holm L, Cassidy JD et al (2004) Diagnostic procedures in mild traumatic brain
injury:resultsoftheWHOCollaboratingCentreTaskForceonMildTraumaticBrainInjury.
JRehabilMed(Suppl43):61–75
20. TeasdaleG,JennettB(1974)Assessmentofcomaandimpairedconsciousness.apractical
scale. Lancet 2:81–84
21. CliftonGL,HayesRL,LevinHSetal(1992)Outcomemeasuresforclinicaltrialsinvolving
traumatically brain-injured patients: report of a conference. Neurosurgery (Baltim)
31:975–978
22. CowenTD,MeythalerJM,DevivoMJetal(1995)Influenceofearlyvariablesintraumatic
brain injury on functional independence measure scores and rehabilitation length of stay and
charges.ArchPhysMedRehabil76:797–803
23. GabbeBJ,CameronPA,FinchCF(2003)ThestatusoftheGlasgowComaScale.Emerg
Med(Fremantle)15:353–360
24. Russell WR, Smith A (1961) Post-traumatic amnesia in closed head injury. Arch Neurol
5:4–17
25. Arciniegas DB, McAllister TW (2008) Neurobehavioral management of traumatic brain
injuryinthecriticalcaresetting.CritCareClin24:viii,737–viii,765
26. LevinHS,O’DonnellVM,GrossmanRG(1979)TheGalvestonOrientationandAmnesia
Test. A practical scale to assess cognition after head injury. J Nerv Ment Dis
167:675–684
27. JacksonWT,NovackTA,DowlerRN(1998)Effectiveserialmeasurementofcognitiveori-
entationinrehabilitation:theOrientationLog.ArchPhysMedRehabil79:718–720
28. Novack TA, Dowler RN, Bush BA et al (2000) Validity of the Orientation Log, relative to
theGalvestonOrientationandAmnesiaTest.JHeadTraumaRehabil15:957–961
29. Frey KL, Rojas DC, Anderson CA et al (2007) Comparison of the O-Log and GOAT as
measures of posttraumatic amnesia. Brain Inj 21:513–520
30. Kay T, Harrington DE, Adams RE et al (1993) Definition of mild traumatic brain injury:
ReportfromtheMildTraumaticBrainInjuryCommitteeoftheHeadInjuryInterdisciplinary
SpecialInterestGroupoftheAmericanCongressofRehabilitationMedicine.JHeadTrauma
Rehabil8:86–87
31. McMillan TM, Jongen EL, Greenwood RJ (1996) Assessment of post-traumatic amnesia
after severe closed head injury: retrospective or prospective? J Neurol Neurosurg Psychiatry
60:422–427
32. WilliamsDH,LevinHS,EisenbergHM(1990)Mildheadinjuryclassification.Neurosurgery
(Baltim)27:422–428
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 143
33. vanderNaaltJ,HewJM,vanZomerenAHetal(1999)Computedtomographyandmagnetic
resonance imaging in mild to moderate head injury: early and late imaging related to out-
come.AnnNeurol46:70–78
34. ZafonteRD,MannNR,MillisSRetal(1997)Posttraumaticamnesia:itsrelationtofunc-
tionaloutcome.ArchPhysMedRehabil78:1103–1106
35. Jennett B, Bond M (1975) Assessment of outcome after severe brain damage. Lancet
1:480–484
36. Katz DI, Alexander MP (1994) Traumatic brain injury. Predicting course of recovery and
outcomeforpatientsadmittedtorehabilitation.ArchNeurol51:661–670
37. EllenbergJH,LevinHS,SaydjariC(1996)Posttraumaticamnesiaasapredictorofoutcome
aftersevereclosedheadinjury.Prospectiveassessment.ArchNeurol53:782–791
38. BrooksDN,AughtonME,BondMRetal(1980)Cognitivesequelaeinrelationshiptoearly
indices of severity of brain damage after severe blunt head injury. J Neurol Neurosurg
Psychiatry43:529–534
39. GeffenGM,EncelJS,ForresterGM(1991)Stagesofrecoveryduringpost-traumaticamne-
sia and subsequent everyday memory deficits. NeuroReport 2:105–108
40. HaslamC,BatchelorJ,FearnsideMRetal(1994)Post-comadisturbanceandpost-traumatic
amnesia as nonlinear predictors of cognitive outcome following severe closed head injury:
findingsfromtheWestmeadHeadInjuryProject.BrainInj8:519–528
41. ShererM,SanderAM,NickTGetal(2002)Earlycognitivestatusandproductivityoutcome
aftertraumaticbraininjury:findingsfromtheTBImodelsystems.ArchPhysMedRehabil
83:183–192
42. Avesani R, Salvi L, Rigoli G et al (2005) Reintegration after severe brain injury: a retrospec-
tivestudy.BrainInj19:933–939
43. Doig E, Fleming J, Tooth L (2001) Patterns of community integration 2-5 years post-dis-
chargefrombraininjuryrehabilitation.BrainInj15:747–762
44.BiglerED(2007)Anteriorandmiddlecranialfossaintraumaticbraininjury:relevantneu-
roanatomy and neuropathology in the study of neuropsychological outcome. Neuropsychology
21:515–531
45. SabetAA,ChristoforouE,ZatlinBetal(2008)Deformationofthehumanbraininducedby
mildangularheadacceleration.JBiomech41:307–315
46. BesenskiN(2002)Traumaticinjuries:imagingofheadinjuries.EurRadiol12:1237–1252
47. Ryan GA, McLean AJ, Vilenius AT et al (1994) Brain injury patterns in fatally injured
pedestrians.JTrauma36:469–476
48. MisraJC,ChakravartyS(1984)Astudyofrotationalbraininjury.JBiomech17:459–466
49. FijalkowskiRJ,StemperBD,PintarFAetal(2007)Newratmodelfordiffusebraininjury
usingcoronalplaneangularacceleration.JNeurotrauma24:1387–1398
50. MarguliesSS,ThibaultLE,GennarelliTA(1990)Physicalmodelsimulationsofbraininjury
in the primate. J Biomech 23:823–836
51. Meythaler JM, Peduzzi JD, Eleftheriou E et al (2001) Current concepts: diffuse axonal
injury-associatedtraumaticbraininjury.ArchPhysMedRehabil82:1461–1471
52. Black KL, Hanks RA, Wood DL et al (2002) Blunt versus penetrating violent traumatic brain
injury: frequency and factors associated with secondary conditions and complications. J
HeadTraumaRehabil17:489–496
53. Povlishock JT, Katz DI (2005) Update of neuropathology and neurological recovery after
traumaticbraininjury.JHeadTraumaRehabil20:76–94
54. BiglerED(2003)Neurobiologyandneuropathologyunderlietheneuropsychologicaldeficits
associatedwithtraumaticbraininjury.ArchClinNeuropsychol18:595–621
55. PhillipsLL,ReevesTM(2001)Interactivepathologyfollowingtraumaticbraininjurymodi-
fieshippocampalplasticity.RestorNeurolNeurosci19:213–235
56. Arciniegas DB (2003) The cholinergic hypothesis of cognitive impairment caused by trau-
maticbraininjury.CurrPsychiatryRep5:391–399
57. ArciniegasDB,SilverJM(2006)Pharmacotherapyofposttraumaticcognitiveimpairments.
BehavNeurol17:25–42
144 D.B. Arciniegas
58. MarkianosM,SeretisA,KotsouSetal(1992)CSFneurotransmittermetabolitesandshort-
termoutcomeofpatientsincomaafterheadinjury.ActaNeurolScand86:190–193
59. MarkianosM,SeretisA,KotsouAetal(1996)CSFneurotransmittermetabolitesincomatose
headinjurypatientsduringchangesintheirclinicalstate.ActaNeurochir(Wien)138:57–59
60. McAllister TW, Flashman LA, Sparling MB et al (2004) Working memory deficits after
traumatic brain injury: catecholaminergic mechanisms and prospects for treatment – a
review. Brain Inj 18:331–350
61. PovlishockJT(1992)Traumaticallyinducedaxonalinjury:pathogenesisandpathobiologi-
cal implications. Brain Pathol 2:1–12
62. Salmond CH, Chatfield DA, Menon DK et al (2005) Cognitive sequelae of head injury:
involvementofbasalforebrainandassociatedstructures.Brain128:189–200
63. American Psychiatric Association (2000) Diagnostic and Statistical Manual of Mental
Disorders, Text Revision, 4th edn. American Psychiatric Association, Washington, DC
64. RappaportM(2005)TheDisabilityRatingandComa/Near-Comascalesinevaluatingsevere
head injury. Neuropsychol Rehabil 15:442–453
65. Trzepacz PT, Mittal D, Torres R et al (2001) Validation of the Delirium Rating Scale-
Revised-98:comparisonwiththedeliriumratingscaleandthecognitivetestfordelirium.
JNeuropsychiatryClinNeurosci13:229–242
66. Sherer M, Nakase-Thompson R, Yablon SA et al (2005) Multidimensional assessment of
acuteconfusionaftertraumaticbraininjury.ArchPhysMedRehabil86:896–904
67. HagenC,MalkmusD,DurhamP(1972)RanchoLosAmigosScale.http://www.rancho.org/
patient_education/bi_cognition.pdf
68. WortzelHS,FreyKL,AndersonCA,ArciniegasDB(2009).Subtleneurologicalsignspre-
dict the severity of subacute cognitive and functional impairments after traumatic brain
injury. J Neuropsychiatry Clin Neurosci 21:463–466
69. Folstein MF, Folstein SE, McHugh PR (1975) Mini-mental state: a practical method for
gradingthecognitivestateofpatientsfortheclinician.JPsychiatryRes12:189–198
70. Dubois B, Slachevsky A, Litvan I et al (2000) The FAB: a frontal assessment battery at
bedside. Neurology 55:1621–1626
71. CrumRM,AnthonyJC,BassettSSetal(1993)Population-basednormsforthemini-mental
stateexaminationbyageandeducationlevel.JAMA269:2386–2391
72. AppollonioI,LeoneM,IsellaVetal(2005)Thefrontalassessmentbattery(FAB):norma-
tive values in an Italian population sample. Neurol Sci 26:108–116
73. ArciniegasDB,FreyK,AlderferBetal(2006)Impairmentsoffrontally-mediatedcognition
characterize posttraumatic encephalopathy following resolution of posttraumatic amnesia.
J Neuropsychiatry Clin Neurosci 18:281
74. VanierM,MazauxJM,LambertJetal(2000)Assessmentofneuropsychologicimpairments
after head injury: interrater reliability and factorial and criterion validity of the Neurobehavioral
RatingScale–Revised.ArchPhysMedRehabil81:796–806
75. McCauleySR,LevinHS,VanierMetal(2001)Theneurobehaviouralratingscale-revised:sensi-
tivityandvalidityinclosedheadinjuryassessment.JNeurolNeurosurgPsychiatry71:643–651
76. RoyallDR,MahurinRK,GrayKF(1992)Bedsideassessmentofexecutivecognitiveimpair-
ment: the executive interview. J Am Geriatr Soc 40:1221–1226
77. Grigsby J, Kaye K, Robbins LJ (1992) Reliabilities, norms and factor structure of the
BehavioralDyscontrolScale.PerceptMotSkills74:883–892
78. KayeK,GrigsbyJ,RobbinsLJetal(1990)Predictionofindependentfunctioningandbehavior
problems in geriatric patients. J Am Geriatr Soc 38:1304–1310
79. CummingsJL,MegaM,GrayKetal(1994)TheNeuropsychiatricInventory:comprehen-
sive assessment of psychopathology in dementia. Neurology 44:2308–2314
80. Kalmar K, Novack TA, Nakase-Richardson R et al (2008) Feasibility of a brief neuropsycho-
logic test battery during acute inpatient rehabilitation after traumatic brain injury. Arch Phys
MedRehabil89:942–949
81. Hanks RA, Millis SR, Ricker JH et al (2008) The predictive validity of a brief inpatient
neuropsychologicbatteryforpersonswithtraumaticbraininjury.ArchPhysMedRehabil
89:950–957
Neuropsychiatric Assessment of Traumatic Brain Injury During Acute Neurorehabilitation 145
82. AkiyamaY,MiyataK,HaradaKetal(2009)Susceptibility-weightedmagneticresonance
imaging for the detection of cerebral microhemorrhage in patients with traumatic brain
injury.NeurolMedChir(Tokyo)49:97–99
83. ArciniegasDB,AndersonCA,RojasDC(2005)Electrophysiologicaltechniques.In:Silver
JM, McAllister TW, Yudofsky SC (eds) Textbook of traumatic brain injury. American
Psychiatric Publishing, Washington, DC
84. RothmanMS,ArciniegasDB,FilleyCMetal(2007)Theneuroendocrineeffectsoftrau-
maticbraininjury.JNeuropsychiatryClinNeurosci19:363–372
85. FreyLC(2003)Epidemiologyofposttraumaticepilepsy:acriticalreview.Epilepsia44(suppl
10):11–17
86. Schierhout G, Roberts I (2000) Anti-epileptic drugs for preventing seizures following acute
traumaticbraininjury.CochraneDatabaseSystRev4:CD000173
87. DikmenSS,TemkinNR,MillerBetal(1991)Neurobehavioraleffectsofphenytoinprophy-
laxisofposttraumaticseizures.JAMA265:1271–1277
88. SmithKRJr,GouldingPM,WildermanDetal(1994)Neurobehavioraleffectsofphenytoin
and carbamazepine in patients recovering from brain trauma: a comparative study. Arch
Neurol 51:653–660
89. Szaflarski JP, Meckler JM, Szaflarski M et al (2007) Levetiracetam use in critically ill
patients.NeurocritCare7:140–147
90. RueggS,NaegelinY,HardmeierMetal(2008)Intravenouslevetiracetam:treatmentexperi-
encewiththefirst50criticallyillpatients.EpilepsyBehav12:477–480
91. MulaM,TrimbleMR,YuenAetal(2003)Psychiatricadverseeventsduringlevetiracetam
therapy.Neurology61:704–706
92. MulaM,TrimbleMR,SanderJW(2004)Psychiatricadverseeventsinpatientswithepilepsy
andlearningdisabilitiestakinglevetiracetam.Seizure13:55–57
93. DikmenSS,MachamerJE,WinnHRetal(2000)Neuropsychologicaleffectsofvalproatein
traumaticbraininjury:arandomizedtrial.Neurology54:895–902
94. Marion DW (2006) Evidenced-based guidelines for traumatic brain injuries. Prog Neurol
Surg19:171–196
95. Fugate LP, Spacek LA, Kresty LA et al (1997) Measurement and treatment of agitation
following traumatic brain injury: II. A survey of the Brain Injury Special Interest Group of
theAmericanAcademyofPhysicalMedicineandRehabilitation.ArchPhysMedRehabil
78:924–928
96.MilbrandtEB,KerstenA,KongLetal(2005)Haloperidoluseisassociatedwith
lower hospital mortality in mechanically ventilated patients. Crit Care Med
33:226–229
97. GoldsteinLB(1993)Basicandclinicalstudiesofpharmacologiceffectsonrecoveryfrom
braininjury.JNeuralTransplantPlast4:175–192
98. Goldstein LB (1999) Pharmacological approach to functional reorganization: the role of
norepinephrine.RevNeurol(Paris)155:731–736
99. Goldstein LB (2003) Neuropharmacology of TBI-induced plasticity. Brain Inj
17:685–694
100. Goldstein LB (2006) Neurotransmitters and motor activity: effects on functional recovery
afterbraininjury.NeuroRx3:451–457
101. Hoffman AN, Cheng JP, Zafonte RD et al (2008) Administration of haloperidol and risperi-
done after neurobehavioral testing hinders the recovery of traumatic brain injury-induced
deficits.LifeSci83:602–607
102. Kline AE, Hoffman AN, Cheng JP et al (2008) Chronic administration of antipsychotics
impede behavioral recovery after experimental traumatic brain injury. Neurosci Lett
448:263–267
103. KlineAE,MassucciJL,ZafonteRDetal(2007)Differentialeffectsofsingleversusmultiple
administrations of haloperidol and risperidone on functional outcome after experimental
braintrauma.CritCareMed35:919–924
104. Stanislav SW (1997) Cognitive effects of antipsychotic agents in persons with traumatic
brain injury. Brain Inj 11:335–341
146 D.B. Arciniegas
105. Rao N, Jellinek HM, Woolston DC (1985) Agitation in closed head injury: haloperidol
effectsonrehabilitationoutcome.ArchPhysMedRehabil66:30–34
106. Buffett-JerrottSE,StewartSH(2002)Cognitiveandsedativeeffectsofbenzodiazepineuse.
Curr Pharm Des 8:45–58
107. BleibergJ,GarmoeW,CederquistJetal(1993)Effectsofdexedrineonperformancecon-
sistency following brain injury: a double-blind placebo crossover case study. Neuropsychiatry
Neuropsychol Behav Neurol 6:245–248
108. SchmittJA,KruizingaMJ,RiedelWJ(2001)Non-serotonergicpharmacologicalprofilesand
associated cognitive effects of serotonin reuptake inhibitors. J Psychopharmacol
15:173–179
109. McCarterRJ,WaltonNH,MooreCetal(2007)PTAtesting,theWestmeadposttraumatic
amnesiascaleandopiateanalgesia:acautionarynote.BrainInj21:1393–1397
110. WardenDL,GordonB,McAllisterTWetal(2006)Guidelinesforthepharmacologictreat-
ment of neurobehavioral sequelae of traumatic brain injury. J Neurotrauma 23:1468–1501
111. CiceroneKD,DahlbergC,KalmarKetal(2000)Evidence-basedcognitiverehabilitation:
recommendationsforclinicalpractice.ArchPhysMedRehabil81:1596–1615
112. CiceroneKD,DahlbergC,MalecJFetal(2005)Evidence-basedcognitiverehabilitation:
updated review of the literature from 1998 through 2002. Arch Phys Med Rehabil
86:1681–1692
113. CappaSF,BenkeT,ClarkeSetal(2005)EFNSguidelinesoncognitiverehabilitation:report
ofanEFNStaskforce.EurJNeurol12:665–680
114. CappaSF,BenkeT,ClarkeSetal(2003)EFNSguidelinesoncognitiverehabilitation:report
ofanEFNStaskforce.EurJNeurol10:11–23
115. Tate RL, Moseley A, Perdices M et al (2006) Update on Cicerone’s systematic review of
cognitiverehabilitation:thePsycBITEperspective.ArchPhysMedRehabil87:446
116. CiceroneKD,MottT,AzulayJetal(2008)Arandomizedcontrolledtrialofholisticneurop-
sychologic rehabilitation after traumatic brain injury. Arch Phys Med Rehabil
89:2239–2249
117. Cicerone KD, Mott T, Azulay J et al (2004) Community integration and satisfaction with
functioningafterintensivecognitiverehabilitationfortraumaticbraininjury.ArchPhysMed
Rehabil85:943–950
Neuropsychiatric Aspects of Vascular
Cognitive Impairment
Ingmar Skoog
Keywords Alzheimer’sdisease•Cerebrovasculardisease•Dementia•Depression
•Whitematterlesions
I. Skoog (*)
Neuropsychiatric Epidemiology Unit, Department of Psychiatry, Institute of Neuroscience
and Physiology, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
e-mail: [email protected]
Background
Cognitive function declines with increasing age. This decline differs between
individuals, probably as a consequence of genetic predisposition and educational
and professional background. The decline may be accelerated by different insults
to the brain, for example, cardiovascular and cerebrovascular diseases, other brain
disorders, preclinical dementia, and terminal decline. The cognitive dysfunction in
individuals with cardiovascular and cerebrovascular disease is often subsumed
under the term vascular cognitive impairment (VCI) [1]. Neuropsychiatric condi-
tions, such as Alzheimer’s disease (AD), depression, and psychotic disorders, are
also related to cognitive dysfunction, and in addition they have all been associated
with cardiovascular and cerebrovascular diseases. AD and cerebrovascular disease
often coexist in the same patient, being then labeled mixed dementia [2]. Vascular
depression is the term for coexistent depression and cerebrovascular disease [3].
Alzheimer’s Disease
The typical clinical picture of AD is dementia with insidious onset and a slowly
progressive loss of memory and other cognitive functions, such as language, visuo-
spatial abilities, executive function, orientation, praxis, and behavior. NINCDS-
ADRDA criteria [4] state that the diagnosis of probable AD requires the absence of
systemic disorders or other brain diseases that alone could account for dementia.
Clinical AD has a long preclinical phase [5, 6]. Thus, some cognitive dysfunctions
in elderly nondemented persons are caused by preclinical AD.
AD in the brain is characterized by marked neuronal and synaptic degeneration
and the presence of extensive amounts of senile plaques, neurofibrillary tangles,
and deposition of the beta-amyloid peptide in specific regions of the brain [7].
However, a large proportion of individuals who were cognitively normal before
death have numerous histopathological hallmarks of AD in the brain at autopsy
[8–11]. Several population-based neuropathological studies suggest that only
approximately 50% of individuals who fulfill neuropathological criteria for AD had
dementia or significant cognitive decline during life [8, 9, 12]. These neuropatho-
logical findings have recently been confirmed by positron emission tomography
studies using the Pittsburgh compound B to detect amyloid deposition in living
individuals [13]. In this study, 21% of normal elderly showed beta-amyloid deposi-
tion compatible with AD. Based on known figures on age-related prevalence of
dementia, this finding indicates that less than 50% of individuals with AD in the
brain show cognitive impairment during life. This finding implies that AD often
does not present with the clinical picture of dementia.
Neuropsychiatric Aspects of Vascular Cognitive Impairment 151
Depression
Psychotic Symptoms
Stroke
Stroke patients typically have history of stroke or transit ischemic attacks (TIA),
including acute focal neurological symptoms and signs, such as hemiparesis or
acute aphasia [39]. The symptoms must be present for 24 h or more for a diagnosis
of stroke. If they are present for a shorter duration, a diagnosis of TIA is given. The
cerebral infarcts are most often caused by thromboembolism from extracranial
arteries and the heart and are often related to large vessel disease. Other cardiovas-
cular manifestations, including myocardial infarction and hypertension, are common
in the patients. The most important risk factors for stroke are hypertension, diabetes
mellitus, atherosclerosis, atrial fibrillation, smoking, overweight, and hypercholes-
terolemia, especially high levels of low-density lipoprotein cholesterol [45]. All these
risk factors are potentially preventable or treatable.
Most epidemiological studies report an increased frequency of dementia in indi-
viduals with stroke. In the studies by Tatemichi et al. [46–48], relatively young
individuals with ischemic stroke had at least nine times greater risk for dementia
Neuropsychiatric Aspects of Vascular Cognitive Impairment 153
than stroke-free controls. Those persons with dementia after stroke had a higher
mortality rate and worse prognosis than those without dementia, which was inde-
pendent of stroke severity. Also, Pohjasvaara et al. [49] reported an increased
prevalence of dementia in stroke victims, as well as a decrease in independent living
for those with dementia. Lindén et al. [44] reported that stroke victims aged above
70 had an odds ratio of 4.7 for dementia. The odds for dementia was higher in those
aged 70 to 80 [odds ratio (OR), 6.7] than in those aged over 80 years, but the
frequency of dementia after stroke was higher after age 80 (34% vs. 18%). In addition,
60% of nondemented stroke victims had some cognitive dysfunction, showing that
very few elderly stroke victims are free from cognitive disturbances. In a population
study on 85-year-olds, Liebetrau et al. [50] reported that the odds ratio for dementia
in stroke was 4.3 and the prevalence of dementia after stroke was 57%. It may be that
the increased risk for dementia with stroke decreases with age at the same time as the
prevalence increases, both among those with and those without a history of stroke.
The pathogenesis of stroke-related dementia is not settled. The main hypothesis
is that dementia is related to the location or the volume of the infarcts, but there are
also other possibilities. The risk factors for dementia in individuals with stroke can be
divided into stroke-related and non-stroke-related factors [51]. The stroke-related
factors are similar to those in stroke, such as male sex, hypertension, diabetes
mellitus, smoking, and cardiac diseases. Non-stroke-related risk factors are similar
to those found in sporadic AD and include higher age, lower level of formal education,
family history of dementia, and the presence of cerebral atrophy. This combination of
risk factors supports the view that stroke-related dementia often is a consequence
of both stroke and preexisting AD pathology. According to neuropathological studies,
pure VAD, without any AD brain changes, is rare [52].
Stroke is an essential part of most criteria for VaD or VCI. The typical clinical
course of stroke-related dementia is sudden onset, stepwise deterioration, and a fluctu-
ating course. In the early stages, the cognitive symptoms may vary between individuals
depending on the site of the lesions. A large proportion of patients with cerebrovas-
cular disease have a gradual onset of dementia with a slowly progressive course [53],
with or without focal signs or infarcts on brain imaging, which makes it difficult to
differentiate from AD or other types of dementia. Changing risk factor patterns
(e.g., decrease in the frequency of smoking, better treatment of hypertension) have
decreased the incidence of stroke and may thus decrease the frequency of VaD. On the
other hand, more individuals survive after stroke, which may act in the opposite direc-
tion. It is not yet known if the prevalence or incidence of VaD increases or decreases.
The associations may also go in the opposite direction, as individuals with cognitive
impairment may be at increased risk for stroke and cerebral infarction. Three studies
[54–56] reported that elderly individuals with decreased cognitive ability and without
previous stroke at baseline were at increased risk for later incidents of stroke. In addi-
tion, one of these studies reported that also mild dementia at baseline was associated
with an increased incidence of new strokes [56]. One reason may be that these indi-
viduals already had silent cerebrovascular disease, such as silent infarcts or WMLs.
Depression is common after stroke [57], both in the acute [58], subacute [59–61],
and long-term perspective [62–64]. The frequency estimates varies from 25% to
154 I. Skoog
48% in the acute stage, from 14% to 50% at 1–12 months after stroke, and from
12% to 42% after a year or more following stroke [62]. In the study by Linden et al.
[62], stroke was related to an odds ratio (OR) for depression of 3.2 in women and
4.0 in men 1.5 years after the index stroke. A systematic review from 51 observa-
tional studies [65, 66] found a pooled estimate of 33% for depression after stroke.
Depression is important to detect in stroke patients, as it has been associated with
worse outcome in numerous studies [67–69]. Depressed stroke patients have more
cognitive impairments [61], spend more days in hospital [70], utilize more care, and
are more often institutionalized than the nondepressed [71]. The etiology of depres-
sion after stroke is debated. Some authors suggest biological factors, such as
disruption of frontostriatal [72] or left-sided prefrontosubcortical pathways [73], or
infarcts in strategic locations, such as right hemisphere strokes [74]. However, a
meta-analysis found no consistent location of infarcts to be associated with depres-
sion after stroke [75]. Others suggest that depression could be a reaction to disability
and loss of autonomy after stroke [76]. However, frequency of depression is increased
after stroke, also when controlling for stroke severity and disability [62].
Also, this association may go in the opposite direction, as several studies have
shown that depression increases the risk for first incidence stroke [77–79]. Reasons
include that depression has been associated with risk factors for stroke, such as
myocardial arrhythmia [80], increased platelet activation [81] and increased insulin
resistance [82]. However, in the study by Liebetrau et al. [78], depression was
related to lower diastolic blood pressure, and not to any other vascular factors.
Another explanation may be that late-life depression has been suggested to be
related to silent cerebrovascular diseases, such as ischemic WMLs, which may
increase the incidence of stroke.
Silent Infarcts
Cerebral infarcts often occur without focal symptoms. The frequency of these silent
infarcts increases with age [83]. These lesions were until recently believed to be
benign incidental findings on brain imaging. It has now been shown that individuals
with silent infarcts are at increased risk for clinical stroke [83] and dementia [84].
Among 85-year-olds, 10% had silent infarcts on CT, and these lesions doubled the
prevalence of dementia [85]. These lesions are thus important to detect, as treatment
of vascular risk factors, such as hypertension, may decrease the risk of new strokes
and thus potentially delay the onset of dementia in these high-risk patients.
White matter lesions (WMLs) are common in the elderly [86]. Subcortical areas of
both hemispheres are affected by marked or diffuse ischemic demyelination and
moderate loss of axons with astrogliosis and incomplete infarction. In addition,
Neuropsychiatric Aspects of Vascular Cognitive Impairment 155
arteriosclerotic changes with thickening of the vessel walls and narrowing of the
lumina of the small penetrating arteries and arterioles caused by hyalinization or
fibrosis are also present [87, 88].
The cause of WMLs is probably that long-standing hypertension causes lipohy-
alinosis and thickening of the vessel walls with narrowing of the lumen of the small
perforating arteries and arterioles that nourish the deep white matter [87]. It has
been suggested that the arterial changes are caused by the exposure of vessel walls
to increased pressure over time. The greater the pressure and/or lifespan, the more
likely are these changes to be present; this may be one reason for the observed
increase with age reported in most studies. Episodes of hypotension may lead to
further hypoperfusion and hypoxia-ischemia, leading to more loss of myelin in
subcortical areas. The deep white matter has few collaterals, which makes it more
vulnerable to ischemia than the cortex. Furthermore, myelin is probably more vulner-
able than axons to ischemia [89]. The cortex, the subcortical U-fibers, and corpus
callosum are thus generally well preserved [89].
The main risk factors for WMLs are high age, hypertension, and hypertension
clustering factors [90]. It was recently shown that increased diastolic blood pressure
and mean arterial pressure in midlife were related to the presence of WMLs in late life,
supporting both the theory that long-standing high blood pressure is significant and
that resistance of the smaller arteries and microvascular network is important [91].
In living individuals, WMLs can be detected on brain imaging. On computerized
tomography (CT) scans they appear as low-density areas and on magnetic reso-
nance imaging (MRI) as hyperdense areas. MRI and CT may not always capture
the same WMLs. WMLs on MRI often show no correlation with cognitive decline
and dementia and correspond to several different histological findings, for example,
état crible. WMLs on CT are most often related to cognitive impairment and
dementia and generally correspond to the histopathological picture described above
[92]. Furthermore, it has been shown that CT is better than MRI in predicting
symptomatic cerebrovascular disease in individuals with AD [93]. MRI is thus
more sensitive than CT to detect changes in the white matter but has a lower speci-
ficity in relation to neuropathological and clinical manifestations.
On CT, WMLs were found in 35% of nondemented 85-year-olds in a study
conducted in 1986–1987 [92], but a study conducted in 2000 found that 55% of
individuals aged 70–84 years had WMLs [94], illustrating that CT is becoming more
sensitive. MRI studies generally report much higher rates of WMLs than CT studies.
The Rotterdam population study [41] reported that 11% in the age strata 65–69 years,
21% in those aged 70–74 years, 27% in those aged 70–79 years, and 54% in those
aged 80–84 years had WMLs on MRI. A recent population-based neuropathological
study reported that 94% of demented subjects had WMLs [86], and another neuro-
pathological study found that 60% of individuals with AD had WMLs [88].
WMLs have been related to dementia both according to neuropathology [86, 88]
and in population-based CT studies [92], and among the nondemented it has been
related to visuo-spatial difficulties and psychomotor slowness [41, 43]. It has also
been found to be more common in individuals with AD than in individuals without
dementia [88, 92]. Investigations using CT of the brain in a population study of
156 I. Skoog
85-year-olds revealed that one-third of the nondemented and about two-thirds of the
AD patients had WMLs [92]. A similar proportion of AD patients had neuropatho-
logical evidence of ischemic WMLs when examined post mortem [88]. Based on
an autopsy study of nondemented individuals who had extensive AD neuropathol-
ogy, it was suggested that white matter degeneration precedes cortical atrophy in
AD [95]. A longitudinal population study also reported that WMLs predicted later
development of dementia [96]. Thus, WMLs seems to be strongly related to cogni-
tive decline and dementia in the elderly. It has even been suggested that individuals
with subcortical cerebrovascular disease show more decline in cognitive function
than those with cortical strokes [97].
The dementia related to WMLs often has an insidious onset and a slowly progres-
sive course [88, 97], which makes it difficult to distinguish from AD. WMLs seldom
occur as the sole cause of dementia, and the clinical picture may vary depending on
what other causes contribute to the dementia. Punctate WMLs on MRI do not prog-
ress, whereas confluent white matter abnormalities are progressive, and are thus
more malignant in the long term [98].
Ischemic WMLs are also related to late-life depression [99]. Two prospective
population-based studies using MRI report that white matter changes increase the
risk of subsequent occurrence of depression [100, 101]. In the latter study, it was
suggested that part of the relationship between white matter changes and depression
was mediated by loss of functional ability [101]. Also, cross-sectional studies
report that changes in the white matter detected with MRI are related to depression
[102]. The association between WMLs on CT and depression is less clear. Cross-
sectional studies from our group showed that WMLs on CT were associated with
dementia but not with depression [92]. WMLs in the frontal region have been asso-
ciated with depression in elderly patients [102], suggesting that WMLs disrupt the
frontostriatal circuitry and thereby lead to the development of depression. All these
findings are in line with the vascular depression hypothesis [3].
WMLs in depressed individuals may also influence outcome of depression.
It has been reported that severe WMLs on MRI predicted a shorter time to relapse
in elderly depressed individuals [103]. Furthermore, depressed individuals with
WMLs may also have a poorer response to antidepressant monotherapy than those
without WMLs [104].
Depression may also influence the development of WMLs. One recent study
reported that depression at baseline was associated with faster progression of
WMLs during follow-up [105].
Mixed Pathology
Conclusion
References
1. Bowler JV, Hachinski V (1995) Vascular cognitive impairment: a new approach to vascular
dementia. Baillieres Clin Neurol 4:357–376
2. Langa KM, Foster NL, Larson EB (2004) Mixed dementia. Emerging concepts and therapeu-
tic implications. JAMA 292:2901–2908
158 I. Skoog
3. Alexopoulos GS, Meyers BS, Young RC et al (1997) ‘Vascular depression’ hypothesis. Arch
Gen Psychiatry 54:915–922
4. McKhann G, Drachman D, Folstein M et al (1984) Clinical diagnosis of Alzheimer’s dis-
ease: Report of the NINCDS-ADRDA work group under the auspices of department of
health and human services task force on Alzheimer’s disease. Neurology 34:939–944
5. Sacuiu S, Sjögren M, Gustafson D et al (2005) Prodromal cognitive signs of dementia in
85-year-olds using four sources of information. Neurology 65:1894–1900
6. Sacuiu S, Gustafson D, Johansson B et al (2009) The pattern of cognitive symptoms predicts
time to dementia onset. Alzheimers Dement 5:199–206
7. Tomlinson B, Henderson G (1976) Some quantitative cerebral findings in normal and
demented old people. In: Terry RD (ed) Neurobiology of aging. Raven, New York
8. White L, Petrovich H, Hardman J et al (2002) Cerebrovascular pathology and dementia in
autopsied Honolulu-Asia Aging Study participants. Ann NY Acad Sci 977:9–23
9. Group N (2001) Medical Research Council Cognitive Function and Aging Study.
Pathological correlates of late-onset dementia in a multicentre, community-based population
in England and Wales. Lancet 357:169–175
10. Davies L, Wolska B, Hilbich C et al (1988) A4 amyloid protein deposition and the diagnosis
of Alzheimer’s disease: prevalence in aged brains determined by immunocytochemistry
compared with conventional neuropathologic techniques. Neurology 38:1688–1693
11. Delaère P, He Y, Fayet G et al (1993) bA4 deposits are constant in the brain of the oldest old:
an immunocytochemical study of 20 French centenarians. Neurobiol Aging 14:191–194
12. Snowdon D, Greiner L, Mortimer J et al (1997) Brain infarction and the clinical expression
of Alzheimer disease. The Nun Study. JAMA 277:813–817
13. Aizenstein HJ, Nebes RD, Saxton JA et al (2008) Frequent amyloid deposition without
significant cognitive impairment among the elderly. Arch Neurol 65:1509–1517
14. American Psychiatric Association (1994) Diagnostic and statistical manual of mental disor-
ders, 4th edn. American Psychiatric Association, Washington, DC
15. Skoog I (2008) Mental disorders associated with aging. In: Heggenhougen K, Quah S (eds)
International encyclopedia of public health, vol 6. Academic, San Diego, pp 174–182
16. Ernst C, Angst J (1995) Depression in old age. Is there a real decrease in prevalence?
A review. Eur Arch Psychiatry Clin Neurosci 245:272–287
17. Riedel-Heller SG, Busse A, Angermeyer MC (2006) The state of mental health in old-age
across the ‘old’ European Union – a systematic review. Acta Psychiatr Scand 113:388–401
18. Skoog I, Nilsson L, Landahl S et al (1993) Mental disorders and the use of psychotropic
drugs in an 85-year-old urban population. Int Psychogeriatr 5:33–48
19. Beekman ATF, Deeg DJH, van Tilburg T et al (1995) Major and minor depression in later
life: a study of prevalence and risk factors. J Affect Disord 36:65–75
20. Ritchie K, Artero S, Beluche I et al (2004) Prevalence of DSM-IV psychiatric disorder in the
French elderly population. Br J Psychiatry 184:147–152
21. McDougall FA, Kvaal K, Matthews FE et al (2007) Medical Research Council Cognitive
Function and Ageing Study. Prevalence of depression in older people in England and Wales:
the MRC CFA Study. Psychol Med 37:1787–1795
22. Steffens DC, Skoog I, Norton MC et al (2000) Prevalence of depression and its treatment in
an elderly population: the Cache County study. Arch Gen Psychiatry 57:601–607
23. Jorm AF (2000) Does old age reduce the risk of anxiety and depression? A review of epide-
miological studies across the adult life span. Psychol Med 30:11–22
24. Palsson S, Skoog I (1997) The epidemiology of affective disorders in the elderly: a review.
Int Clin Psychopharm 12(suppl 2):S3–S13
25. Palsson SP, Östling S, Skoog I (2001) The incidence of first-onset depression in a population
followed from the age of 70 to 85. Psychol Med 31:1159–1168
26. Palsson S, Johansson B, Berg S et al (2000) A population study on the influence of depres-
sion on neuropsychological functioning in 85-year-olds. Acta Psychiatr Scand
101:185–193
Neuropsychiatric Aspects of Vascular Cognitive Impairment 159
27. Persson G, Skoog I (1992) Subclinical dementia: relevance of cognitive and affective symp-
toms and signs. A nine year prospective study. J Geriatr Psychiatry Neurol 5:172–178
28. Copeland JRM, Davidson IA, Dewey ME et al (1992) Alzheimer’s disease, other dementias,
depression and pseudodementia: prevalence, incidence and three-year outcome in Liverpool.
Br J Psychiatry 161:230–239
29. Henderson AS, Korten AE, Jacomb PA et al (1997) The course of depression in the elderly:
a longitudinal community-based study in Australia. Psychol Med 27:119–129
30. Palsson S (2000) Population studies on depression in the elderly. Prevalence, incidence and
relation to cognitive function and dementia. Dissertation, University of Gothenburg
31. Ownby RL, Crocco E, Acevedo A et al (2006) Depression and risk for Alzheimer disease:
systematic review, meta-analysis, and metaregression analysis. Arch Gen Psychiatry
63:530–538
32. Skoog I (1993) The prevalence of psychotic, depressive and anxiety syndromes in demented
and non-demented 85-year-olds. Int J Geriatr Psychiatry 8:247–253
33. Savva GM, Zaccai J, Matthews FE et al (2009) Medical Research Council Cognitive
Function and Ageing Study. Prevalence, correlates and course of behavioural and psycho-
logical symptoms of dementia in the population. Br J Psychiatry 194:212–219
34. Petrovic M, Hurt C, Collins D et al (2007) Clustering of behavioural and psychological
symptoms in dementia (BPSD): a European Alzheimer’s disease consortium (EADC) study.
Acta Clin Belg 62:426–432
35. Östling S, Skoog I (2002) Psychotic symptoms and paranoid ideation in a nondemented
population-based sample of the very old. Arch Gen Psychiatry 59:53–59
36. Östling S, Börjesson-Hansson A, Skoog I (2007) Psychotic symptoms and paranoid ideation
in a population-based sample of 95-year-olds. Am J Geriatr Psychiatry 15:999–1004
37. Sigström R, Skoog I, Sacuiu S et al (in press) The prevalence of psychotic symptoms and
paranoid ideation in a non-demented population sample of 70-82 year olds. Int J Geriatr
Psychiatry
38. Östling S, Johansson B, Skoog I (2004) Cognitive test performance in relation to psychotic
symptoms and paranoid ideation in non-demented 85-year-olds. Psychol Med 34:443–450
39. Skoog I (2006) Vascular dementia. In: Pathy MSJ, Sinclair AJ, Morley JE (eds) Principles
and practice of geriatric medicine, 4th edn. Wiley, Chichester, pp 1103–1110
40. Breteler MMB, Claus JJ, Grobbee DE et al (1994) Cardiovascular disease and distribution of
cognitive function in elderly people: the Rotterdam Study. BMJ 308:1604–1608
41. Breteler MMB, van Swieten JC, Bots ML et al (1994) Cerebral white matter lesions, vascular
risk factors, and cognitive function in a population-based study: the Rotterdam Study.
Neurology 44:1246–1252
42. Breteler MMB, van Amerongen NM, van Swieten JC et al (1994) Cognitive correlates of
ventricular enlargement and cerebral white matter lesions on magnetic resonance imaging.
The Rotterdam Study. Stroke 25:1109–1115
43. Skoog I, Berg S, Johansson B et al (1996) The influence of white matter lesions on neurop-
sychological functioning in demented and non-demented 85-year-olds. Acta Neurol Scand
93:142–148
44. Linden T, Skoog I, Fagerberg B et al (2004) Cognitive impairment and dementia 20 months
after stroke. Neuroepidemiology 23:45–52
45. Qiu C, Skoog I, Fratiglioni L (2002) Occurrence and determinants of Vascular Cognitive
Impairment. In: Erkinjuntti T, Gauthier S (eds) Vascular cognitive impairment. Martin
Dunitz, London, pp 61–83
46. Tatemichi TK, Desmond DW, Mayeux R et al (1992) Dementia after stroke: baseline fre-
quency, risks, and clinical features in a hospitalized cohort. Neurology 42:1185–1193
47. Tatemichi TK, Desmond DW, Paik M et al (1993) Clinical determinants of dementia related
to stroke. Ann Neurol 33:568–575
48. Tatemichi TK, Paik M, Bagiella E et al (1994) Dementia after stroke is a predictor of long-
time survival. Stroke 25:1915–1919
160 I. Skoog
49. Pohjasvaara T, Erkinjuntti T, Vataja R et al (1997) Dementia three months after stroke.
Baseline frequency and effect of different definitions of dementia in the Helsinki Stroke
Aging Memory (SAM) cohort. Stroke 28:785–792
50. Liebetrau M, Steen B, Skoog I (2003) Stroke in 85-year-olds. Prevalence, incidence, risk
factors and relation to mortality and dementia. Stroke 34:2617–2622
51. Skoog I (1998) Guest editorial. Status of risk factors for vascular dementia. Neuroepidemiology
17:2–9
52. Barker WW, Luis CA, Kashuba A et al (2002) Relative frequencies of Alzheimer disease,
Lewy body, vascular and frontotemporal dementia, and hippocampal sclerosis in the State of
Florida Brain Bank. Alzheimer Dis Assoc Disord 16:203–212
53. Fischer P, Gatterer G, Marterer A et al (1990) Course characteristics in the differentiation
of dementia of the Alzheimer type and multi-infarct dementia. Acta Psychiatr Scand
81:551–553
54. Ferrucci L, Guralnik J, Salive M et al (1996) Cognitive impairment and risk of stroke in the
older population. J Am Geriatr Soc 44:237–241
55. Gale C, Martyn C, Cooper C (1996) Cognitive impairment and mortality in a cohort of
elderly people. Br Med J 312:608–611
56. Zhu L, Fratiglioni L, Guo Z et al (2000) Incidence of stroke in relation to cognitive function
and dementia in the Kungsholmen Project. Neurology 54:2103–2107
57. Folstein MF, Maiberger R, McHugh PR (1977) Mood disorder as a specific complication of
stroke. J Neurol Neurosurg Psychiatry 40:1018–1020
58. Rao R, Jackson S, Howard R (2001) Depression in older people with mild stroke, carotid
stenosis and peripheral vascular disease: a comparison with healthy controls. Int J Geriatr
Psychiatry 16:175–183
59. Burvill PW, Johnson GA, Jamrozik KD et al (1995) Prevalence of depression after stroke:
the Perth Community Stroke Study. Br J Psychiatry 166:320–327
60. Ng KC, Chan KL, Straughan PT (1995) A study of post-stroke depression in a rehabilitative
center. Acta Psychiatr Scand 92:75–79
61. Pohjasvaara T, Leppavuori A, Siira I et al (1998) Frequency and clinical determinants of
poststroke depression. Stroke 29:2311–2317
62. Linden L, Blomstrand C, Skoog I (2007) Depressive disorders after 20 months in elderly
stroke patients. A case-control study. Stroke 38:1860–1863
63. Astrom M, Adolfsson R, Asplund K (1993) Major depression in stroke patients. A 3-year
longitudinal study. Stroke 24:976–982
64. Kauhanen ML, Korpelainen JT, Hiltunen P et al (2000) Aphasia, depression, and non-verbal
cognitive impairment in ischaemic stroke. Cerebrovasc Dis 10:455–461
65. Hackett ML, Yapa C, Parag V et al (2005) Frequency of depression after stroke: a systematic
review of observational studies. Stroke 36:1330–1340
66. Hackett ML, Anderson CS (2005) Predictors of depression after stroke: a systematic review
of observational studies. Stroke 36:2296–2301
67. Ramasubbu R, Robinson RG, Flint AJ et al (1998) Functional impairment associated with
acute poststroke depression: the Stroke Data Bank Study. J Neuropsychiatry Clin Neurosci
10:26–33
68. Schulz R, Beach SR, Ives DG et al (2000) Association between depression and mortality in
older adults: the Cardiovascular Health Study. Arch Intern Med 160:1761–1768
69. Pohjasvaara T, Vataja R, Leppavuori A et al (2001) Depression is an independent predictor
of poor long-term functional outcome post-stroke. Eur J Neurol 8:315–319
70. Schubert DS, Taylor C, Lee S et al (1992) Detection of depression in the stroke patient.
Psychosomatics 33:290–294
71. Kotila M, Numminen H, Waltimo O et al (1999) Post-stroke depression and functional recov-
ery in a population-based stroke register. The Finnstroke study. Eur J Neurol 6:309–312
72. Beblo T, Wallesch CW, Herrmann M (1999) The crucial role of frontostriatal circuits for
depressive disorders in the postacute stage after stroke. Neuropsychiatry Neuropsychol
Behav Neurol 12:236–246
73. Vataja R, Pohjasvaara T, Leppavuori A et al (2001) Magnetic resonance imaging correlates
of depression after ischemic stroke. Arch Gen Psychiatry 58:925–931
Neuropsychiatric Aspects of Vascular Cognitive Impairment 161
74. Starkstein SE, Robinson RG, Honig MA et al (1989) Mood changes after right-hemisphere
lesions. Br J Psychiatry 155:79–85
75. Carson AJ, MacHale S, Allen K et al (2000) Depression after stroke and lesion location: a
systematic review. Lancet 356:122–126
76. Kneebone II, Dunmore E (2000) Psychological management of post-stroke depression. Br J
Clin Psychol 39:53–65
77. Bos MJ, Lindén T, Koudstaal PJ et al (2008) Depressive symptoms and risk of stroke: the
Rotterdam Study. J Neurol Neurosurg Psychiatry 79:997–1001
78. Liebetrau M, Steen B, Skoog I (2008) Depression as a risk factor for the incidence of first-
ever stroke in 85-year-olds. Stroke 39:1960–1965
79. Salaycik KJ, Kelly-Hayes M, Beiser A et al (2007) Depressive symptoms and risk of stroke:
the Framingham Study. Stroke 38:16–21
80. Frasure-Smith N, Lesperance F, Talajic M (1995) Depression and 18-month prognosis after
myocardial infarction. Circulation 91:999–1005
81. Musselman DL, Tomer A, Manatunga AK et al (1996) Exaggerated platelet reactivity in
major depression. Am J Psychiatry 153:1313–1317
82. Okamura F, Tashiro A, Utumi A et al (2000) Insulin resistance in patients with depression and its changes
during the clinical course of depression: minimal model analysis. Metabolism 49:1255–1260
83. Vermeer SE, Koudstaal PJ, Oudkerk M et al (2002) Prevalence and risk factors of silent brain
infarcts in the population-based Rotterdam Scan Study. Stroke 33:21–25
84. Vermeer SE, Prins ND, Den Heijer T et al (2003) Silent brain infarcts and the risk of demen-
tia and cognitive decline. N Engl J Med 348:1215–1222
85. Liebetrau M, Steen B, Hamann GF et al (2004) Silent and symptomatic infarcts on cranial
computerized tomography in relation to dementia and mortality. A population-based study in
85-year-olds. Stroke 35:1816–1820
86. Fernando MS, Ince PG, on behalf of the MRC Cognitive function and Ageing Neuropathology
Study Group (2004) Vascular pathologies and cognition in a population-based cohort of
elderly people. J Neurol Sci 226:13–17
87. Román GC (1987) Senile dementia of the Binswanger type. A vascular form of dementia in
the elderly. JAMA 258:1782–1788
88. Brun A, Englund E (1986) A white matter disorder in dementia of the Alzheimer type:
A pathoanatomical study. Ann Neurol 19:253–262
89. Englund E, Brun A (1990) White matter changes in dementia of Alzheimer’s type. The dif-
ference in vulnerability between cell compartments. Histopathology (Oxf) 16:433–439
90. Skoog I, Gustafson D (2003) Hypertension, hypertension-clustering factors and Alzheimer’s
disease. Neurol Res 25:675–680
91. Guo X, Pantoni L, Simoni M et al (2009) Midlife blood pressure and late-life white matter
lesions: the Prospective Population Study of Women in Gothenburg, Sweden. Hypertension
54:57–62
92. Skoog I, Palmertz B, Andreasson L-A (1994) The prevalence of white matter lesions on
computed tomography of the brain in demented and non-demented 85-year-olds. J Geriatr
Psychiatry Neurol 7:169–175
93. Lopez OL, Becker JT, Jungreis CA et al (1995) Computed tomography–but not magnetic
resonance imaging–identified periventricular white-matter lesions predict symptomatic cere-
brovascular disease in probable Alzheimer’s disease. Arch Neurol 52:659–664
94. Simoni M, Pantoni L, Pracucci G et al (2008) Prevalence of CT-detected cerebral abnormali-
ties in an elderly Swedish population sample. Acta Neurol Scand 118:260–267
95. De la Monte S (1989) Quantitation of cerebral atrophy in preclinical and end-stage
Alzheimer’s disease. Ann Neurol 25:450–459
96. Prins ND, van Dijk EJ, den Heijer T et al (2004) Cerebral white matter lesions and the risk
of dementia. Arch Neurol 61:1531–1534
97. Gunstad J, Brickman AM, Paul RH et al (2005) Progressive morphometric and cognitive
changes in vascular dementia. Arch Clin Neuropsychol 20:229–241
98. Schmidt R, Enzinger C, Ropele S et al (2003) Austrian Stroke Prevention Study. Progression
of cerebral white matter lesions: 6-year results of the Austrian Stroke Prevention Study.
Lancet 361:2046–2048
162 I. Skoog
99. Herrmann LL, Le Masurier M, Ebmeier KP (2008) White matter hyperintensities in late life
depression: a systematic review. J Neurol Neurosurg Psychiatry 79:619–624
100. Steffens DC, Krishnan KR, Crump C et al (2002) Cerebrovascular disease and evolution of
depressive symptoms in the cardiovascular health study. Stroke 33:1636–1644
101. Teodorczuk A, Firbank MJ, Pantoni L et al (2009) Relationship between baseline white-
matter changes and development of late-life depressive symptoms: 3-year results from the
LADIS study. Psychol Med 12:1–8
102. Greenwald BS, Kramer-Ginsberg E, Krishnan KR et al (1998) Neuroanatomic localization
of magnetic resonance imaging signal hyperintensities in geriatric depression. Stroke
29:613–617
103. O’Brien J, Ames D, Chiu E et al (1998) Severe deep white matter lesions and outcome in
elderly patients with major depressive disorder: follow up study. BMJ 317:982–984
104. Simpson S, Baldwin RC, Jackson A et al (1998) Is subcortical disease associated with a poor
response to antidepressants? Neurological, neuropsychological and neuroradiological find-
ings in late-life depression. Psychol Med 28:1015–1026
105. Chen PS, McQuoid DR, Payne ME et al (2006) White matter and subcortical gray matter
lesion volume changes and late-life depression outcome: a 4-year magnetic resonance imag-
ing study. Int Psychogeriatr 18:445–456
106. Lim A, Tsuang D, Kukull W et al (1999) Clinico-neuropathological correlation of Alzheimer’s
disease in a community-based case series. J Am Geriatr Soc 47:564–569
107. Nagy Z, Esiri M, Jobst K et al (1997) The effects of additional pathology on the cognitive
deficit in Alzheimer disease. J Neuropathol Exp Neurol 56:165–170
108. Heyman A, Fillenbaum G, Welsh-Bohmer K et al (1998) Cerebral infarcts in patients with
autopsy-proven Alzheimer’s disease: CERAD, part XVIII. Consortium to Establish a
Registry for Alzheimer’s Disease. Neurology 51:159–162
109. Etiene D, Kraft J, Ganju N et al (1998) Cerebrovascular pathology contributes to the hetero-
geneity of Alzheimer’s disease. J Alzheimers Dis 1:119–134
110. Erkinjuntti T, Hachinski V (1993) Dementia post stroke. In: Teasell R (ed) Physical medicine
and rehabilitation: state of the art reviews, vol 7. Hanley & Belfus, Philadelphia, pp 195–212
111. Pasquier F, Leys D (1997) Why are stroke patients prone to develop dementia? J Neurol
244:135–142
112. Skoog I, Nilsson L, Palmertz B et al (1993) A population-based study of dementia in
85-year-olds [see comments]. N Engl J Med 328:153–158
113. Wetterling T, Kanitz R-D, Borgis K-J (1996) Comparison of different diagnostic criteria for
vascular dementia (ADDTC, DSM-IV, ICD-10, NINDS-AIREN). Stroke 27:30–36
114. Amar K, Wilcock G, Scott M (1996) The diagnosis of vascular dementia in the light of the
new criteria. Age Ageing 25:51–55
115. Skoog I, Gustafson D (2006) Lessons learned from primary prevention trials in dementia. In:
Rockwood K, Gauthier S (eds) Trial designs and outcomes in dementia therapeutic research.
Taylor & Francis, Abingdon, pp 189–211
Neuropsychiatric Complications
of Cerebrovascular Disease
Introduction
StrokeisthethirdleadingcauseofdeathintheUnitedStates,thesecondleading
cause of death worldwide, and a major cause of long-term physical and mental dis-
abilityinstrokesurvivors.WithestimatesbytheAmericanStrokeAssociationof
750,000 strokes occurring annually in the United States, there are likely about
4.5 million stroke survivors in America today [1]. Additionally, economic costs of
strokeintheUnitedStateshavebeenestimatedtobeatleast$43billioneachyear
[2]. A major portion of these costs are allocated toward the treatment of secondary
complications, especially disability caused by depression, cognitive impairment,
and other neuropsychiatric complications.
The symptomatology of stroke survivors is an array of comorbid symptoms,
many of which were previously assumed to be of the same etiology or interrelated.
However, given recent research among this patient population, more understanding
has been gathered regarding the individual pathogenesis for many of these complica-
tions.Someoftheneuropsychiatricsequelaesecondarytocerebrovasculardisease
and their hypothesized mechanisms will be discussed here as the individual entities
thattheyare.Itisinthiswaythatthecliniciancanmorecloselyexamineeachsymp-
tom the individual patient suffers from and appropriately treat the exact cause of that
symptom rather than approaching collective complaints and applying a generalized
treatment plan. This approach may prove to be useful in the rehabilitation and treatment
of neuropsychiatric complications in stroke patients leading to historical recovery in
the quality of life of these patients and their loved ones.
Mania
Mania secondary to stroke, given its rarity and likely underdiagnosis, has limited
appearancethroughouttheneuropsychiatricliterature.Inpriorstudieslookingatneu-
ropsychiatric complications following stroke, enrolled patients uncommonly suffered
from poststroke mania in comparison to other psychiatric complaints [1].Oftwomajor
studies investigating poststroke complications, one following 700 consecutive stroke
patients [3] and another looking at 661 stroke survivors [4], only 6 patients in total
between the two studies presented with mania secondary to stroke. Additionally, in two
other considerably large community studies, the Oxfordshire Community Stroke
ProjectandthePerthCommunityStrokeProject,bothofwhichwereinvestigatingthe
prevalence of neuropsychiatric disorders following stroke, zero patients were found to
suffer from poststroke mania [1]. When witnessed, however, mania secondary to
stroke demonstrates very similar symptomatology in comparison to patients suffering
from primary mania. These frequently include elation, pressured speech, insomnia,
grandiosity, and flight of ideas with racing thoughts. Although the prognostic charac-
teristics of poststroke mania are limited, it does appear that a family history of mood
disorder is correlated with a higher risk of mania after stroke [1].
Despite the infrequency of poststroke mania, the lesion localization in patients
suffering from secondary mania is more definitively understood. Most hypotheses
related to mania and stroke location concur that right hemispheric strokes are likely
responsibleforthemanifestationofthissymptom.Priorstudieshavediscussedthat
lesions in the right hemisphere lead to anterior limbic circuit dysfunction involving
the orbitofrontal and basotemporal cortex in addition to the head of the caudate and
dorsomedial thalamic nucleus. It has been hypothesized that a more generalized
cerebral distribution may be responsible for the rarity of this symptom, given that
both subcortical and limbic involvement is necessary for manic symptoms [1].
NeuropsychiatricComplicationsofCerebrovascularDisease 165
Anxiety
The DSM-IV-TR criteria for primary generalized anxiety disorder includes the
presence of a sustained worrying state, in addition to a minimum of three other anxiety
symptoms, such as concentration difficulties, irritability, muscle tension, sleep
disturbances, restlessness, and decreased energy, for a period of at least 6 months.
Althoughnotasprevalentaspoststrokedepression(PSD),anxietyhasalsocometo
be regarded as a major neuropsychiatric complication severely impacting the daily
functioningandqualityoflifeofstrokesurvivors.In1997,Robinsonreportedamean
prevalenceofpoststrokeanxietytobe14.4%,withsomestudiesreportingupto28%;
however, with improved screening and diagnosis, this value is surely more common
today [7]. Recent studies have demonstrated that patients who present with anxiety
disorder after suffering a stroke are more likely to have impairment in psychosocial
functioningandactivitiesofdailyliving(AODL)thanpatientswithgeneralizedanxiety
disorder not suffering from cerebrovascular disease [7]. Additionally, these patients
were at a greater risk of comorbid insomnia and depression than their nonstroke
166 M. Gaviria and R.D. Verzal
counterparts. These findings are not only significant for the increased burden of disease
on the patient but have ramifications regarding the prescribing of sleep-promoting and
anxiolytic drugs, which also contribute to further difficulty in daily functioning.
IncontrasttostudiesinvestigatingPSD,aprevioushistoryofmigrainesand/or
epilepsy before the insighting cerebrovascular event seem to increase the risk of
poststroke anxiety [7].Inaddition,anxietysecondarytostrokewasrelatedtothe
following demographic and clinical factors: smoking, migraine, epilepsy, previous
mental disorders, comorbid depression and insomnia, stroke severity, and impair-
mentinpsychosocialfunctioningandinAODL.Womenalsoseemtobemorelikely
to suffer anxiety secondary to stroke than men, although reasons for this finding are
still under investigation.
Patients diagnosed with generalized anxiety disorder secondary to stroke
demonstrated lesion localization predominantly in the territory supplied by anterior
circulation.Strokesurvivorswhosufferedalesionintheanteriorcirculationdemon-
strated worse scores on the anxiety scale than those survivors with lesions in the
posterior circulation [8]. This finding may support recent hypotheses that suggest a
distinction between the mechanism of poststroke anxiety and generalized anxiety.
Severalrecentstudiesreportthatalthoughlefthemisphericstrokeswerefound
to cause comorbid depression and anxiety disorders, isolated anxiety secondary to
stroke was localized to right hemispheric lesions [5], although a more specific
lesion localization has yet to be identified.
Treatment modalities used for patients suffering from poststroke anxiety have
primarily been based on those implicated for patients with primary generalized
anxiety disorder. Studies on therapy for anxiety secondary to stroke are limited.
Thus far, the recommended treatment is benzodiazepines, excluding the elderly
population given reports of significant side effects. The preferable therapeutic
modality in elderly patients who are unable to tolerate benzodiazepines are buspirone
andselectiveserotoninuptakeinhibitors(SSRIs).
Apathy
Symptomaticlesionshavebeenfoundinevendistributionbilaterallythroughout
the brain among apathetic and nonapathetic stroke patients. Continued research is
warranted by the recurrent inconsistencies regarding lesion localization in patients
suffering from poststroke apathy. Chemerinski et al. reported a collective literature
review demonstrating the occurrence of 93 left-sided lesions, 77 right-sided
lesions, and 94 bilateral strokes resulting in secondary apathy [1].Severalfactors
that have been previously correlated with apathy following stroke include cogni-
tivedysfunction,advancedage,deficitsinAODL,decreasedattentionandinfor-
mation processing, and poor fluency [1]. When examining the functional level of
stroke survivors, apathetic patients demonstrate a significantly lower recovery
level in aspects of self-care. This aspect is of great importance when evaluating
caregiver burden and the long-term ramifications that these patients and their fami-
lies face during stroke rehabilitation.
Treatment recommendations for poststroke apathy include the use of various
stimulating antidepressants, such as fluoxetine, for first-line therapy. Additionally,
other stimulating agents, such as amphetamine, methylphenidate, selegiline, and
bupropionhavebeenusedsuccessfullytotreatapathy;however,onlyincasereports.
Recent studies have examined the role of cholinesterase inhibitors in the treatment
of apathy given the activating properties of these agents and the correlation with
poststroke apathy and cholinergic deficits [11].
Sleepdisordershaverecentlybecomeanareaofmedicalinterestgiventheirsignificant
impactonapatient’sdailyliving.Pathologyofsleepisaspectrumofabnormalities
thataffectsleeptime,thesleepcycle,andbreathingandactivityduringsleep.One
example includes dyssomnias, such as insomnia and hypersomnia, in which a patient
hasanalterationintheamountoftimeasleepwithinagiven24-hperiod.Onaver-
age,7–9hofsleeppernightisconsideredadequatebytheNationalSleepFoundation
intheUnitedStates,althoughtherearesomenormalvariantsthatslightlydeviate
from this amount. Chen et al. defined short-sleep duration as a night’s sleep of 6 h
or less and a long-sleep duration as more than 9 h of sleep [12].Othersleepdisorders
involve disruption in the normal pattern of the sleep cycle between non-REM and
REM (rapid eye movement sleep). Abnormalities within either the amount or
cycling of the sleep cycle can consequentially lead to deficits in attention, memory,
daily functioning, an increase in accidents, and a diminishment of overall health.
Inpatientswhohavesufferedstroke,thereductioninqualityoflifehasevoked
great focus. Although the etiology of this change is multifactorial, disturbance of the
sleep–wakecycleinthesepatientsappearstobeasignificantcontributor.Long-term
follow-up of stroke survivors who have suffered from both ischemic stroke and/or
subarachnoid hemorrhage reveals difficulty in initiation of sleep, irritability, loss of
interests, poor concentration, fatigue, and falling asleep at inappropriate times
throughout the day. Assessment of functional outcome, and therefore quality of life,
168 M. Gaviria and R.D. Verzal
hasbeenevaluatedinstrokepatientsusingtheRankinScale,a6-pointhandicapscale
focusing on restrictions of lifestyle in coordination with polysomnographic studies.
It appears that up to one-third of stroke survivors suffer from either insomnia,
excessive daytime sleepiness, or both, resulting in lower values when scoring quality
oflife.Itisimportanttonotethatsleepdisturbancescausedbystrokemustbediffer-
entiated from sleep disturbances that are secondary to other neuropsychiatric compli-
cationsofstroke,suchasdepression.Forexample,Schuilingetal.reportedthatsleep
disturbances resulting from depression are characteristically demonstrated by short-
REM sleep latency and an increased amount of REM sleep overall, whereas sleep
disturbances that do not show this pattern, but occur after stroke, are more likely a
result of the stroke alone and may secondarily lead to the initiation of sleep disturbance
depression [13].Landauetal.demonstratedlesionlocalizationinpatientswithsleep
disturbances secondary to stroke concentrated in the right pons, followed by bilateral
pontine strokes, and then left pontine lesions [14].Inpatientsdiagnosedwithrestless
leg syndrome following stroke, Kim et al. demonstrated lesion topography predomi-
nantly in subcortical regions, such as the pyramidal tract and basal ganglia–brainstem
axis [15]. This result reinforces that compromise of cerebral perfusion in areas of the
brain controlling motor function and the sleep–wake cycles may result in restlessness
symptoms. Treatment modalities available for these symptoms include dopamine ago-
nists, which have delivered relatively significant relief in symptoms [15].
Fatigue
Fatigue has been known to be a common sequelae in stroke survivors, with a prevalence
rangingfrom38%to68%[16]. Treating this phenomenon has proven to be quite
challenging given the lack of established standards in its measurement and diffi-
culty in determining its exact etiology. Additionally, the diagnostician must take
into consideration other possible contributing factors associated with fatigue from
whichnumerousstrokepatientssufferfrommonthstoyearsaftertheincitingevent;
these include mental factors, such as depression and anxiety, but also might be
related to physical deconditioning and gross motor deficits. Because the etiology of
fatigue has proven to be so complex, it is not until recently that research has began
to control for some of these confounding factors and evaluate a more central origin
of fatigue in stroke survivors.
Inastudyinvestigatingfatigueinstrokesurvivors,patientswhosufferedfrom
minor strokes were compared to those who experienced a transient ischemic attack
(TIA).Whilepreviousstudiessuggestedthatfatigueafterstrokewasattributableto
compensatory behaviors related to gross neurological deficits, the comparison used
in this study demonstrated that poststroke fatigue may be of central origin rather
than physical sequelae secondary to increased effort required after suffering a
stroke [16]. By investigating patients who suffered from minor rather than major
strokes, there were minimal to no residual neurological deficits to which secondary
poststroke fatigue might be attributable. These results were the first set of published
data investigating fatigue in relationship to stroke using this approach.
NeuropsychiatricComplicationsofCerebrovascularDisease 169
The study evaluated 149 patients: 73 status post minor stroke and 67 status post
TIA.Demographiccharacteristics,recentlifeevents,vascularriskfactors,medica-
tions, and mental and physical disabilities were all variables that were controlled
among the two patient groups. Fatigue was evaluated at 6 months after minor stroke
orTIAusingtheChalderFatigueScale[17]. The results of the study demonstrated
that patients who suffered from a minor stroke had a 56% fatigue prevalence in
comparisontotheTIApatientswithaprevalenceof29%.Thisfindingisonethat
should serve to motivate neuropsychiatrists and other clinicians to move toward
the consideration of a central mechanism underlying fatigue in patients who
have suffered a stroke. Additionally, collaborative efforts between researchers,
physicians, and pharmaceutical experts should investigate central-acting treatment
modalities for these patients. Treatment is mainly symptomatic, with the use of
stimulating agents, in addition to treating concurrent neuropsychiatric complaints.
Sexual Dysfunction
Inadultpatients,cerebrovasculardiseasecausesgreatersecondaryimpairmentthan
anyothermajorillnessintheUnitedStates.Perhapsoneofthemostlimitedareas
of investigation among poststroke complications is that of sexual dysfunction.
Historically, the pathogenesis of sexual dysfunction was viewed as predominantly a
psychogenic disorder; however, after more closely investigating patients suffering
from various sexual impairments, additional hypotheses have begun to reveal them-
selves.Priorstudieshavedemonstratedthatsexualdysfunctionafterstrokeismulti-
factorial, with both organic and psychological etiologies at its origin. Organic
influences on sexual function include certain comorbid medical conditions, such as
diabetes,hypertension,andcardiacdisease.Psychosocialfactorsincludelossofself-
esteem, fear of another stroke, and changes within the spousal relationship [18].
More recently, physicians have recognized that in stroke patients there likely exists
both a psychogenic and neurogenic pathophysiology to erectile dysfunction.
Although most research on sexual dysfunction has involved male patients, there
is no doubt that this is a complication experienced by both male and female patients.
OneScandinavianstudyreportedadeclineinsexualdesireinthree-quartersofmale
stroke patients and two-thirds of female stroke patients [19]. From previous studies,
it is evident that major complications of concern include decreased sexual desire in
both male and female patients, diminished vaginal lubrication and orgasm in female
patients, and impairment in erection and ejaculation in men [18]. Additionally,
these poststroke consequences extend beyond the individual to also affect their
intimate relationships with their partners.
A recent study investigating sexual dysfunction in male stroke patients was aimed
at correlating this impairment with brain localization of the stroke lesions [20];this
appears to be the first study to correlate specific aspects of sexual dysfunction with
localization of strokes. The comparison was made between 109 male stroke patients
and 109 aged-matched controls. A five-item version of the International Index of
Erectile Function was use to evaluate changes in sexual desire, ejaculatory function,
170 M. Gaviria and R.D. Verzal
and sexual satisfaction after stroke. These results were then analyzed with the
location of the respective patients’ brain lesions.
Overall,thestudydemonstratedasignificantdecreaseinerectilefunctioninthe
stroke patient group. Frequency of sexual intercourse and sexual desire were both
significantly reduced in this patient group. The diminished frequency in intercourse
was found to be most commonly (almost 60%) secondary to the lack of sexual
desire after stroke. More specifically, stroke patients suffering from ejaculation
disorders demonstrated lesions in the right cerebellum. Additionally, patients expe-
riencing decreases in sexual desire showed imaging correlations such that their
brain lesions were concentrated to the left basal ganglia. Overall, these findings
were able to localize patterns with certain aspects of sexual dysfunction. A larger
and more inclusive study would likely be helpful in confirming these associations
between sexual dysfunction and the respective neuroanatomy.
The common implication among studies evaluating poststroke sexual dysfunc-
tion is such that these impairments should be more frequently addressed and
aggressively treated during the rehabilitation process in stroke survivors so that
they can enjoy a healthier quality of life and continue to share and participate in
intimate relationships with their partners. Although poststroke rehabilitation has
shown to help in the recovery of sexual functioning, additional therapies used for
sexual dysfunction unrelated to stroke may also be used.
Cognitive Performance
Neurosciencehasdemonstratedtheassociationbetweenincreasedcognitiveimpairment
followinginfarction;however,thepathophysiologybywhichinfarctsresultinthis
cognitive dysfunction is incompletely understood. A popular hypothesis involves the
disruption of the cerebral circuitry with resultant deficits in cognition, usually with
thefrontal-subcorticalcircuitsbeingaffected.Injurytothesecircuitsresultsindeficits
in memory, information processing, and executive dysfunction. Additionally, disrup-
tion of cerebro-cerebellar circuits often causes problems in higher cognitive functions.
Inparticular,manystudieshaveexaminedtheimpactoflocationandnumberof
infarcts on cognitive impairment. In a recent study by Saczynski and colleagues,
three separate cognitive domains were investigated following the diagnosis of
cerebralinfarct:processingspeed,memory,andexecutivedysfunction.Inthe4,030
nondemented participants enrolled in the Age Gene/Environment Susceptibility –
ReykjavikStudy,itwasfoundthatthosepatientswhosufferedinfarctsinmultiple
locations had slower processing speed, poorer performance in memory, and execu-
tive dysfunction in comparison to patients with infarcts concentrated to a single
location.Interestingly,thoseparticipantswhodidsuffermultipleinfarctswithinone
region did not demonstrate any cognitive discrepancies from the noninfarct control
group after model adjustment [21]. These findings are all independent of white matter
lesions, brain atrophy, cardiovascular comorbidities, and depressive symptoms.
Treatment modalities for post-stroke cognitive impairment include the use of acetyl-
cholinesterase inhibitors, in addition to recent developments, such as the use of
NeuropsychiatricComplicationsofCerebrovascularDisease 171
repetitivetranscranialmagneticstimulation(TMS).Afocallesionsuchasastroke
may produce a state of hemispheric imbalance. TMS can be used to repair this
disequilibrium between the ipsi-lesional and contra-lesional cerebrum.
exclude this diagnosis as most patients with corticobulbar infarcts will present
without them [1]. With the use of the Pathological Laughter and Crying Scale,
previous studies have demonstrated the effectiveness of using nortriptyline for 4–6
weekstotreatpatientswithIEED[24]. Additionally, a double-blind drug trial using
citalopraminpatientswithpoststrokeIEEDalsodemonstratedareductioninthe
number of crying spells by more than 50% [25].
Irritability
Psychosis
Incomparisontomanyoftheotherneuropsychiatriccomplicationsofstroke,psychotic
symptoms,suchashallucinationsanddelusions,arerare.Onlyfivepatientsfollowed
in a 9-year longitudinal study demonstrated symptoms consistent with poststroke
NeuropsychiatricComplicationsofCerebrovascularDisease 173
psychosis [29]. Another study following stroke patients up to 11 years later found a
total of eight patients who exhibited psychotic symptoms after right-sided temporopa-
rieto-occipital infarcts [30]. Many of these stroke patients also experienced seizures
closetothetimeatwhichpsychosiswasobserved.Improvementoftheirpsychotic
symptoms was noted after treatment with antiepileptic medications [30].
A frontal lobe syndrome and psychosis have been observed after infarction in
the brainstem dopaminergic nuclei, specifically at the ponto-mesencephalic junc-
tion.StereotacticlesionlocalizationonMRIinadditiontoconcordantanalysisof
regional cerebral blood flow demonstrated that infarction within these nuclei likely
has resultant disruption of the ascending dopaminergic projections to the frontal-
subcortical circuit components [31]. In addition, a longitudinal study of stroke
patients demonstrated that right frontoparietal lesions and subcortical atrophy were
significant risk factors for psychosis following stroke given that nonpsychotic
stroke survivors did not have these findings [29].
Agitation
Agitationfollowingstrokeisobservedinapproximately28%ofpatientswhohave
sufferedfromacerebrovascularevent.Poststrokeagitationmanifestsprimarilyby
stubbornness(81%),noncompliance,suchaswithrehabilitation,refusaltocooperate
with the patient’s caregiver (75%), crying and cursing (75%), and less commonly
violencetowardobjects,suchasslammingdoors(2%),andneverviolencetoward
people [26].Lesionsresultinginpoststrokeagitationareneuroanatomicallyconfined
to the bitemporal lobes and/or the amygdale [28]. The current treatment recom-
mendation includes mood stabilizing agents.
Depression
Conclusion
References
1. ChemerinskiE,LevineSR(2006)Neuropsychiatricdisordersfollowingvascularbraininjury.
MtSinaiJMed73(7):1006–1014
2. HadidiN,Treat-JacobsonDJ,LindquistR(2009)Poststrokedepressionandfunctionalout-
come:acriticalreviewofliterature.HeartLung38(2):151–162
3. StarksteinSE,BostonJ,RobinsonRG(1988)Mechanismsofmaniaafterbraininjury:12case
reportsandreviewoftheliterature.JNervMentDis176:87–100
4. DunneJW,LeedmanPJ,EdisRH(1986)Inobviousstroke:acauseofdeliriumanddementia.
AustNZJMed16(6):771–778
5. BraunCMJ,DaigneaultR,GaudeletS(2008)DSMIVsymptomsofmania:whichonesresult
moreoftenfromrightthanlefthemispherelesions?ComprPsychiatry49:441–459
6. FilardidaRochaF,HorizonteB,CarneiroJG(2008)Poststrokemanicsymptoms:anunusual
neuropsychiatriccondition.RevBrasPsiquiatr30(2):168–176
7. LeppavuoriA,PohjasvaaraT,VatajaRetal(2003)Generalizedanxietydisordersthreetofour
monthsalterischemicstroke.CerebrovascDis16:257–264
Table 1 Neuropsychiatricsymptomsandlesionlocalization(modiiedfrom[3])
Neuropsychiatricsymptoms Lesionlocalization Treatment
Depression Lcerebralcortex,frontallobe,frontosubcorticalcircuits, Fluoxetine, followed by nortriptyline, and in
basal ganglia and limbic system some cases, citalopram, trazadone, and
methylphenidate
Mania Anterior region of bilateral hemispheres, R temporal lobe, Olanzapine,carbamazepine,andvalproicacid
R caudate nucleus, R subcortical region, R ventral pons,
Lbasalganglia
Apathy Frontal lobe, cingulate gyrus, supplemental motor area, Stimulatingantidepressants,i.e.,fluoxetine.
amygdala,capsule,insula,caudatenucleus/nuclei, Stimulatingagents,i.e.,amphetamine,
bilateral anterior thalamic nuclei methylphenidate,
selegiline,bupropion.AChE-I
IEED Bilateralcorticobulbartracts,Lfrontalcortex Nortriptyline,citalopram
Psychosis R hemisphere, especially temporo-parieto-occipital or Antiepileptics
frontoparietal, and thalamus
Agitation Bitemporal, amygdala Mood stabilizing agents
Disturbance of sleep–wake cycle Pons(R>bilateral>L),subcorticalregionssuchasthe Dopamine agonists
NeuropsychiatricComplicationsofCerebrovascularDisease
8. Dennis M, O’Rrourke S, Lewis S, Sharpe M, Warlow C (2000) Emotional outcomes after
stroke:factorsassociatedwithpooroutcome.JNeurolNeurosurgPsychiatry68:47–52
9. StarksteinSE,FedoroffJP,PriceTRetal(1993)Apathyfollowingcerebrovascularlesions.
Stroke24:1625–1631
10. SantaN,SugimoriH,KusudaKetal(2008)Apathyandfunctionalrecoveryfollowingfirst-
everstroke.IntJRehabilRes31(4):321–326
11. ParanthamanG,BalwinR(2006)Treatmentofpsychiatricsyndromesduetocerebrovascular
disease.IntRevPsychiatry18(5):453–470
12. ChenJC,BrunnerRL,RenHetal(2008)Sleepdurationandischemicstrokeinolderwomen.
Stroke39:315–3192
13. Schuiling WJ, Rinkel GJE, Walchenbach R et al (2005) Disorders of sleep and wake in
patientsafterSAH.Stroke36:578–582
14. Landau ME, Maldonado JY, Jabbari B (2005) The effects of isolated brainstem lesions on
humanREMsleep.SleepMed6:37–40
15. Lee SJ, Kim JS, Song IU (2009) Poststroke restless legs syndrome and lesion location:
anatomicalconsiderations.MovDisord24(1):77–84
16.WinwardCetal(2009)FatigueafterTIAandminorstroke.Stroke40(3):757–766
17. Chalder T, Berelowitz G, Pawlikowska T et al (1993) Development of a fatigue scale. J
PsychosomRes37(2):147–153
18. Tamtam Y, Tamtam L, Akil E et al (2008) Post-stroke sexual functioning in first stroke
patients.EurJNeurol15:660–666
19. SjogrenK,DamberJE,LiliequistB(1983)Sexualityafterstrokewithhemiplegia:aspectsof
sexualfunction.ScandJRehabilMed15:55–61
20.JungJ-H,KamS-C,ChoiS-M(2008)Sexualdysfunctioninmalestrokepatients:correlation
betweenbrainlesionsandsexualfunction.Urology71:99–103
21. SaczynskiJS,SigurdssonS,JonsdottirMKetal(2009)Cerebralinfarctsandcognitiveper-
formance.Stroke40:677–682
22. CummingsJL,ArciniegasDB,BrooksBR(2006)Defininganddiagnosinginvoluntaryemo-
tionalexpressiondisorder.CNSSpectr11(6suppl6):1–7
23. Cummings JL (2007) Involuntary emotional expression disorder: definition, diagnosis and
measurementscales.CNSSpectr12(4suppl5):11–16
24. RobinsonRG,ParikhRM,LipseyJRetal(1993)Pathologicallaughingandcryingfollowing
stroke:validationofameasurementscaleanddouble-blindtreatmentstudy.AmJPsychiatry
150:286–293
25. Andersen G, Verstergaard K, Riis JO et al (1993) Citalopram for post-stroke pathological
crying.Lancet342:837–839
26. AngelelliP,PaolucciS,BivonaUetal(2004)Developmentofneuropsychiatricsymptomsin
poststrokepatients:across-sectionalstudy.ActaPsychiatrScand110:55–63
27. WilliamsA(1994)Whatbotherscaregiversofstrokevictims?JNeurosciNurs26(3):155–161
28. Luna-MatosM,McGrathH,GaviriaM(2007)Neuropsychiatricmanifestationsincerebro-
vasculardiseases.RevChilNeuro-Psiquiatr45(2):129–140
29. RabinsPV,StarksteinSE,RobinsonRG(1991)Riskfactorsfordevelopingatypical(schizo-
phreniform)psychosisfollowingstroke.JNeuropsychiatryClinNeurosci3:6–9
30. Levin DN, Finklestein S (1982) Delayed psychosis after right temporoparietal stroke or
trauma:relationtoepilepsy.Neurology32:267–273
31. NishioY,IshiiK,KazuiH(2007)Frontal-lobesyndromeandpsychosisafterdamagetothe
brainstemdopaminergicnuclei.JNeurolSci260:271–274
32. EversonSA,RobertsonRE,GoldbergDE(1998)Depressivesymptomsandincreasedriskof
strokemortalityovera29-yearperiod.ArchInternMed158:1133–1138
33. HadidiN,Treat-JacobsonDJ,LindquistR(2009)Poststrokedepressionandfunctionalout-
come:acriticalreviewoflitreature.IssuesNeurol38(2):151–162
34. NarushimaK,KosierJT,RobinsonRG(2003)AreappraisalofPSD,intra-andinter-hemi-
sphericlesionlocationusingmeta-analysis.JNeuropsychiatryClinNeurosci15:422–430
Systemic Inflammation and Cognition
in the Elderly
Introduction
Background
IL-6 gene is associated with lower risk of developing AD [15], and IL-6 mRNA
levels may also predict the clinical progression of the disease [16]. Of interest is the
demonstration of interactive effects with other alleles known to confer risk for AD.
Forexample,McCuskeretal.[17] demonstrated that, in those carrying the apoli-
poprotein E4 (ApoE 4) allele, TNF-a substantially increased the risk of AD. Nicoll
et al. [18] demonstrated that simultaneous inheritance of the high-risk alleles for
IL-1a (IL-1a-889) and IL-b (IL-b + 3953) increased the odds of developing AD by
10.8 [18]. Taken together, available data suggest that AD risk may be influenced by
inflammatory factors in relationship to both serum measures and “genetic suscep-
tibility profile.” The data not only underscore the importance of inflammation in
AD but raise the possibility that inflammatory processes may be relevant to cognitive
ageing in nonclinical populations. The relevance of inflammation in this context is
best examined in community-dwelling ageing populations, which are the main
focus in this chapter.
“Inflammaging”
The term “inflammaging” has been used to describe the progressive increase in
systemic inflammation with advancing age and is characterized by a decline in
adaptive immune mechanisms, upregulation of the innate immune system, and a
resultant low-grade, chronic inflammatory response. Central to this process is the
overactivation of mononuclear phagocytes, which results in elevation of cytokine
levels [19, 20]; this comes at a time when the brain may be more susceptible to
adverse effects of inflammation. For example, animal studies demonstrate an increased
susceptibility of the aged brain to oxidative stress and inflammation [21], affording
a greater potential impact of inflammation on cognitive function. The brain has
been traditionally viewed as an immune-privileged site. However, recent research
indicates the peripheral immune system and the central nervous system (CNS)
communicate extensively [22]. Such communication raises a number of possibili-
ties regarding the relationship between cognitive function and inflammation. For
example, peripherally generated inflammation may be a response or contributor to
CNS inflammation and pathology [7].
Whether “inflammaging” or increased susceptibility to its effects translates into
meaningful change in cognitive function or confers greater risk of cognitive decline
isuncertainandisthesubjectofthisreview.Toexplorethisissue,aMEDLINE
searchwasconductedusingtheMESHterm“inflammation”orkeyword“inflam-
matorymarkers”andMESHterms“cognition”or“cognitiondisorders.”Thesearch
strategy identified those articles published in the English language until July 2009.
Articles were examined in detail only if the participants were drawn from non-
clinical populations, and only if the study included participants ³65 years of age.
Reference lists were cross-checked for additional articles not initially identified by the
search strategy. The cross-sectional and longitudinal data from studies fulfilling
the selection criteria are summarized in Tables 1 and 2, respectively.
Table 1 Cross-sectional studies of circulating inflammatory markers and cognitive function in community-dwelling elderly populations
180
Inflammatory
marker/s Study population Cognitive tests Outcomes Covariates Results
Alley IL-6 851 Community- Similarities subset of Individual Age, sex, race, Before covariate
et al.[23] CRP dwelling elders, Wechsler Adult cognitive education, inclusion, raised
70–79 years, Intelligence Scale- tests/ income,DM, IL-6 and CRP were
USA Revised, copying domains MI,stroke,hip inversely associated
geometric figures, and #, BP, HbA1c, with abstraction and
delayed recognition global HDL, waist language ability
Span test, modified cognitive circumference, (P < 0.05) and
Boston Naming test, function NSAIDs, global cognition
incidental recall alcohol, (P < 0.001); IL-6
on naming items, smoking, alone associated
abbreviated short physical with poor spatial
portable mental activity ability (P < 0.01)
status questionnaire and CRP with short
portable mental
status questionnaire
(P < 0.01);
all results
nonsignificant
after inclusion of
covariates
Baune IL-1b 369 Community- Three word recall test Individual Age, gender, Increased IL-8 inversely
et al.[24] sIL-4R dwelling (Tulving and Colotla cognitive education, associated with
IL-6 participants, lag measures), word tests/ smoking, memory (P < 0.01),
IL-8 >65 years, mean fluency, Stroop color- domains depressive cognitive speed
IL-10 age 72.6 years, word test, Wechsler symptoms, TIA, (P < 0.001) and motor
IL-12 Germany Adult Intelligence stroke,DM function (P < 0.01)
TNF-a scales digit symbol
test, Purdue Pegboard
test,MMSE
J. Trollor and E. Agars
Sweat CRP 125 Community- California Verbal Learning Individual Age,education,DM, Significant inverse
et al. [25] dwelling test,WechslerMemory cognitive statins association between
42–82-year-olds, Scale-Revised, Digit tests/ CRP and cognition
categorized by Span Backwards, domains only in overweight
BMI,USA VisualMemorySpan and obese women:
Backwards, Colorado executive function
Assessment tests: and figural memory
Tower of London, (P < 0.01), estimated
Shipley Institute of IQ and general
Living Scale cognitive functioning
(P < 0.05)
Schram
et al.[26]
1. Rotterdam IL-6 3,874 Community- MMSE,abbreviated Individual Age, sex, education CRP and IL-6 inversely
Study CRP dwelling Stroop test part cognitive associated with
a1- elderly, mean 3, Letter Digit tests/ global cognition and
Antichymo- age, 72 years, Substitution Task, domains executive functioning
trypsin Netherlands word fluency, Geriatric (P < 0.01), IL-6
MentalStatusschedule was also inversely
Systemic Inflammation and Cognition in the Elderly
associated with
MMSEscore
(P < 0.05)
2. Leiden IL-6 491 Community- MMSE,abbreviated Individual Age, sex, education No significant association
85-plus CRP dwelling 85-year- Stroop test part cognitive
Study olds, Netherlands 3, Letter Digit tests/
Substitution Task, domains
GeriatricMentalStatus
schedule, 12-Picture
Learning test
Fischer CRP 606 Community- MMSE Global Sex, BP, total No significant association
et al.[27] Homocysteine dwelling 75-year- cognitive cholesterol,
Fibrinogen olds, Austria function LDL, HDL, TG,
antihypertensive
medications,
181
statins, smoking
(continued)
Table 1 (continued)
182
Inflammatory
marker/s Study population Cognitive tests Outcomes Covariates Results
Dik IL-6 1,284 Community MMSE,AuditoryVerbal Individual Age, sex, education Serum a1-
et al. [28] CRP dwelling Learning task, Raven’s cognitive antichymotrypsin
a1- 62–85-year-olds, colored progressive tests/ inversely associated
Antichymo- Netherlands matrices, Alphabet domains with delayed recall
trypsin coding task-15 (P < 0.05) and albumin
Albumin inversely associated
withMMSEscore
(P = 0.05)
Ravaglia CRP 540 Cognitively MMSE Global Age, sex, education, Raised CRP inversely
et al. [29] intact elders, cognitive fibrinogen, associated with
mean age, function leukocyte count, MMSEscore
73 years, Italy albumin,BMI,
DM,IHD,PVD,
cerebrovascular
disease, chronic
pulmonary
disease, peptic
ulcer, edentulism
Elwan IL-6 94 Neurologically Paced Auditory Serial Individual Age, sex, education Raised IL-6 inversely
et al. [30] ICAM-1 intact healthy Addition test, cognitive associated with
ApoE genot- Egyptians, age Intentional and tests/ attention and
yping range, 40–82 Incidental memory domains intentional (sensory)
years, mean, test, Digit Symbol memory (P < 0.05);
58.5 years Substitution test, Trail ApoE 4 genotype
MakingtestAandB, associated with
Eysenck Personality significantly poorer
questionnaire intentional memory
(P < 0.05)
J. Trollor and E. Agars
Teunissen IL-6 65 Community- MAASprotocol:Auditory Individual Age, sex, education Serum haptoglobin
et al. [31] IL-6 receptor dwelling subjects, Verbal Learning task, cognitive and CRP inversely
CC16 mean age, Letter Digit coding test, tests/ correlated with Stroop
CRP 54 years baseline, Stroop color-word test domains test and delayed recall
Haptoglobin Netherlands (P < 0.05)
Weaver IL-6 779 Community- Boston naming test, Individual Age, sex, income, No significant
et al. [32] dwelling delayed verbal cognitive education, associations
70–79-year-olds, memory test, delayed tests/ ethnicity, alcohol,
USA recognition Span test, domains physical activity,
similarities subset of BMI,HbA1c,
the Weschler Adult DM,cancer
intelligence Scale-
revised, copying of
geometric figures
Bruunsgaard IL-6 126 Centenarians, 45 MMSE,Lawton’s Dementia Age, cancer, acute TNF-a levels correlated
et al. [33] TNF-a 81-year-olds, 23 instrumental ADL severity and chronic with dementia severity
CRP 55–65-year-olds, scale, caregiver’s inflammatory (P < 0.05)
Systemic Inflammation and Cognition in the Elderly
Haan 5 years CRP 1,118Mexican Verbal episodic memory All cause Age,sex,DM, CRP lower in ApoE 4
et al. [34] APOE Americans, and modified mini- dementia, stroke, carriers; in ApoE 4
genotype without mental state exam AD, CIND medications, carriers higher CRP
dementia/ (3MSE),DSM–IVR total cholesterol, associated with lower
CIND criteria, NINCDS- LDL all cause dementia
and aged ADRDA criteria and AD but after
60–101 years adjustment for
at baseline, metabolic disease/
USA stroke only associated
with AD; in non-
ApoE 4 carriers there
was no effect of CRP
on dementia/CIND
J. Trollor and E. Agars
Inflammatory Cognitive tests/
Duration marker/s Study population diagnostic tools Outcomes Covariates Results
Jordanova 3 years IL-6 216 African- MMSE(orientation), Individual Age, sex, school Raised IL-6 associated
et al. [35] CRP Caribbean CERAD tests cognitive leaving age, with decline in
Serum amy- participants, (immediate/delayed tests/ HT, stroke, orientation and
loid A 55–75 years word list recall, domains DM,smoking, immediate verbal
at baseline, delayed word list alcohol, anti- recall (P < 0.05); no
England recognition), Trail inflammatory association found
MakingtestA medications/ for CRP or serum
aspirin,BMI, amyloid A
disability
Komulainen 12 years CRP 97 Women MMSE,WordRecall Individual Age, education, Baseline CRP was
et al. [36] 50–60 years test, prospective cognitive depression, inversely related to
at baseline, memory test, Stroop tests/ hormone memory function at
Finland test, Letter digit domains replacement follow-up (P < 0.05);
Substitution test therapy, LDL, no association with
BMI cognitive speed or
MMSE
Rafnsson 4 years IL-6CRP 452 Community Weschler Logical Individual Age, sex, depressed Fibrinogen predicted
Systemic Inflammation and Cognition in the Elderly
et al. [37] ICAM-I dwelling Memorytest,Raven’s cognitive mood, peak prior decline in non-verbal
VCAM-1 subjects, aged Standard Progressive tests/ cognitive ability, reasoning and logical
Fibrinogen 55–74 years, matrices, Verbal domains smoking, alcohol memory (P < 0.05),
Scotland fluency test, Digit consumption, IL-6 was inversely
Symbol-Coding test cardiovascular related to information
disease, glucose processing speed and
intolerance/DM ICAM-Iassociated
with decline in general
cognitive ability and
non-verbal reasoning
(P < 0.05)
(continued)
185
Table 2 (continued)
186
Table 2 (continued)
Inflammatory Cognitive tests/
Duration marker/s Study population diagnostic tools Outcomes Covariates Results
Dik 3 years IL-6 1,284 Community MMSE,AuditoryVerbal Individual Age, sex, education Serum a1-antichymo-
et al. [28] CRP dwelling Learning task, cognitive trypsin associated
a1- 62–85 year Raven’s coloured tests/ with decline in
Antichym- olds, progressive matrices, domains MMSE,OR=1.60,
otrypsin Amsterdam alphabet coding 95% CI (1.05–2.43);
Albumin task-15 CRP, IL-6 and
albumin not related
to cognitive decline
Tilvis 10 years CRP 650 Community MMSE,CDR Dementia Age, sex, baseline Elevated CRP associated
et al. [42] dwelling 75, severity MMSEscore with drop in CDR
80 and 85 year class at 5 years
olds, Finland (RR = 2.32, 95% CI
(1.01–5.46) but not at
10 years
Yaffe 4 years IL-6 2,632 Community Modified Global Age, race, sex, Only in participants
et al. [43] CRP dwelling black mini-mental cognitive education, with metabolic
TNF-a and white state examination function alcohol, stroke, syndrome were
elders, mean (3MSE) depressive raised inflammatory
age 74 years, symptoms, markers significantly
USA statins, baseline associated with
cognitive score cognitive decline
(RR = 1.66, 95% CI
(1.19–2.32) those
with metabolic
syndrome and
low inflammation
(RR = 1.08, 95% CI
J. Trollor and E. Agars
(0.89–1.30)
Inflammatory Cognitive tests/
Duration marker/s Study population diagnostic tools Outcomes Covariates Results
and those without
metabolic
syndrome and
high inflammation
(RR = 0.81, 95% CI
(0.60–1.08) were not
at elevated risk of
decline
Engelhart 4–9 years IL-6 727 (Random MMSE,Geriatric All cause Age, sex, Elevated a1-
et al. [44] CRP subcohort of MentalState dementia, education level, antichymotrypsin
a1- 6,713) and Schedule, AD, VaD smoking, anti- and IL-6 associated
Antichym- 188 cases Cambridge inflammatory with increased risk
otrypsin classified examination for medications, dementia, AD,
ICAM-1 dementia at mental disorders statins,BMI, VaD; less strong
VCAM-1 follow-up in elderly persons, SBP,DM,Ankle- association present
Community DSM-III-R, brachial index, CRP, no association
dwelling NINCDS-ADRDA intima media ICAMorVCAM
Systemic Inflammation and Cognition in the Elderly
ADRDA (AD)
(continued)
Table 2 (continued)
190
Cross-Sectional Studies
Eleven studies report the relationship between various inflammatory markers and
cognitive function (see Table 1) from cross-sectional analyses. The studies vary in
size, ranging from a small Egyptian study (n = 94) [30] to a large study of community-
dwelling elders (n = 3,874) [26]. Studies involve a diverse range of ethnic groups [24,
29, 30, 35]. Some studies include participants from mid to late life [25, 30].
The most consistently reported finding is a cross-sectional association between ele-
vated levels of IL-6 and C-reactive protein (CRP) and poorer cognitive performance
[23, 26, 32, 36].TheMacArthurStudyofSuccessfulAging(n = 851) assessed IL-6 and
CRP in 70–79-year-olds [23]. Participants were selected on the basis of minimal cogni-
tive disability and intact functional status. Global cognitive function was derived from
a composite of five neuropsychological tests. Cross-sectional analysis revealed that both
CRP and IL-6 levels were inversely related to global cognitive function (P < 0.001) as
well as individual tests of abstraction and language (P < 0.05). IL-6 levels were inversely
related to spatial ability, and CRP was inversely related to the score on the short portable
mental status questionnaire. However, after inclusion of covariates in the analysis, the
results of the analyses were no longer significant for IL-6 or CRP.
Schram et al. [26] investigated inflammatory markers and cognitive function in
two large community-derived cohorts with conflicting results. Both were conducted
in the Netherlands: the Rotterdam Study investigated 3,874 individuals aged
55 years and over (age range, 55–99 years; mean, 72) and the Leiden 85-plus Study
investigated 491 individuals aged 85 years at baseline. Circulating levels of IL-6 and
CRP were measured in both studies, with the addition of a1-antichymotrypsin in the
Rotterdam Study. Neuropsychological testing measured mini-mental state examina-
tionscore(MMSE),individualmeasuresofexecutivefunctionandmemory,anda
global score derived from summation of individual neuropsychological test scores.
In cross-sectional analysis, the Rotterdam Study revealed that IL-6 and CRP were
inversely related to global cognition and executive function (P < 0.01) and IL-6 was
inverselyassociatedwithMMSE(P < 0.05). The Leiden 85-plus Study demonstrated
no significant cross-sectional associations between inflammatory markers and cog-
nition. This variability in results highlights the impact of methodological issues in
the wider literature and may arise from differences in cohort size and age, cytokine
assays (e.g., high-sensitivity CRP measured in Rotterdam Study, whole blood rather
than plasma in Leiden 85-plus analysis), and inclusion of different cognitive tests.
The Longitudinal Aging Study Amsterdam reported by Dik et al. [28] was a
similarly large prospective population-based cohort study. This study measured a1-
antichymotrypsin, albumin, CRP, and IL-6 in participants aged 65–88 years
(n = 1,284; mean age, 75.4 years). Cognitive testing included global cognition and
individual neuropsychological domains such as information processing speed, fluid
intelligence, and memory (see Table 1). After adjustment, only a1-antichymotrypsin
andalbuminweresignificantlyassociatedwithdelayedrecallandMMSE,respec-
tively (P < 0.05). Similarly to observations by Schram et al. [26], Dik et al. [28]
found no effect of ApoE 4 on cross-sectional analysis. Dik et al. [28] categorized
192 J. Trollor and E. Agars
markers into tertiles and dichotomized IL-6 around the detection limit to compensate
for reduced assay sensitivity. However, disparities in cytokine measurement and
analysis may in part explain the lack of significant results in this study. A number
of other studies of community-dwelling elders have failed to establish a relation-
ship between cognitive function and CRP [27, 28, 33]. The negative findings by
Fischer et al. [27] may relate to a number of methodological problems including a
very high dropout rate (60%), and the study by Bruunsgaard et al. [33] used a lim-
ited range of cognitive tests and, as it contained a large sample of centenarians, was
biased toward a very old population.
Interaction between inflammatory factors, gender, and other risk variables has
been demonstrated in some studies. For example, in a study of 125 community-
dwelling subjects 42–82 years old, Sweat et al. [25] demonstrated an association
between raised CRP and cognitive impairment in women only. As the relationship
wasstrongerinoverweightandobesesubjects[bodymassindex(BMI)>25]and
was particularly notable for frontal-executive function, the authors suggested vas-
cular mediation of the relationship.
Few studies have evaluated the relationship between cognitive function and a
comprehensive array of inflammatory markers while controlling for possible cova-
riates. A notable exception is the study by Baune et al. [24], which found an iso-
lated association between IL-8 levels and poorer memory, processing speed, and
motor function in a cohort of 369 older Germans [24]. This same study failed to
find an association between cognitive function and IL-6 or TNF-a.
Of additional note is that our literature search also identified a small number of
studies that explored the relationship between proinflammatory cytokines and cog-
nition in midlife (not tabulated). A small initial study [31] suggested an association
between higher CRP levels and poorer performance on a test of episodic verbal
memory in middle-aged participants. Another study in 460 middle-aged partici-
pants demonstrated an association between higher IL-6 levels and poorer perfor-
mance on tests of attention, working memory, and executive function. More
recently, in a large study (n = 4,200) of predominantly male office staff aged
between 35 and 55 years, Gimeno et al. [46] demonstrated an association between
higher levels of CRP and poorer memory function and between higher IL-6 levels
and performance on a semantic fluency task.
Longitudinal Studies
example, those studies that failed to find a relationship were of similar overall
duration, included participants of similar ages, and included similar covariates in
analyses as those which found a significant relationship between IL-6 at baseline and
long-term cognitive outcome. Of note is that several studies failing to find an asso-
ciation between cognition and IL-6 levels tended to use a less detailed cognitive
batteryoremployedlesssensitivecognitivemeasuressuchasMMSE[26, 39].
Two studies evaluated IL-6 levels at baseline in nondemented community-dwelling
elders and specifically related this to dementia diagnosis at follow-up [38, 39]. The
Conselice Study of Brain Aging [38] investigated the role of inflammatory markers
on development of dementia in a community cohort of 804 elderly Italians cogni-
tively intact at baseline (mean age, 74 years). After 4 years, combination IL-6 and
CRP predicted all-cause dementia and vascular dementia (VaD) but not AD [hazard
ratio, 2.56; 95% confidence interval (CI), 1.21–5.20]. IL-6 alone did not predict
AD diagnosis. Tan et al. [39] followed 691 elders in the United States for a mean of
7 years but found no relationship between IL-6 and AD diagnosis. The small number
of studies in this area makes firm conclusions impossible. However, taken together
these results suggest that IL-6 levels at baseline are not strongly predictive of AD on
follow-up. There is some preliminary support for a longitudinal association between
IL-6 and VaD, which would be in keeping with a cross-sectional study by Zuliani
et al. [11] in a clinical cohort of 222 patients derived from outpatient clinics in which
higher IL-6 was associated with VaD but not AD (P < 0.05).
Fifteen studies have examined the relationship between raised CRP at baseline
and cognitive outcome at longitudinal follow-up in community-dwelling elders
who were nondemented at initial assessment. Studies are divided regarding the
presence of an association, with eight studies finding an association [23, 34, 36,
41–45] and seven studies failing to find an association [26, 28, 35, 37–40]. There
is no specific pattern of methodological differences between those studies that did
or did not find an association between CRP and cognitive outcome. Those studies
that have found an association between baseline CRP and change in cognition have
highlighted an association with decline in global measures of cognition [26, 43],
memory function [36], dementia diagnosis [34, 38, 45], or dementia severity [42].
A specific association with dementia subtype has not been observed, but available
data suggest that raised CRP at baseline is more strongly associated with VaD
rather than AD [38]. This issue is complicated by the association between CRP and
other known vascular risk factors, and can only be resolved by studies that compre-
hensively control for common vascular risks.
An interaction between CRP and some other risk variables for cognitive decline
has been observed. For example, Yaffe et al. [43] observed that inflammatory markers
only impacted on cognition in those with metabolic syndrome. An interaction
between CRP and ApoE 4 genotype in which CRP has greater impact on cognitive
decline in the presence of ApoE 4 genotype [26] has also been observed, but a
contrary finding has also been published [34].
A limited number of other inflammatory markers have been examined in com-
munity samples to determine any longitudinal impact on cognitive function. Serum
amyloid A (SAA) was measured in one study [35] but had no bearing on long-term
194 J. Trollor and E. Agars
Discussion
Conclusion
References
30. Elwan O, Madkour O, Elwan F et al (2003) Brain aging in normal Egyptians: cognition,
education, personality, genetic and immunological study. J Neurol Sci 211:15–22
31. TeunissenCE,vanBoxtelMPJ,BosmaHetal(2003)Inflammationmarkersinrelationto
cognition in a healthy aging population. J Neuroimmunol 134:142–150
32. Weaver JD, Huang MH, Albert M et al (2002) Interleukin-6 and risk of cognitive decline:
MacArthurstudiesofsuccessfulaging.Neurology59:371–378
33. Bruunsgaard H, Andersen-Ranberg K, Jeune B et al (1999) A high plasma concentration of
TNF-a is associated with dementia in centenarians. J Gerontol A Biol Sci Med Sci
54A(7):357–364
34. HaanMN,AielloAE,WestNAetal(2008)C-reactiveproteinandrateofdementiaincarriers
and non carriers of apolipoprotein APOE4 genotype. Neurobiol Aging 29:1774–1782
35. JordanovaV,StewartR,DaviesEetal(2007)Markersofinflammationandcognitivedecline
in an African-Caribbean population. Int J Geriatr Psychiatry 22:966–973
36. KomulainenP,LakkaTA,KivipeltoMetal(2007)SerumhighsensitivityC-reactiveprotein
and cognitive function in elderly women. Age Ageing 36:443–448
37. Rafnsson SB, Deary IJ, Smith FB et al (2007) Cognitive decline and markers of inflammation
and hemostasis: the Edinburgh artery study. J Am Geriatr Soc 55:700–707
38. RavagliaG,FortiP,MaioliFetal(2007)Bloodinflammatorymarkersandriskofdementia:
the Conselice study of brain aging. Neurobiol Aging 28:1810–1820
39. Tan ZS, Beiser AS, Vasan RS et al (2007) Inflammatory markers and the risk of Alzheimer’s
disease: the Framingham study. Neurology 68:1902–1908
40. VandenBiggelaarAHJ,GusseklooJ,deCraenAJMetal(2007)Inflammationandinterleukin-1
signaling network contribute to depressive symptoms but not cognitive decline in old age. Exp
Gerontol 42:693–701
41. WeuveJ,RidkerPM,CookNRetal(2006)High-sensitivityC-reactiveproteinandcognitive
function in older women. Epidemiology 17:183–189
42. TilvisRS,Kahonen-VareMH,JolkkonenJetal(2004)Predictorsofcognitivedeclineand
mortality of aged people over a 10-year period. J Gerontol A Biol Sci Med Sci
59A(3):268–274
43. Yaffe K, Kanaya A, Lindquist K et al (2004) The metabolic syndrome, inflammation, and risk
ofcognitivedecline.JAMA292(18):2237–2242
44. EngelhartM,GeerlingsMI,MeijerJetal(2004)Inflammatoryproteinsinplasmaandtherisk
of dementia. Arch Neurol 61:668–672
45. Schmidt R, Schmidt H, Curb D et al (2002) Early inflammation and dementia: a 25-year
follow-up of the Honolulu-Asia aging study. Ann Neurol 52:168–174
46. GimenoD,MarmotMG,Singhetal(2008)Inflammatorymarkersandcognitivefunctionin
middle-aged adults: the Whitehall II study. Psychoneuroendocrinology 33:1322–1334
Diagnosis and Clinical Relevance of Depression
and Apathy in Alzheimer’s Disease
Keywords Anxiety•Apathy•Dementia•Depression•Motivation
Diagnosis of Depression in AD
One of the major challenges in neuropsychiatry is how to obtain a valid and reliable
diagnosis of a psychiatric disorder when its symptoms overlap with the symptoms
of the neurological condition. Another limitation is that common disorders such as
depression in Alzheimer’s disease (AD) are rarely isolated phenomena and usually
coexist with other psychiatric conditions such as anxiety, apathy, pathological
affective display, psychotic symptoms, and poor insight. These confounding factors
may account for the lack of general consensus on the most valid and reliable
method to diagnose depression in AD.
Four different strategies have been used to diagnose depression in chronic
degenerative disorders. The “inclusive approach” [1] diagnoses depression based
on symptoms that may or may not be related to the physical illness. This approach
is the most commonly used diagnostic strategy in neuropsychiatry. The “exclusive
approach,” on the other hand, does not include for diagnosis those symptoms con-
sidered to be related to the physical illness [2]. The “substitutive approach” replaces
overlapping symptoms of depression with psychological symptoms [3]. Finally, the
“specific symptom approach” only considers for diagnosis those symptoms that
were identified as belonging to a “depressive cluster” using specific statistical tech-
niques, such as latent class analysis. In a recent study Verkaik and coworkers [4]
examined potential confounders for the diagnosis of depression in AD. One main
confounder is that some studies diagnosed depression on the basis of a cutoff score
on a depression scale (the “continuous method”) whereas other studies diagnosed
depression using standardized diagnostic criteria (the “categorical method”). Other
confounders identified by the authors are (1) the grouping of different types of
dementia, (2) different criteria to diagnose AD, (3) different instruments to assess
the severity of AD and depression, and (4) heterogeneous samples in terms of size
and sociodemographic characteristics.
Lyketsos and coworkers were the first to propose standardized criteria to diagnose
depression in AD [5]. They assessed a large sample of individuals living in the com-
munity using the Neuropsychiatric Inventory (NPI). A latent class analysis produced
three clusters, one of which was characterized by depression, anxiety, irritability, and
apathy. Based on this cluster, Lyketsos and coworkers proposed the diagnostic crite-
ria summarized in Table 1. One limitation of the study lies with the use of the NPI,
Depression and Apathy in Alzheimer’s Disease 203
Taken together, these findings suggest that symptoms of depression are not rife among
euthymic AD patients. It is important to note that AD patients may have poor insight
into their own depressive symptoms and tend to minimize or deny their presence.
Therefore, it is important to obtain collateral information when assessing patients’
mood.
In a more recent study we assessed the construct of major and minor depression in
a series of 670 patients with AD [8]. In this cross-sectional study, we found that 26%
had major depression and an identical percentage had minor depression. The percentage
of patients with three or more symptoms of depression (as required by the NIMH-dAD
diagnostic criteria) but without sad mood was 22% among patients with mild AD, 23%
among patients with moderate AD, and 41% among patients with severe AD,
suggesting that the NIMH-dAD may have high sensitivity but poor specificity.
Another strategy to clarify the phenomenology of depression in AD is to assess the
pattern of symptom improvement on the remission of depression. To this end, we exam-
ined a series of 65 patients with AD and depression who received a follow-up assess-
ment 1–3 years later [9]. At follow-up, 33 patients had a full remission of depression
whereas 32 patients continued to be depressed (with major or minor depression).
The main finding was that patients with a full remission of depression had a significantly
lower score on the Hamilton depression scale for the symptoms of sad mood, guilt,
Depression and Apathy in Alzheimer’s Disease 205
suicide ideation, insomnia, loss of interest, psychomotor changes, loss of energy, social
withdrawal, and loss of appetite/weight as compared to patients with no remission of
depression. Moreover, patients on remission also showed a significant decrease in the
severity of anxiety. On the other hand, there were no significant between-group differ-
ences in the severity of apathy, supporting previous findings that showed that apathy and
depression are overlapping but independent syndromes in AD [10].
Frequency of Depression in AD
Before discussing the frequency of depression in AD, it is important to note that esti-
mates of depression in AD depend on sampling issues, diagnostic methods, and clinical
manifestations. The prevalence of major and minor depression has been estimated to
range between 30% and 50% [3]. Population studies reported a prevalence of dysphoria
of 20% and an 18-month incidence of 18% [11, 12]. A population case-report study
from the United Kingdom diagnosed major depression in 24% of their sample [13],
and similar frequencies were reported in a recent study from Maastricht [14].
Depression in AD has been associated with worse quality of life, greater impair-
ments in activities of daily living, decrease in caregiver’s well-being [15], faster
cognitive decline, greater health care utilization [16], higher mortality rates [17],
and higher rates of nursing home placement [18]. Our group [8] found that patients
meeting DSM-IV criteria for either minor or major depression had more severe
social dysfunction and greater impairment in activities of daily living than AD
patients without depression. Furthermore, AD patients with major depression had
more severe anxiety, apathy, delusions, and parkinsonism than patients with minor
depression, suggesting that the severity of psychopathological and neurological
impairments in AD increases with increasing severity of depression. On the other
hand, patients with either major or minor depression had similar deficits on activi-
ties of daily living and social functioning, suggesting that even mild levels of
depression are significantly associated with increased functional impairment in
AD. Depression among nursing home patients with AD has been associated with
relatively more severe malnutrition, behavioral problems, noncompliance with treat-
ment, increased nursing staff time, and excessive mortality rate [19].
Conclusion
Depression is among the most frequent psychiatric disorders in AD. One important
limitation to the diagnosis of depression in AD is the lack of valid and reliable criteria.
206 S.E. Starkstein and S. Brockman
Apathy in AD
Diagnosis of Apathy in AD
Although the ICD-10 makes no reference to apathy, the DSM-IV mentions apa-
thy as a subtype of personality disorder caused by a General Medical Condition.
Our group has validated a set of standardized criteria for the diagnosis of apathy
in AD [23]. To this aim, we assessed a series of 319 patients with AD using the
apathy scale (a severity rating scale) and found that 13% of the sample met our
ad hoc criteria for apathy. These criteria have been recently updated by Starkstein
and Leentjens [24] (Table 3). On the other hand, none of a series of 46 age-
comparable healthy controls demonstrated apathy. It is important to note the
overlap between depression and apathy in AD: 13% of the AD sample had apa-
thy and no depression, but 24% had both depression and apathy. AD patients
with apathy only and patients with neither apathy nor depression had similar
scores on the Hamilton depression scale, suggesting that apathy does not artifi-
cially increase depression scores in this population.
The European Psychiatric Association Task Force on apathy has recently
published standardized diagnostic criteria for apathy in AD [25]. These criteria
Depression and Apathy in Alzheimer’s Disease 207
be made using the Starkstein and Leentjens’ or the European diagnostic criteria for
apathy.
Frequency of Apathy in AD
In a recent study, Starkstein and coworkers [35] examined the frequency of apathy
in a series of 319 patients with AD, 117 patients with depression but no dementia,
and 36 age-comparable healthy individuals. Apathy was diagnosed in 37% of the
AD patients, in 32% of the depressed patients, and in none of the healthy controls.
A Latin American study [36] examined 60 AD patients with the NPI, reporting a
frequency of apathy of 53%. A similar frequency (59%) was recently reported by
an American study [37].
In a recent series of longitudinal studies, our group examined the predictive validity
of apathy in AD. The first study included a series of patients who were followed for
1–4 years [38]. At baseline, apathy was significantly associated with older age and
depression (both major and minor). The frequency of apathy increased from 14%
in the stage of very mild AD to 61% in the stage of severe AD. Therefore, cognitive
deficits are not sufficient to produce apathy in AD because about half the patients
with moderate or severe dementia did not show apathy. Whether cognitive deficits
Depression and Apathy in Alzheimer’s Disease 209
are necessary to produce apathy has not been yet determined, but most studies
assessing patients with a variety of neurological conditions such as stroke and
Parkinson’s disease found a significant association between greater apathy and
more severe cognitive impairments [39, 40]. At follow-up, patients with apathy at
baseline or patients who developed apathy during follow-up had a significant
increase in Hamilton depression scale scores compared to AD patients with no
apathy at baseline. Moreover, patients with apathy at baseline or those who devel-
oped apathy during follow-up had a significantly greater functional and cognitive
decline and more severe parkinsonism than patients without apathy at baseline [41].
Based on these findings, we proposed that apathy may be a behavioral marker of a
more “malignant” type of AD, with more severe behavioral problems and a faster
cognitive, functional, and motor decline.
Conclusions
Acknowledgments This study was partially supported with grants from the University of
Western Australia, and the National Health and Medical Research Council.
References
1. Cohen-Cole SA, Stoudemire A (1987) Major depression and physical illness. Psychiatr Clin
North Am 10:1–17
2. Gallo JJ, Rabins PV, Anthony JC (1999) Sadness in older persons: 13-year follow-up of a
community sample in Baltimore, Maryland. Psychol Med 29(2):341–350
3. Olin JT, Katz IR, Meyers BS et al (2002) Provisional diagnostic criteria for depression of
Alzheimer disease: rationale and background. Am J Geriatr Psychiatry 10(2):129–141
4. Verkaik R, Nuyen J, Schellevis F et al (2007) The relationship between severity of Alzheimer’s
disease and prevalence of comorbid depressive symptoms and depression: a systematic
review. Int J Geriatr Psychiatry 22(11):1063–1086
5. Lyketsos CG, Sheppard JM, Steinberg M et al (2001) Neuropsychiatric disturbance in
Alzheimer’s disease clusters into three groups: the Cache County study. Int J Geriatr
Psychiatry 16(11):1043–1053 (see comment)
6. Olin JT, Schneider LS, Katz IR et al (2002) Provisional diagnostic criteria for depression of
Alzheimer disease. Am J Geriatr Psychiatry 10(2):125–128
210 S.E. Starkstein and S. Brockman
Keywords Alzheimerdisease•Earlydiagnosis•Geneticbiomarkers•Treatment
symptoms that significantly affect the life quality of patients and cohabitants usually
appear in the intermediate and late stages of the disease.
Changes in the biological rhythms involve quantitative and qualitative alterations
of sleep, increase of evening symptoms when the sun sets (sundowning), and
flattening of biological rhythms, including plane body rhythm with a decline of the
patient’s daytime mobility and increased activity at night, thus affecting their
environment.
The diagnosis of AD is mainly clinical and, to date, we lack a biological marker
or diagnostic test that could positively and selectively identify the disease, beyond
the final anatomopathological assessment.
Accordingly, the clinical assessment remains the most important tool in the
diagnostic process, but neuropsychology and some complementary methods of
diagnosis also collaborate [1].
The distinction between the cognitive and noncognitive aspects (also called
primary and secondary) will be used with an organizational and didactic criterion
because the coexistence and overlapping of these phenomena is normal in the clinical
field and is beyond this dichotomy.
Current diagnostic criteria allow for high sensitivity and diagnostic efficiency in
close to 85% of cases. Standardizations propose a consensus of diagnostic standards
to be used in our environment, thus minimizing disputes arising from the use of the
different proposals.
The most commonly used criteria for disease diagnosis are based on the
Diagnostic and Statistical Manual, Fourth Revision (DSM IV TR), or the
National Institute of Neurologic, Communicative Disorders and Stroke/
Alzheimer’s Disease and Related Disorders Association (NINCDS–ADRDA).
The American Academy of Neurology has evaluated these criteria and considered
them reliable. However, there are several points of evidence indicating the
need to update the current criteria for definition, such as inadequate diagnostic
specificity: the criteria of DSM IV TR and NINCDS–ADRDA have been vali-
dated using neuropathological gold standards, showing a diagnostic accuracy
that ranges between 65% and 96%. When the diagnosis was for other demen-
tias and not AD, it was only between 23% and 88%.
The patient’s medical history is a key element in the diagnosis of dementia
syndromes. Input from informants (not just the story of the patient) is also
important. A number of tools are available that can be used to obtain information
about the everyday activities and performance of these patients, although none
has been able to demonstrate superior efficacy over another; therefore, the doctor’s
careful inquiry about this item is crucial.
The differential diagnosis of AD is one of the most complex challenges of
medicine: not only because it is a disease that compromises the person’s thought
and behavior, dissocializing them and generating a substantial change in their
integrity, but also because it is based on a complex set of diagnostic premises
through which we reach a diagnosis that can only be confirmed by analyzing the
patient’s brain neuropathology, something that is generally not done.
216 L.I. Brusco
in all AD patients, they are often the main reason why a patient seeks medical attention.
It is therefore obvious that a correct treatment of the behavioral changes of an AD
patient not only generates an improvement of the behavioral performance of the
subject but also an increase of their intellectual possibilities [5]. Also, these
disorders limit the patient at a social level and may be the most frequent cause for
an early hospitalization, which undoubtedly changes the future situation and
clearly increases the costs generated by this disease.
Treatment of AD
The other drug used in combination with the aforementioned ones is memantine,
a partial antagonist of glutamate used in combination therapy with cholinergic
therapy [8].
ThereisevidenceindicatingthatAEisassociatedwithanimportantlossofsyn-
apses, dendritic changes, and cell death. The severity of the dementia is clearly
relatedtotheextensionofthesynapticloss.Eventhoughtheexactcauseandthe
underlying mechanisms of neuronal loss are not clear, there are several possible
factors, such as endogenous and exogenous neurotoxins, metabolic alterations,
alterations in calcium homeostasis, abnormal processing of the amyloid precursor
protein, and changes in the proteins of neurons.
To delay the progression of the degenerative process, it is essential to develop
treatments that maintain synaptic stability and ensure the survival of neurons. Some
treatments that use neuronal growth factors and genetic therapy techniques could
be fruitful in the future [9].
The primary symptoms are defined as those that involve memory loss (both fixation
and evocation memory) and other cognitive disorders (such as aphasia, apraxia,
agnosia, and further impairment of the executive function).
Cognitive Decline and Treatment of Alzheimer’s Disease 219
Cholinergic Agents
Multiple data prove the significant role that cholinergic mechanisms play in AD:
1. Anticholinergic agents with core action produce cognitive deficit in humans
2. Cholinergic neurotransmission shapes memory and learning
3. The lesions that affect central cholinergic pathways cause learning and memory
impairment, which may be reversed through the administration of cholinomimetics
4. PostmortemstudiesofADpatientsshowlossofcholinergicneuronsinthesep-
tum and basal nucleus of Meynert, reduction of the cholines acetyltransferase
and acetylcholinesterase, and correlation between these changes and the level of
cognitive impairment
Relative to these points, it was theorized that increasing central cholinergic trans-
mission would be useful in the treatment of AD. This effect may be achieved by
using acetylcholine precursors, by inhibiting cholinesterase, the enzyme that
reduces it, or by using direct postsynaptic agonists.
Acetylcholine Precursors
Acetylcholinesterase Inhibitors
EventhoughacetylcholinesteraseinhibitorsarenotusefulforallADpatients,there
is a group of subjects who might benefit from treatment with acetylcholinesterase
inhibitors; these are patients with late onset of the symptoms, initial stages of the
disease, and lack of a related organic disease.
Donepezil is a benzyl methyl piperidine, mixed inhibitor (competitive-
noncompetitive) of the enzyme, which shows a therapeutic profile similar to that of the
prototype drug, tacrine, although it is much more powerful [10]. It presents a pro-
longed half-life, more than 80 h, which facilitates its administration in just one daily
intake, 5 or 10 mg/day being the indicated dose [11]. In contrast to tacrine, it does not
show hepatotoxicity or high incidence of digestive intolerance. The most common
adverse effects are nausea, vomiting, anorexia, insomnia, cramps, and bradycardia.
Rivastigmine is an acetylcholinesterase inhibitor also approved by the FDA.
It is a pseudo-irreversible inhibitor that dissociates the enzyme slowly. It is
220 L.I. Brusco
Glutamate Regulators
Memantine is a new therapy that has recently been approved by the FDA for the
treatment of moderate to severe AD [12]. It belongs to the family of partial antago-
nists of the NMDA receptor and can added to the treatment with inhibitors [13]. Its
use has been approved both alone and in combination with the mentioned inhibitors.
It could also have an effect on the basic physiopathogenic process of the disease
through the inhibition of the entrance of calcium at rest, which would exist as a
pathological process in neurodegenerative disease. The adverse reactions are milder
than those with anticholinesterase drugs: anxiety, akathisia, and mild gastrointestinal
pain. There is a new presentation and administration of memantine, based on work
that guarantees the use in a single intake of 20 mg, with the same effect and
tolerance.
It is the only drug approved for the severe form of the disease: pharmacoeco-
nomic analyses have been conducted in most cases by comparing the drug to non-
pharmacological treatment [14].
In general, the different trials have shown that memantine is less costly and more
effective than nonpharmacological treatment, although, given the limited number of
pharmacoeconomic studies performed, it is still too early to draw conclusions on
the drug’s cost-effectiveness.
Antioxidants
Within this heterogeneous group we must mention vitamin E, selegiline, and
bifemelane.
Cognitive Decline and Treatment of Alzheimer’s Disease 221
Antiinflammatory Drugs
Similar to the use of the drugs already described, there has been epidemiological
proof that postmenopausal women who have received hormone replacement therapy
with estrogens would be more protected against AD. It is likely that estrogens may
have a neuroprotective effect in delaying the onset of the disease, but the data on
the use of estrogens have not been positive [16].
An open, randomized, double-blind, placebo-controlled trial on mild to moder-
ate AD failed to prove the benefits of hormone replacement after a term of
12 months. There were no changes in the primary measures of efficacy in this trial,
and concern was raised regarding deep vein thrombosis as a possible adverse effect.
An additional smaller trial of 16 weeks also failed to show benefits with this ther-
apy. Consequently, there are no data supporting the recommendation of the use.
Cognitive Decline and Treatment of Alzheimer’s Disease 223
Neurotrophic Factors
Although the primary symptoms of AD are memory impairment and loss of other
cognitive abilities, patients also develop secondary symptoms, among which are
depression, anxiety, agitation, delirium, hallucinations, and insomnia. It is recom-
mendable to manage with caution the use of antipsychotics and anxiolytics, keeping
in mind that both have side effects [22]. The first behavior is to wait for the
therapeutic effects of memantine and anticholinesterase medication as, besides
cognition, both drugs also improve the neuropsychiatric inventory [23].
With antipsychotics it is necessary to avoid the typical ones that cause parkin-
sonism, given the predisposition of elderly dementia patients to experience
extrapyramidal symptoms. However, the atypical drugs have proved an increase in
mortality in late-life psychosis. Quetiapine (with the family’s informed consent)
may be the drug to indicate in the psychotic disorders of dementia, given the low
production of extrapyramidal effects in low doses.
224 L.I. Brusco
It is important to consider not only the patient but also his or her caretaker, who in
general is a relative and is under chronic stress. Therefore, aid measures through
support groups for relatives and caretakers such as those at the Asociación
Alzheimer Argentina, that advise, orient, and support relatives and caretakers, are
particularly important.
Assistance to the patient involves not only medication, but also the work of
psychologists, occupational therapists, voice specialists, and physical therapists in
a treatment modality in which multiple stimulation is essential.
It is important to remember that this disease affects the entire family and that
interdisciplinary work is essential for the correct approach to the patient and his
or her environment.
References
1. American Academy of Neurology (2009) AAN guideline summary for clinicians: detection,
diagnosis and management of dementia. http://www.aan.com/professionals/practice/pdfs/
dementia_guideline.pdf. Accessed 15 April 2009
Cognitive Decline and Treatment of Alzheimer’s Disease 225
2. Small GW, Siddarth P, Burggren AC et al (2009) Influence of cognitive status, age, and
APOE-4 genetic risk on brain FDDNP positron-emission tomography imaging in persons
withoutdementia.ArchGenPsychiatry66(1):81–87
3. ReisbergB,AuerSR(1996)BehavioralpathologyinAlzheimer’sdisease.Psychogeriatrics
8:301–308
4. WraggRE,JesteDV(1989)OverviewofdepressionandpsychosisinAlzheimer’sdisease.
AmJPsychiatry146:577–587
5. Salzman C (1997) Treatment of the elderly agitated patient. J Clin Psychiatry 48(5
suppl):19–22
6. Schatzberg A, Nemeroff C (1998) Textbook of psychopharmacology, 2nd edn. American
Psychiatric,Washington,DC
7. American Psychiatric Association (2009) Practice guideline and resources for treatment of
patients with Alzheimer´s disease and other dementias, 2nd edn. http://www.psychiatryonline.
com/pracGuide/pracGuideTopic_3.aspx. Accessed 15 April 2009
8. APAWorkGrouponAlzheimer´sDiseaseandotherDementias,RabinsPN,BlackerDetal
(2007)AmericanPsychiatricAssociationpracticeguidelineforthetreatmentofpatientswith
Alzheimer´s disease and other dementias. Second edition. Am J Psychiatry 164(12
suppl):5–56
9. Moizeszowicz J (1998) Psicofarmacología psicodinámica IV: estrategias terapéuticas y psi-
coneurobiológicas,4thedn.Paidós,BuenosAires
10. RaskindMA,SadowskyCHetal(1997)Effectoftacrineonlanguage,praxisandnoncogni-
tive behavioral problems in Alzheimer’s disease. Arch Neurol 54:836–840
11. HowardRJ,JuszczakE,BallardCGetal(2007)Donepezilforthetreatmentofagitationin
Alzheimer´sdisease.NEnglJMed357(14):1382–1392
12. Tariot PN, Farlow MR, Grossberg GT et al (2004) Memantine treatment in patients with
moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled
trial.JAMA291:317–324
13. Farlow M et al (2003) Memantine/donepezil dual therapy is superior to placebo/donepezil
therapy for treatment of moderate to severe Alzheimer´s disease. Neurology 60(suppl
1):A412
14. Reisberg B, Doody R, Söffler A et al (2003) Memantine in moderate to severe Alzheimer’s
disease.NEnglJMed348:1333–1341
15. Tolbert SR, Fuller MA (1996) Seleginine in treatment of behavioral and cognitive symptoms
ofAlzheimerdisease.AnnPharmacother30:1122–1129
16. Kawas C, Resnik S, Morrison A (1997) A prospective study of estrogen replacement therapy
and the risk of developing Alzheimer disease: the Baltimore longitudinal study of aging.
Neurology 48:1517–1521
17. RütherE,RitterR,ApececheaM,FreytagS,GmeinbauerR,WindischM(2000)Sustained
improvements in patients with dementia of Alzheimer’s type (DAT) 6 months after termina-
tionofcerebrolysintherapy.JNeuralTransm107:815–829
18. RuetherE,AlvarezXA,RainerM,MoesslesH(2002)Sustainedimprovementofcognition
and global function in patients with moderately severe Alzheimer’s disease: a double-blind,
placebo-controlledstudywiththeneurotrophicagentCerebrolysin®.JNeuralTransmSuppl
62:265–275
19. Panisset M et al (2002) Cerebrolysin in Alzheimer’s disease: a randomized, double-blind,
placebo-controlledtrialwithaneurotropicagent.JNeuralTransm109:1089–1104
20. Xiao SF et al (2000) Efficacy of FPF 1070 (Cerebrolysin) in patients with Alzheimer’s
disease: a multicentre, randomized, double-blind placebo-controlled trial. Clin Drug Invest
19:43–53
21. Alvarez XA et al (2006) A 24-week double blind, placebo-controlled study of three dosages
of Cerebrolysin in patients with mild to moderate Alzheimer’s disease. Eur J Neurol
13:43–54
22. BarnesR,VeithR,OkimotoJetal(1992)Efficacyofantipsychoticmedicationsinbehavior-
allydisturbeddementiapatients.AmJPsychiatry139:1170–1174
226 L.I. Brusco
23. QaseemA,SnowV,CrossJTetal(2008)Currentpharmacologictreatmentofdementia:a
clinical practice guideline from the American College of Physicians and the American
AcademyofFamilyPhysicians.AnnInternMed148(5):370–378
24. SternRG,DuffelmeyerMEetal(1991)Theuseofbenzodiazepinesinthemanagementof
behavioralsymptomsindementiapatients.PsychiatrClinNorthAm14:375–384
25. TariotPN,ErbRetal(1994)Carbamazepinetreatmentofagitationinnursinghomepatients
withdementia:apreliminarystudy.JAmGeriatrSoc42:1160–1166
26. GleasonRP,SchneiderLS(1990)CarbamazepinetreatmentofagitationinAlzheimer’soutpatients
refractorytoneuroleptics.JClinPsychiatry51:115–118
27. Lott AD, Mcelroy SL, Keys MA (1995) Valproate in the treatment of behavioral agitation in
elderlypatientswithdementia.JNeuropsychiatryClinNeurosci7:314–319
Alzheimer’s Disease in Japan: Current
Situation and Issues of the Care for Persons
with Dementia
Akira Homma
Abstract It is estimated that the number of persons with dementia in 2005 will
double in 2035 in Japan from more than 2 million to 4.5 million. In the wake
of the introduction of long-term care insurance in Japan in 2000, there have
been particularly marked changes in the awareness and recognition of dementia.
However, considering that the aim of the care for persons with dementia is to
support their independence or daily life, we envisage many more issues ahead of us.
Early detection and diagnosis are important for all diseases, and particularly in the
case of dementia. Underdetection of persons with dementia means underdiagnosis
and treatment. At present, no data are available on the proportion of persons with
dementia diagnosed and treated appropriately. Such a situation clearly indicates the
role of primary care physicians to detect persons with dementia in the early stage.
Also, “dignity” is emphasized in Article 1 of the Long-term Care Insurance Law
revised in 2006. That is, persons with dementia must not be discriminated for the
reason of dementia. However, persons with dementia are often discriminated in
various situations. Here, I consider some of these issues with a view to establishing
a future ideal model.
Keywords Alzheimer’sdisease•Dementiacare•Humanright•Management
•Personswithdementia
Introduction
Dementia is an umbrella term used to refer to pathology from various causes.
According to the estimates made by Awata et al. [1], the number of persons with
dementia in 2005 will double in 30 years (in 2035) from more than 2 million to 4.5
A. Homma (*)
CenterforDementiaCareResearchandTraininginTokyo,Tokyo,Japan
e-mail: [email protected]
Fig. 1 Projectedincreaseofpersonswithdementia
million. In Japan, the number of persons with dementia will be increasing most
rapidlyinSaitamaPrefecture(3.2times).Theseestimateswereobtainedfromthe
epidemiological surveys that have been conducted by experts in certain areas
throughoutJapan.Otherestimateshavebeenobtainedbyestimatingthepercentage
of persons with dementia among those designated for the long-term care insurance
[2].Accordingtothese,approximatelyhalf(48%)ofthosedesignatedforlong-term
care insurance are suspected of having dementia. Furthermore, a tentative report on
thelong-termcareinsuranceinSeptember2008indicatedthattherewereapproxi-
mately 4.6 million individuals designated for this insurance in Japan. Forty-eight
percent of this figure is 2.21 million, which is comparable to the estimate made by
Awata et al. Given these numbers, dementia can be regarded as a very familiar
condition, as shown in Fig. 1.
In the wake of the introduction of long-term care insurance in Japan in 2000, there
have been particularly marked changes in the awareness and recognition of dementia.
However, considering that the aim of the care for persons with dementia is to support
their independence or daily life, we envisage many more issues ahead of us. Here, I
consider some of these issues with a view to establishing a future ideal model.
Early detection and diagnosis are important for all diseases, and particularly in the
case of dementia, because persons with dementia are less likely to complain of
memory impairment and visit medical facilities by themselves than those with other
diseases such as hypertension and diabetes. Those with very mild dementia may,
Alzheimer’sDiseaseinJapan 229
nevertheless, seek help for their memory loss as a chief complaint. However, most
of these individuals rarely visit a hospital, even if their family members, or others,
notice forgetfulness. Accordingly, the estimated number of persons with dementia
is relatively small compared to, for example, the estimated 16 million persons with
diabetes. However, it is critically important to determine the actual percentage of
potential persons with dementia who are able to visit a hospital and receive
appropriate diagnosis and treatment. Currently, there are no data available on the
number of such persons. Relatively old survey results provide several examples
demonstrating that early diagnosis in the community is not necessarily sufficient.
In an epidemiological survey of the elderly aged 65 and above, conducted
throughout Tokyo in 1995, approximately 5,000 persons were randomly sampled
from approximately 1.49 million people aged 65 and above [3]. Eventually, 123
personswerediagnosedashavingdementia.Ofthese123individuals,only26.5%
were diagnosed as having dementia by their primary care physicians. Thus,
three-fourths of persons with dementia may not actually be diagnosed as having
dementia. The severity of dementia among these patients was classified as mild
(18.5%),moderate(12.5%),andsevere(46.7%).Severedementiaisaccompanied
by total disorientation and requires total care in daily life. We should note that half
of the elderly with severe dementia were not diagnosed as having dementia. In the
results, the diagnoses of dementia by primary care physicians were Alzheimer’s
dementia (20.8%) and vascular dementia (56.8%). The percentage of those
diagnosed as having Alzheimer’s dementia was lower than those having vascular
dementia. These data seem very important, because primary care physicians were
the second person with whom family members taking care of the elderly with
dementia consulted at that time.
Further data were obtained from a questionnaire survey conducted to members
of the regional medical association in Tokyo before the introduction of the
long-term care insurance [4].Theresponseratewasfoundtobeaslowas50%
(n = 56). However, a relatively high percentage of the physicians replying
commented that they took care of symptoms of dementia in their daily practice
(73.2%)andthattheyrespondedtoconsultationswithpatients’familiesregarding
dementia(69.6%).However,only19.6%ofthephysicianssaidthattheyreferred
their patients to special medical facilities for diagnoses, etc., or that they used
public health centers. This situation is far from satisfactory. At this time,
immediatelybeforetheenforcementofthelong-termcareinsurance,only32.1%
of physicians were aware of the criteria for determining the independence level of
the elderly with dementia, which was an item of attending physicians’ written
opinion to be submitted to municipalities to determine the eligibility for the
long-term care insurance.
I give a third example here. Diagnoses of dementia made by physicians and
experts immediately before the enforcement of the long-term care insurance were
directly compared [5]. Primary care physicians and experts (four persons) made
diagnoses independently for 36 subjects, including healthy individuals, using the
NM scale, a behavior rating scale that measures the severity of dementia. The
results indicated that there was consistency in the diagnosis of severe dementia
230 A. Homma
between the physicians and specialists. However, of the eight persons diagnosed as
having mild dementia by the experts, the primary care physicians diagnosed four as
normal and one as having moderate dementia. Further, of the eight persons
diagnosed as having suspected dementia by the experts, the primary care physicians
diagnosed three as normal, another three as having mild dementia, and two as
having moderate dementia. Thus, the diagnoses were inconsistent. Of the eight
persons diagnosed with moderate dementia by the experts, six were diagnosed
similarly as having moderate dementia by the primary care physicians. Thus, the
less severe the disorder, the lower was the consistency rate.
Given these results, we can easily understand that primary care physicians play
a key role in early detection of dementia. However, in a paper entitled “Screening
forCognitiveImpairmentInGeneralPractice:TowardaConsensus”[6], Brodaty,
an Australian geriatric psychiatrist specializing in the community care of dementia,
considered various expenses, and concluded that it was inappropriate to seek
dementia screening from primary care physicians. I completely agree with this
view: one should not directly seek diagnosis from a primary care physician.
Establishing a network, including experts and health and welfare professionals with
whom you can consult at any time, is important. Also, in a model project of total
care collaborative system for the elderly with dementia, implemented by the Tokyo
Metropolitan Government, capable experts were secured in areas where a
counseling system for the elderly with dementia was established [7]. Conversely,
experts skilled in clinical practice for dementia are indispensable for facilitating
consultation projects, including the early detection of dementia.
Respondingtothissituation,in2000welaunchedatrainingmodelfortheearly
detectionanddiagnosisofdementiabyprimarycarephysicians.Onthebasisofthe
results of this model, in 2006 the Ministry of Health, Labour and Welfare
commenced education for primary care physicians to improve medical skills for
treating persons with dementia. To date, approximately 21,000 primary care
physicians have undergone this training. Such training has been reported to improve
the diagnostic rate of dementia, including Alzheimer disease, by primary care
physicians [8]. Figure 2 shows a conceptual structure for early detection and
management of persons with dementia in the community. At present, although not
every resource shown in the figure works satisfactorily, it seems that the structure
itself is quite valid in the management of persons with dementia. Furthermore, the
former Centers for Dementia Disorders were abolished in 2008. Subsequently,
MedicalCentersforDementiaDisordershavebeenfoundedascoreorganizations
responsible for the local management of dementia (Fig. 3). The target is to establish
150 such centers throughout Japan. In addition, since June 2009, a screening test
for dementia has been introduced into the training course for driver license renewal
at the age of 75. Those suspected of having dementia should undergo diagnosis for
the presence or absence of dementia by primary care physicians or experts when
they have a history of specific traffic violations within the past year. Thus, in view
of the rapidly increasing number of persons with dementia, primary care physicians
in local areas will be required to play more important roles, because dementia
cannot be handled only by experts.
Alzheimer’sDiseaseinJapan 231
Supporting physicians
Physicians in 47 prefectures and cities
of 500,000 or more, granted special Consultation
rights by government (supporting Consultationand Care Managers
physicians/regional medical advicetoPCPs Care staff
associations) Collaboration
Planning
Experts
Collaboration
Administration of educational Collaboration
program for PCPs “How to manage
persons with dementia”
Collaboration
Regional Medical Association Community
Primary Care Physicians Comprehensive
Support Center
Early detection placed in the
Referring to experts area of a junior
Management of physical Personwithdementia high school
conditions Family Support
Emotional support to family Support
caregivers
Collaboration of health
professionals in the community
Collaboration
Fig. 2 Supporting system for persons with dementia in the community with the involvement of
primarycarephysicians(PCPs)
Fig. 3 MedicalCenterforDementiaDisorders
232 A. Homma
The long-term care insurance has now been in force for 10 years. Certainly, our
understanding and recognition of dementia have improved markedly in some areas.
With this improvement, medical technology to facilitate early diagnosis has also
advanced, and the local network for early detection is also being consolidated. “Care
for the Elderly in 2015” [2], proposed by the Health and Welfare Bureau for the
Elderly, the Ministry of Health, Labor and Welfare in 2003, presents care for the
elderly with dementia as a new care model. In addition, the following is described in
Article 1 of the Long-term Care Insurance Law revised in 2006: “This law will allow
persons under conditions requiring care, such as bathing, excretion, and feeding,
functional training, and medical care, such as nursing and care management because
of illness resulting from physical and mental changes with aging, to maintain dignity
and lead an independent daily life according to their abilities.” (The rest is omitted.)
Thus, the term “dignity” was used for the first time. However, discrimination continues
to remain in spite of the changes in attitudes toward dementia [9]. A revised adult
guardianship law was introduced in April 2000. A total of 24,988 applications
associated with the adult guardianship law were filed in the year between April 2007
andMarch2008.Thelong-termcareinsurancecontractsaccountedfor6.2%(1,560
persons) of all the motives for the applications (Fig. 4; http://www.courts.go.jp/). In
2007, 2.3 million people were suspected of having dementia [1].Ofthese,thosewho
used the adult guardianship law for long-term care insurance contracts accounted for
only 0.007%. Not all people suspected of having dementia seem to use the adult
guardianship law for long-term care insurance contracts. However, it does not
necessarily seem to be wrong to consider that those with dementia have slightly
impaired judgment. Thus, it is certainly desirable to use the adult guardianship law
more actively also for application for long-term care insurance contracts, because it is
Inheritance 3,050
Others 2,005
Fig. 4 Numberofapplicantstotheadultguardianshipsystemanditsmotives:April2007–March
2008,n=24,988
Alzheimer’sDiseaseinJapan 233
a useful means for protecting the human rights of persons with dementia. All persons
involved should always consider health, medicine, welfare, care, and family in connec-
tion with dementia, thereby supporting the dignity of persons with dementia.
The second issue naturally concerns measures against behavioral and psychological
symptoms. According to the results of a questionnaire survey that we conducted
among members (n=1,049)oftheJapanesePsychogeriatricSocietyandtheJapanese
AssociationofPsychiatricHospitals,whichhasestablisheddementiatreatmentwards
and medical treatment wards [10], among behavioral and psychological symptoms, the
drug of first choice for hallucination, delusion, abusive language and violence, and
delirium was risperidone, an atypical antipsychotic agent. As yet, no final conclusion
has been reached concerning the effectiveness of antipsychotic drugs on the behavioral
and psychological symptoms of dementia. However, antipsychotic drugs are
indispensable for supporting the lives of the elderly with dementia, either at home or
in institutions, as a therapeutic procedure for hallucination and delusion that may cause
dangerous situations for persons with dementia and their families when they develop.
We should administer such medication with periodic monitoring, comprehensively
considering the actual needs and health conditions of the patients and the effectiveness
and side effects expected. In the present situation, off-label use of antipsychotic drugs
is inevitable for the behavioral and psychological symptoms of dementia. Experts are
expected to be skilled in the use of these drugs. However, the most serious issue in the
surveillance is the situation where no informed consent has been obtained in the more
than half of patients who are administered antipsychotic drugs. Although no stratified
analysis has been conducted on this data, regardless of use status, informed consent
should be obtained appropriately for any treatment using an antipsychotic drug.
Furthermore, the concept of informed consent should be examined for persons with
dementia with notably impaired mental capacity. This is one of the serious issues
regarding the current adult guardianship law and requires immediate revision.
The third issue is the response to caregivers and families. In recent years, many
outpatients with milder dementia have visited a hospital for diagnosis. In less severe
cases, disclosing to persons with dementia the proper name of their disease is more
important. Next, the circumstances of caregivers and families, as well as those of the
persons with dementia, should be accurately evaluated. Caregivers often find it difficult
to recognize the facts. Naturally, they may be unwilling to accept or want to deny the
reality, and may therefore be confused for a while. In such situations, it is impossible
for a caregiver to properly take in the variety of information that they may be provided.
As a matter of priority, caregivers’ feelings should be acknowledged with sympathy.
Such responses are required from all persons concerned, including physicians.
Early detection, precise diagnoses, and management are essential requirements for
persons with dementia. Simultaneously, support is needed to allow those with dementia
to live in the community. To meet these requirements, we should maintain close
234 A. Homma
communication with those involved in health, medicine, welfare, and care based on
appropriate diagnoses. This activity will greatly influence the therapeutic effects,
course, and prognosis. However, in view of the present educational curriculum described
above, we cannot deny the bias of its contents toward diagnostics. In contrast to
diagnosis or treatment skills, it is certainly difficult to evaluate these roles to support
persons with dementia in the community. Currently, five dementia-associated academic
societies(theJapanPsychogeriatricSociety,theJapanSocietyforDementiaResearch,
the Japanese Society of Neurology, the Japan Geriatrics Society, and the Japanese
Society of Neurological Therapeutics) are jointly drawing up a guideline for the
diagnoses and treatment of dementia. In this guideline, emphasizing their philosophy
for diagnoses, treatment, and care of persons with dementia is extremely important.
In addition, we should continuously construct a structure to maintain the quality
of care for dementia and simultaneously conduct positively educational and
enlightening activities, including familiarizing such care to the public.
References
1. Awata S et al (2000) Model of integrated emergency care for dementia; Grants-in-Aid for
Scientific Research, Ministry of Health, Labour and Welfare, 2007; The Health Science
Research of Heart, Psychiatric Emergency Care; Study report on treatment of physical
disorders and complications of dementia, pp 135–156
2. Elderly Care Study Group (2003) Elderly Care in 2015 – for the establishment of care to
support dignity of the elderly – Elderly Care Study Group. Houken, Tokyo
3. Bureau of Social Welfare and Public Health, Tokyo Metropolitan Government: Special
investigation report on daily life and health of the elderly in 1995; 1995, Bureau of Social
WelfareandPublicHealth,TokyoMetropolitanGovernment,1995
4. HommaA(2000)Geriatricpsychiatry:millennium.JpnJGeriatrPsychiatry11:68–69
5. Office for the Promotion of Measures for the Elderly, Tokyo Metropolitan Government:
SummaryoftheinterimreportfromtheCommitteeforEstablishmentofServiceOrganization
fortheElderly,TokyoMetropolitanGovernmentin1998;1999,OfficeforthePromotionof
MeasuresfortheElderly,TokyoMetropolitanGovernment,1999
6. Brodaty H et al (1998) Screening for cognitive impairment in general practice: toward a
consensus.AlzheimerDisAssocDisord12:1–13
7. Office for the Promotion of Measures for the Elderly, Tokyo Metropolitan Government:
ReportfromtheCommitteeforEvaluationofTotalCareCollaborationSystemfortheElderly
withDementia,TokyoMetropolitanGovernment2000,OfficeforthePromotionofMeasures
fortheElderly,TokyoMetropolitanGovernment,2000
8. Homma A, Awata S, Ikeda M, Ueki A, Urakami K, Kitamura S, Shigeta M, Nakamura Y,
NakamuraM,OhtaK(2006)Localcollaborationfortheelderlywithdementia:modelproject
toimprovediagnosticskillsofdementiabyprimarycarephysicians.JpnJGeriatrPsychiatry
17:483–489
9. Homma A (2007) Issues of the adult guardianship law concerning informed consent for
treatment of the elderly with dementia. In: Arai M (ed) Adult guardianship and medical
practice. Nippon Hyoronsha, Tokyo, pp 19–31
10. Homma A (2006) Questionnaire survey about use status of antipsychotic drugs for behavioral
andpsychologicalsymptomsofdementia(BPSD).JpnJGeriatrPsychiatry17:799–783
AD-FTLD Spectrum: New Understanding
of the Neurodegenerative Process
from the Study of Risk Genes
Keywords Alzheimer’sdisease•Frontotemporallobardegeneration•Progranulin
•Tau•TDP-43
Introduction
and burden to caregivers, dementia is regarded one of the most malignant diseases
in this century. There are 24.3 million dementia patients in the world, and 4.6
million people are newly diagnosed every year [1]. In Japan alone there are now 1.5
million dementia patients, and the number of dementia patients is steadily
increasing for reasons of the extension of the average lifespan. It is expected there
will be 3.5 million dementia patients in Japan by the year 2035.
Alzheimer’s disease (AD) is the most frequent primary neurodegenerative
disease, accounting for 50–70% of all dementia patients. Higher age, female
gender, family history of dementia, head injury, and lower education are reported
to be the risks for AD, among which higher age is the most significant. The
prevalence rate of AD increases age after 65, almost doubling by every 5 years, and
reaching more than 40% prevalence in the elderly over 85 years old [2].
Primary neurodegenerative diseases causing dementia include AD, diffuse Lewy
body disease (DLB), frontotemporal lobar degeneration (FTLD), and corticobasal
degeneration (CBD). Each disease shows characteristic clinical signs and symptoms.
The initial sign of AD is memory impairment, and later impairment in cognitive
function follows, such as aphasia, apraxia, agnosia, and executive dysfunction.
Those impaired cognitive functions together result in dysfunction in judgment. DLB
is the second most common disease among the elderly more than 75 years old, char-
acterized by varying levels of cognitive function, visual hallucination, and parkin-
sonism. The neuropathological feature of DLB is abundant Lewy bodies in the
cerebral cortical neurons. In persons of younger age, less than 65 years old, FTLD
is the second predominant dementing disease [3]; it is characterized by personality
change, dysinhibition, abnormal behavior, and language disability. In the early phase
of FTLD, memory can be preserved despite the deteriorating daily life of the
patients. The classification of frontotemporal dementia has been discussed for many
years, and FTLD is the broadest concept, which includes Pick’s disease and other
diseases characterized by progressive lobar atrophy of frontal and temporal lobe.
FTLD is classified into three types: (1) frontotemporal dementia (FTD), (2) progres-
sive aphasia, and (3) semantic dementia. FTD is further divided into frontal lobar
degeneration, Pick’s type, and motor neuron disease type (Fig. 1).
FLD type
Cause % of Cases
Chromosomal (Down syndrome) <1%
Familial ~25%
Late-onset familial (AD2) 15% -25%
Amyloid beta
AD1 10%-15% APP 21q21 Clinical
A4 protein
Chromosomal
Locus name Gene symbol Protein name Test availability
locus
protein (VCP) was reported as the component of FTLD-U [19], and in 2005
charged multivesicular body protein (CHMP2B) was reported [20] as genes related
to FTLD-U inclusion body formation. However, there are only a few cases caused
by mutation in VCP or CHMP2B, and further search for the genetic cause of
FTLD-U has continued.
Copy number variation (CNV) is the repetition of a sequence 1 kb long and more
including several genes. CNV is now shown to be the causative factor of many dis-
orders, including chronic neuroimmunological (CND) disease. More than 20,000
CNVs exist in the whole genome, which has more than 7,000 genes. In 2006, CNV
including the APP gene was reported with five families of AD [36] whose CNV
region covers 0.58–6.37 M, including APP and 5–12 other genes. The phenotype of
the cases of this CNV is AD with cerebral amyloid angiopathy (CAA), and it was
implied that an increased copy number of APP results in AD pathogenesis [37].
Considering the pathogenesis of Down syndrome, which is caused by the increased
copy number of APP that is included in the 21q21region, it is quite reasonable to
understand this increased copy number of APP is causing AD pathology in the brain
of Down syndrome patients more than 30 years old [38]. CNV of the alpha-cynuclain
gene is also reported with familial PD patients [39, 40].
These findings support the notion that increased copy number of a certain gene
can lead to the deposition of abnormal protein, causing neurodegenerative demen-
tia. Increased numbers of APP may explain the pathogenesis of AD, and this also
supports the validity of the amyloid cascade hypothesis.
Increased expression of APP may be caused by several different mechanisms
other than CNV. For example, there might be mutations in promoter regions, which
could increase the expression level of APP gene up to 50%. Increased expression
of APP may increase the risk of AD. In fact, there are papers reporting that
AD-FTLD Spectrum: New Understanding of the Neurodegenerative Process 241
mutations in the promoter region of APP cause a 1.2–1.8 increased expression level
of APP in Dutch and Belgium families with AD [41, 42].
In addition to gene replication, several different types of partial or complete
deletion of genes are reported with families with AD or FTLD. For example, exon
9 deletion of PSEN1 causes AD with massive deposition of characteristic cotton
wool plaques in the brain of the patients [43]. Deletion of PGRN or MAPT are
reported with FTLD families [44]. There is a paper reporting partial deletion of
exon 6–9 of MAPT gene [45], and the tau protein coded by the deleted MAPT gene
shows decreased binding activity with microtubules and shows more binding with
MAP1B, which might result in the neurodegeneration caused by defective tau
protein.
The importance of miRNA should be mentioned in this context. miRNA is an
endogenous small RNA molecule that supposedly stimulates mRNA degradation or
regulates gene expression after binding with the target mRNA. There are more than
1,000 different kinds of miRNA in the human genome, which are supposed to be
relevant to control of the expression level of more than 30% of genes. Especially,
more miRNA exist in brain tissue. Search with miRNA array has revealed the
significant reduction of miR-107 in the brain with AD [46], which is correlated
with the severity of AD pathology. It was also shown that miR-107 binds with
mRNA of BACE1 and speculated that reduction of miR-107 results in the increased
expression level of BACE1. It is quite reasonable that reduction in miRNA will
cause AD pathology through higher expression of BACE1. Significant increase of
miR-146a in the AD brain tissue is reported compared with that of the healthy
control brain by pooled RNA sample study [47].
These findings just described show the possibility that the change in expression
level of certain genes will increase the risk of neurodegenerative diseases such as
AD and FTLD, which may lead to a proposal that explains the common neuro-
pathological process among neurodegenerative dementia.
AD-FTLD Spectrum
In the brain of AD patients, there are abnormal depositions of beta amyloid and tau
protein. Beta amyloid deposits in the core of senile plaque as well as in the cerebral
vessel wall of CAA. Tau is deposited in the soma of degenerating neurons, in ghost
tangles, and in dystrophic neuritis as NFT. On the other hand, in the brain of FTLD,
tau protein is deposited in Pick body (FTLD-tau) and TDP-43 is deposited
intracellularly and intranuclearly as a ubiquitin-positive inclusion body as inclusion
body (FTLD-U).
There are 32 different missense mutations reported with the APP gene. Most of
these mutations cause AD pathology, such as KM670/671NL Swedish mutation,
V717I,P,G London mutation, V715M France mutation, V715A German mutation,
and I716V Florida mutation. However, some APP mutations are observed in
hereditary cerebral hemorrhage with angiopathy, such as HCHWA Dutch (E693Q)
242 M. Takeda et al.
and CAA Italian (E693K). Flemish-type APP mutation (Flemish A692G) causes
CAA as well as AD pathology.
There are 177 different mutations reported with the PSEN gene, most of which
cause early-onset type AD. The earliest onset of AD is reported with a subject of
24 years old, implying strong pathogenic effects of PSEN1 gene mutation. It should
be noticed that there are PSEN1 mutations whose phenotype is FTLD symptoms
(L113P, G183V, insArg352).
There are 44 different mutations reported with MAPT, most of which are
concentrated in 9–13 exons and an intron between exon 10 and 11. The major
phenotype of MAPT mutations is FTLD with typical Pick body and FTLD-tau inclu-
sion body in the intracellular and intranuclear space. The characteristics of MAPT
mutations is the fact that they show a wide range of symptoms of FTLD, whose
major symptom is parkinsonism (i.e., FTDP-17), or with behavioral abnormalities,
or aphasia. There are also tau mutations whose symptoms are very similar to those
of AD. For example, patients with the MAPT R406W mutation show memory
impairment in the initial stage and then gradually show FTLD symptoms.
FTLD-tau inclusion is observed in 25% of FTLD patients, and the majority of
FTLD patients show a tau-negative, ubiquitin-positive inclusion (FTLD-U), which
is composed of TDP-43. LOF of PGRN is closely related with pathogenesis of
FTLKD-U formation. Mutation of PGRN explains about 25% of familial FTLD,
which coincides with the frequency of the MAPT mutation among FTLD cases.
There is a wide variety of onset age with FTLD patients with MAPT or PGRN
mutations: 20–70 years for MAPT mutations and 30–80 years for PRGN mutations.
When the average onset age is compared between patients with PGRN mutation
(61 ± 9 years old) and those with MAPT mutations (48 ± 10 years old), the PGRN
mutation causes FTLD with later-onset age.
Furthermore, there are cases with AD phenotypes and ALS phenotypes among
the patients with PGRN mutations.
PSEN PGRN
References
1. Ferri CP, Prince M, Brayne C et al (2005) Global prevalence of dementia: a Delphi consensus
study. Lancet 366:2112–2117
2. Breteler MM, Claus JJ, van Duijn CM et al (1992) Epidemiology of Alzheimer’s disease.
Epidemiol Rev 14:59–82
3. Graff-Radford NR, Woodruff BK (2007) Frontotemporal dementia. Semin Neurol 27:48–57
4. Neary D, Snowden JS, Gustafson L et al (1998) Frontotemporal lobar degeneration: a consen-
sus on clinical diagnostic criteria. Neurology 51(6):1546–1554
5. Goate A, Chartier-Harlin MC, Mullan M et al (1991) Segregation of a missense mutation in
the amyloid precursor protein gene with familial Alzheimer’s disease. Nature (Lond)
349:704–706
6. Sherrington R, Rogaev EI, Liang Y et al (1995) Cloning of a gene bearing missense mutations
in early-onset familial Alzheimer’s disease. Nature (Lond) 375:754–760
7. Levy-Lahad E, Wasco W, Poorkaj P et al (1995) Candidate gene for the chromosome 1
familial Alzheimer’s disease locus. Science 269:973–977
8. Hardy J (1997) Amyloid, the presenilins and Alzheimer’s disease. Trends Neurosci
20:154–159
9. De Strooper B (2003) Aph-1, Pen-2, and nicastrin with presenilin generate an active
gamma-secretase complex. Neuron 38:9–12
10. Bird TD (2008) Genetic aspects of Alzheimer disease. Genet Med 10:231–239
11. Strittmatter WJ, Saunders AM, Schmechel D et al (1993) Apolipoprotein E: high-avidity
binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial
Alzheimer disease. Proc Natl Acad Sci USA 90:1977–1981
12. Farrer LA, Cupples LA, Haines JL et al (1997) Effects of age, sex, and ethnicity on the
association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis.
APOe and Alzheimer disease meta analysis consortium. JAMA 278:1349–1356
13. Corder EH, Saunders AM, Strittmatter WJ et al (1993) Gene dose of apolipoprotein E type 4
allele and the risk of Alzheimer’s disease in late onset families. Science 261:921–923
14. Mahley RW, Weisgraber KH, Huang Y (2006) Apolipoprotein E4: a causative factor and
therapeutic target in neuropathology, including Alzheimer’s disease. Proc Natl Acad Sci USA
103:5644–5651
244 M. Takeda et al.
15. Drzezga A, Grimmer T, Henriksen G et al (2009) Effect of APOe genotype on amyloid plaque
load and gray matter volume in Alzheimer disease. Neurology 72:1487–1494
16. Reiman EM, Chen K, Liu X et al (2009) Fibrillar amyloid-beta burden in cognitively normal
people at 3 levels of genetic risk for Alzheimer’s disease. Proc Natl Acad Sci USA
106:6820–6825
17. Rademakers R, Hutton M (2007) The genetics of frontotemporal lobar degeneration. Curr
Neurol Neurosci Rep 7:434–442
18. Hutton M, Lendon CL, Rizzu P et al (1998) Association of missense and 5¢-splice-site
mutations in tau with the inherited dementia FTDP-17. Nature (Lond) 393:702–705
19. Watts GD, Wymer J, Kovach MJ et al (2004) Inclusion body myopathy associated with Paget
disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein.
Nat Genet 36:377–381
20. Skibinski G, Parkinson NJ, Brown JM et al (2005) Mutations in the endosomal ESCRTIII
complex subunit CHMP2B in frontotemporal dementia. Nat Genet 37:806–808
21. Baker M, MacKenzie IR, Pickering-Brown SM et al (2006) Mutations in progranulin cause
tau-negative frontotemporal dementia linked to chromosome 17. Nature (Lond)
442:916–919
22. Cruts M, Gijselinck I, van der Zee J et al (2006) Null mutations in progranulin cause
ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature (Lond)
442:920–924
23. He Z, Bateman A (2003) Progranulin (granulin–epithelin precursor, PC-cell-derived growth
factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med 81:600–612
24. He Z, Ong CH, Halper J et al (2003) Progranulin is a mediator of the wound response. Nat
Med 9:225–229
25. Van Damme P, Van Hoecke A, Lambrechts D et al (2008) Progranulin functions as a neurotrophic
factor to regulate neurite outgrowth and enhance neuronal survival. J Cell Biol 181:37–41
26. Neumann M, Sampathu DM, Kwong LK et al (2006) Ubiquitinated TDP-43 in frontotemporal
lobar degeneration and amyotrophic lateral sclerosis. Science 314:130–133
27. Buratti E, Baralle FE (2008) Multiple roles of TDP-43 in gene expression, splicing regulation,
and human disease. Front Biosci 13:867–878
28. Gregory RI, Yan KP, Amuthan G et al (2004) The microprocessor complex mediates the
genesis of microRNAs. Nature (Lond) 432:235–240
29. Amador-Ortiz C, Lin WL, Ahmed Z et al (2007) TDP-43 immunoreactivity in hippocampal
sclerosis and Alzheimer’s disease. Ann Neurol 61:435–445
30. Higashi S, Iseki E, Yamamoto R et al (2007) Concurrence of TDP-43, tau and alpha-synuclein
pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res
1184:284–294
31. Nakashima-Yasuda H, Uryu K, Robinson J et al (2007) Co-morbidity of TDP-43 proteinopathy
in Lewy body related diseases. Acta Neuropathol 114:221–229
32. Kabashi E, Valdmanis PN, Dion P et al (2008) TARDBP mutations in individuals with spo-
radic and familial amyotrophic lateral sclerosis. Nat Genet 40:572–574
33. Sreedharan J, Blair IP, Tripathi VB et al (2008) TDP-43 mutations in familial and sporadic
amyotrophic lateral sclerosis. Science 319:1668–1672
34. Morita M, Al-Chalabi A, Andersen PM et al (2006) A locus on chromosome 9p confers sus-
ceptibility to ALS and frontotemporal dementia. Neurology 66:839–844
35. Vance C, Al-Chalabi A, Ruddy D et al (2006) Familial amyotrophic lateral sclerosis with
frontotemporal dementia is linked to a locus on chromosome 9p13.2–21.3. Brain
129:868–876
36. Rovelet-Lecrux A, Hannequin D, Raux G et al (2006) APP locus duplication causes auto-
somal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet
38:24–26
37. Sleegers K, Brouwers N, Gijselinck I et al (2006) APP duplication is sufficient to cause early
onset Alzheimer’s dementia with cerebral amyloid angiopathy. Brain 129:2977–2983
AD-FTLD Spectrum: New Understanding of the Neurodegenerative Process 245
38. Wisniewski KE, Dalton AJ, McLachlan C et al (1985) Alzheimer’s disease in Down’s syn-
drome: clinicopathologic studies. Neurology 35:957–961
39. Singleton AB, Farrer M, Johnson J et al (2003) Alpha-synuclein locus triplication causes
Parkinson’s disease. Science 302:841
40. Chartier-Harlin MC, Kachrgus J, Roumier C et al (2004) Alpha-synuclein locus duplication
as a cause of familial Parkinson’s disease. Lancet 364:1167–1169
41. Brouwers N, Sleegers K, Engelborghs S et al (2006) Genetic risk and transcriptional vari-
ability of amyloid precursor protein in Alzheimer’s disease. Brain 129:2984–2991
42. Theuns J, Brouwers N, Engelborghs S et al (2006) Promoter mutations that increase amyloid
precursor-protein expression are associated with Alzheimer disease. Am J Hum Genet
78:936–946
43. Crook R, Verkkoniemi A, Perez-Tur J et al (1998) A variant of Alzheimer’s disease with
spastic paraparesis and unusual plaques due to deletion of exon 9 of presenilin 1. Nat Med
4:452–455
44. Rovelet-Lecrux A, Deramecourt V, Legallic S et al (2008) Deletion of the progranulin gene in
patients with frontotemporal lobar degeneration or Parkinson disease. Neurobiol Dis
31:41–45
45. Rovelet-Lecrux A (2009) Partial deletion of the MAPT gene: a novel mechanism of FTDP-17.
Hum Mutat 30:E591–E602
46. Wang WX, Rajeev BW, Stromberg AJ et al (2008) The expression of microRNA miR-107
decreases early in Alzheimer’s disease and may accelerate disease progression through regula-
tion of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci 28:1213–1223
47. Lukiw WJ, Zhao Y, Cui JG (2008) An NF-kB-sensitive micro RNA-146amediated inflamma-
tory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem
283:31315–31322
Dementia with Lewy Bodies
Abstract Dementia with Lewy bodies (DLB) has over the last two decades become
recognised as a common dementia subtype in older people. Since it shares clinical and
pathological features with both Alzheimer’s disease and Parkinson’s disease, knowl-
edge of DLB is essential not only in its own right, but also to obtain a full understand-
ing of its two prototypical boundary disorders. This chapter reviews what is currently
known about the clinical, pathological and management aspects of DLB.
Keywords Alpha-synuclein•Dementia•Diagnosis•Lewybody•Parkinson’s
Introduction
Dementia with Lewy bodies (DLB), which is now thought to be the second most
prevalent cause of degenerative dementia in older people [1] has previously carried
a variety of diagnostic labels, including diffuse Lewy body (LB) disease [2], LB
dementia [3], the LB variant of Alzheimer’s disease [4], senile dementia of LB type
[5], and dementia associated with cortical Lewy bodies [6]. The latest International
Consensus criteria describe a spectrum of LB disorders with DLB and Parkinson’s
disease (PD) dementia as two operationally defined phenotypes which are now in
widespread clinical use [7]. Primary autonomic failure, rapid eye movement sleep
disorder, and PD itself are other syndromes considered to lie along the same con-
tinuum. and debate continues about the nature of their interrelationships. The
importance of recognizing DLB relates particularly to its pharmacological man-
agement, with reports of good responsiveness to acetylcholinesterase inhibitors
(AChEIs)[8], extreme sensitivity to the side effects of neuroleptics [9, 10], and
limited responsiveness to levo-dopa [11].
Epidemiology
Alpha (a)-synuclein-positive LBs and Lewy neurites (LNs) are the characteristic
autopsy lesions of DLB. Up to a quarter of LB disease cases show a pathological
distribution that has extensive neocortical and limbic involvement with relative
sparing of midbrain and subcortical structures, that is, not conforming to typical
“Braak-like” Parkinson’s disease (PD) spread. There is often also abundant
Alzheimer-type pathology, predominantly in the form of amyloid plaques.
Tau-positive inclusions and neocortical neurofibrillary tangles sufficient to
qualify for a diagnosis of concomitant AD (Braak stages V or VI) occur in only
10–25% of cases. Alzheimer pathology is not needed for dementia to occur
because a small number of “pure” DLB cases are seen with typical cognitive
impairment and neuropsychiatric features. Cortical LB and LN density is not
robustly correlated with either the severity or duration of dementia [17, 18], and
in some community-based series, Lewy-type pathologies are frequently seen in
nondemented, neurologically normal individuals [19, 20]. This finding raises the
intriguing possibility that LBs are a neurprotective response to the generation of
low molecular weight species of a-synuclein that are located in synaptosomes and
impair synaptic function [21]. Synaptic dysregulation and neurotransmitter defi-
cits may therefore be turn out to be better correlates of the fluctuating clinical
picture [22] and provide the best drug targets. a-Synuclein immunoreactive
deposits with many of the characteristics of LB have also been reported in a high
proportion of AD cases usually occurring exclusively in the amygdala [23]. In this
context they may simply reflect end-stage aggregation of a-synuclein in severely
dysfunctional neurons that are already heavily damaged by plaque and tangle
pathology [24, 25]. Triplication of the a-synuclein gene (SNCA) can cause DLB,
PD, and Parkinson’s disease dementia (PDD), whereas gene duplication is asso-
ciated only with motor PD, suggesting a gene dose effect [26]. However, SNCA
multiplication is not found in most LB disease patients [27]. Mutations in the
Dementia with Lewy Bodies 249
Clinical Features
Investigations
matter changes, and rates of progression of whole brain atrophy [52] appear to be
unhelpful in differential diagnosis. An important recent development for the diag-
nosis of DLB is the visualization of presynaptic dopaminergic deficits in the striatum
using 123I-radiolabeled 2-beta-carbomethoxy-3 beta-(4-iodophenyl)-N-(3-fluoropropyl)
nortropane (FP-CIT; trade name, DaTSCAN). This technique demonstrates high
sensitivity (78%) and specificity (90%) in identifying probable DLB versus
non-DLB dementia [53, 54]. Although the current (European) regulatory approval
for FP-CIT imaging is in distinguishing probable DLB from AD, the more valuable
clinical use may be in determining clinically uncertain cases [55].
Diagnostic Criteria
References
1. McKeith IG, Galasko D, Kosaka K et al (1996) Consensus guidelines for the clinical and
pathologic diagnosis of dementia with Lewy bodies (DLB): report of the Consortium on DLB
International Workshop. Neurology 47:1113–1124
2. Kosaka K, Yoshimura IK et al (1984) Diffuse type of Lewy body disease: progressive dementia
with abundant cortical Lewy bodies and senile changes of varying degree – a new disease?
Clin Neuropathol 3(5):185–192
3. Gibb WRG, Esiri MM, Lees AJ (1987) Clinical and pathological features of diffuse cortical
Lewy body disease (Lewy body dementia). Brain 110:1131–1153
4. Hansen L, Salmon D, Galasko D et al (1990) The Lewy body variant of Alzheimer’s disease:
a clinical and pathologic entity. Neurology 40:1–8
5. Perry RH, Irving D, Blessed G et al (1990) Senile dementia of Lewy body type. A clinically and
neuropathologically distinct form of Lewy body dementia in the elderly. J Neurol Sci 95:119–139
6. Byrne EJ, Lennox G, Godwin-Austen LB et al (1991) Dementia associated with cortical Lewy
bodies. Proposed diagnostic criteria. Dementia 2:283–284
7. McKeith IG, Dickson DW, Lowe J et al (2005) Diagnosis and management of dementia with
Lewy bodies. Third report of the DLB consortium. Neurology 65:1863–1872
8. McKeith I, Del Ser T, Spano P et al (2000) Efficacy of rivastigmine in dementia with Lewy
bodies: a randomised, double-blind, placebo-controlled international study. Lancet
356:2031–2036
9. McKeith I, Fairbairn A, Perry R et al (1992) Neuroleptic sensitivity in patients with senile
dementia of Lewy body type. Br Med J 305:673–678
10. Ballard C, Grace J, McKeith I et al (1998) Neuroleptic sensitivity in dementia with Lewy
bodies and Alzheimer’s disease. Lancet 351:1032–1033
11. Molloy S, McKeith IG, O’Brien JT et al (2005) The role of levodopa in the management of
dementia with Lewy bodies. J Neurol Neurosurg Psychiatry 76:1200–1203
12. Rahkonen T, Eloniemi-Sulkava U, Rissanen S et al (2003) Dementia with Lewy bodies
according to the consensus criteria in a general population aged 75 years or older. J Neurol
Neurosurg Psychiatry 74(6):720–724
13. Shergill S, Mullen E, D’Ath P et al (1994) What is the clinical prevalence of Lewy body
dementia? Int J Geriatr Psychiatry 9:907–912
14. Stevens T, Livingston G, Kitchen G et al (2002) Islington study of dementia subtypes in the
community. Br J Psychiatry 180:270–276
15. Holmes C, Cairns N, Lantos P et al (1999) Validity of current clinical criteria for Alzheimer’s
disease, vascular dementia and dementia with Lewy bodies. Br J Psychiatry 174:45–51
16. Singleton AB, Wharton A, O’Brien KK et al (2002) Clinical and neuropathological correlates
of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord
14:167–175
17. Harding AJ, Halliday GM (2001) Cortical Lewy body pathology in the diagnosis of dementia.
Acta Neuropathol 102:355–363
18. Gómez-Tortosa E, Newall K, lrizarry MC et al (1999) Clinical and quantitative pathological
correlates of dementia with Lewy bodies. Neurology 53:1284–1291
19. Parkkinen L, Kauppinen T, Pirttilä T et al (2005) Alpha-synuclein pathology does not predict
extrapyramidal symptoms or dementia. Ann Neurol 57:82–91
20. Zaccai J, Brayne C, McKeith I et al (2008) Patterns and stages of alpha-synucleinopathy:
relevance in a population-based cohort. Neurology 70:1042–1048
21. Kramer ML, Schulz-Schaeffer WJ (2007) Presynaptic alpha-synuclein aggregates, not Lewy
bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci 27:1405–1410
22. Perry EK, Piggott MA, Johnson M et al (2003) Neurotransmitter correlates of neuropsychiatric
symptoms in dementia with Lewy bodies. In: Bedard M-A, Agid Y, Chouinard S et al (eds)
Mental and behavioral dysfunction in movement disorders. Humana, Totowa, NJ, pp. 285–294
23. Hamilton RL (2000) Lewy bodies in Alzheimer’s disease: a neuropathological review of 145
cases using alpha-synuclein immunohistochemistry. Brain Pathol 10:378–384
Dementia with Lewy Bodies 253
24. Lippa CF, McKeith I (2003) Dementia with Lewy bodies: improving diagnostic criteria.
Neurology 60:1571–1572
25. Uchikado H, Lin WL, DeLucia M et al (2006) Alzheimer disease with amygdala Lewy bodies:
a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol 65:685–697
26. Singleton A, Gwinn-Hardy K (2004) Parkinson’s disease and dementia with Lewy bodies:
a difference in dose? Lancet 364(9440):1105–1107
27. Johnson J, Hague SM, Hanson M et al (2004) SNCA multiplication is not a common cause of
Parkinson disease or dementia with Lewy bodies. Neurology 63(3):554–556
28. Clark LN, Kartsaklis LA, Wolf Gilbert R et al (2009) Association of glucocerebrosidase mutations
with dementia with Lewy bodies. Arch Neurol 66:578–583
29. Hardy J, Lewis P, Revesz T et al (2009) The genetics of Parkinson’s syndromes: a critical
review. Curr Opin Genet Dev 19:254–265
30. Ballard C, O’Brien J, Morris CM et al (2001) The progression of cognitive impairment in
dementia with Lewy bodies, vascular dementia and Alzheimer’s disease. Int J Geriatr
Psychiatry 16(5):499–503
31. Walker Z, Allen RL, Shergill S et al (2000) Three years survival in patients with a clinical
diagnosis of dementia with Lewy bodies. Int J Geriatr Psychiatry 15:267–273
32. Hanyu H, Sato T, Hirao K et al (2009) Differences in clinical course between dementia with
Lewy bodies and Alzheimer’s disease. Eur J Neurol 16:212–217
33. Armstrong TP, Hansen LA, Salmon DP et al (1991) Rapidly progressive dementia in a patient
with the Lewy body variant of Alzheimer’s disease. Neurology 41:1178–1180
34. Aarsland D, Ballard C, McKeith I et al (2001) Comparison of extrapyramidal signs in dementia
with Lewy bodies and Parkinson’s disease. J Neuropsychiatry Clin Neurosci 13:374–379
35. Burn DJ, Rowan EN, Minett T et al (2003) Extrapyramidal features in Parkinson’s disease
with and without dementia and dementia with Lewy bodies: a cross-sectional comparative
study. Mov Disord 18(8):884–889
36. Ballard C, O’Brien J, Swann A et al (2000) One year follow-up of parkinsonism in dementia
with Lewy bodies. Dement Geriatr Cogn Disord 11:219–222
37. Duda JE, Giasson BI, Mabon ME et al (2002) Novel antibodies to synuclein show abundant
striatal pathology in Lewy body diseases. Ann Neurol 52(2):205–210
38. Walker MP, Ayre GA, Cummings JL et al (2000) The Clinician Assessment of Fluctuation and
the One Day Fluctuation Assessment Scale. Two methods to assess fluctuating confusion in
dementia. Br J Psychiatry 177:252–256
39. Ballard CG, Mohan RNC, Patel A et al (1993) Idiopathic clouding of consciousness – do the
patients have cortical Lewy body disease? Int J Geriatr Psychiatry 8:571–576
40. Bradshaw J, Saling M, Hopwood M et al (2004) Fluctuating cognition in dementia with Lewy
bodies and Alzheimer’s disease is qualitatively distinct. J Neurol Neurosurg Psychiatry
75:382–387
41. Ferman T, Smith GE, Boeve BF et al (2004) DLB fluctuations: specific features that reliably
differentiate from AD and normal aging. Neurology 62(2):181–187
42. Mosimann UP, Rowan EN, Partington CE et al (2006) Characteristics of visual hallucinations
in Parkinson disease dementia and dementia with Lewy bodies. Am J Geriatr Psychiatry
14(2):153–60
43. Birkett DP, Desouky A, Han L et al (1992) Lewy bodies in psychiatric patients. Int J Geriatr
Psychiatry 7:235–240
44. Boeve BF, Silber MH, Ferman TJ (2004) REM sleep behavior disorder in Parkinson’s disease
and dementia with Lewy bodies. J Geriatr Psychiatry Neurol 17(3):146–157
45. Klatka LA, Louis ED, Schiffer RB (1996) Psychiatric features in diffuse Lewy body disease:
findings in 28 pathologically diagnosed cases. Neurology 46:A180
46. Ballard C, Shaw F, McKeith I et al (1998) High prevalence of neurovascular instability in
neurodegenerative dementias. Neurology 51:1760–1762
47. Del-Ser T, Munoz DG, Hachinski V (1996) Temporal pattern of cognitive decline and incontinence
is different in Alzheimer’s disease and diffuse Lewy body disease. Neurology 46:682–686
48. McKeith I, Mintzer J, Aarsland D et al (2004) Dementia with Lewy bodies. Lancet Neurol
3:19–28
254 I.G. McKeith
Kenji Kosaka
Abstract The history of Parkinson disease (PD) and dementia with Lewy bodies
(DLB) is briefly presented. Since Lewy reported Lewy bodies in the brainstem
nuclei of the PD brain in 1912, Lewy bodies had been considered an essential
pathological finding for the diagnosis of PD. It had been, however, considered that
there were almost no Lewy bodies occurring in the cerebral cortex. In 1976, we
reported our first autopsied case showing numerous Lewy bodies in the cerebral
cortex. In 1978, we reported the detailed characteristics and distribution pattern of
cortical Lewy bodies, based on three autopsied cases showing diffuse Lewy body
disease (DLBD), a term that we proposed in 1984. We also reported two German
autopsied cases showing DLBD in 1979, which were the first DLBD cases reported
in Europe. In 1980, we also proposed the term Lewy body disease and classified it
into three types: brainstem type, transitional type, and diffuse type. The brainstem
type is the same as PD, and the diffuse type was later designated DLBD. In 1990,
we reviewed all the 37 DLBD cases reported in Japan and classified DLBD into
two forms: a common form with more or less Alzheimer pathology and a pure form
without such pathology. Since then, we have reported many papers concerning
DLBD. The term dementia with Lewy bodies (DLB) was proposed at the first
international workshop held in 1995. CDLB guidelines were published in 1996, and
the CDLB guidelines–revised were reported in 2005. In the revised guidelines the
term Lewy body disease was used as a generic term that included DLB, PD, and
PDD, as we had insisted since 1980.
Keywords DementiawithLewybodies(DLB)•DiffuseLewybodydisease
• Lewy body disease • Parkinson disease • Parkinson disease with dementia
(PDD)
K. Kosaka (*)
Yokohama Houyuu Hospital, Yokohama, Japan
e-mail: [email protected]
Introduction
Dementia with Lewy bodies (DLB) is now the second most frequent dementing
illness in the elderly, following Alzheimer-type dementia (ATD). The term DLB
was proposed at the first International Workshop [1], which was held in Newcastle
upon Tyne in 1995. Lewy bodies are essential for the neuropathological diagnosis
of Parkinson’s disease (PD). PD patients frequently show dementia, and such
patients are diagnosed as having PD with dementia (PDD). Recently, it has usually
been considered that DLB and PDD are almost the same disease not only clinically
but also neuropathologically.
In this chapter, the author briefly introduces the history of PD and DLB, and
insists that DLB and PDD should be understood within the spectrum of Lewy body
disease, a term that we proposed in 1980 [2].
James Parkinson [3] published the first report detailing the clinical features of
“shaking palsy” in 1817. Then, he described that cognition remained intact in this
disease. Charcot [4] proposed the term “Parkinson disease” in 1968, and described
cognitive disturbance in PD. In 1912, Lewy [5] reported eosinophilic round intra-
neuronal inclusions in the substantia innominata and dorsal vagal nuclei of PD
brain. Tretiakoff [6] nominated these inclusions “Lewy bodies” and indicated the
importance of the substantia nigra in PD in 1919. Thereafter, the difference of PD
and postencephalitic parkinsonism was discussed for a long time. Hassler [7]
reported the difference in the distribution of neuronal loss in the substantia nigra
between these two diseases in 1938. In 1953, Greenfield and Bosanquet [8] pointed
out for the first time that the presence of Lewy bodies is the essential pathological
finding for the diagnosis of PD whereas the presence of neurofibrillary tangles is
essential for the diagnosis of postencephalitic parkinsonism. Furthermore, den
Hartog Jager and Bethlem [9] described the detailed distribution of Lewy bodies in
the brainstem of PD brain in 1960. Thus, the pathological basis of PD was estab-
lished about one and a half centuries after the first report of Parkinson in 1817. It
had been, however, considered that there were almost no Lewy bodies occurring in
the cerebral cortex. In 1976, we [10] reported our first autopsied case showing
numerous Lewy bodies in the cerebral cortex as well as in the brainstem nuclei. In
1978, we [11] reported the detailed characteristics and distribution pattern of corti-
cal Lewy bodies, based on our own three autopsied cases with “diffuse Lewy body
disease (DLBD),” a term that we [12] proposed in 1984. We [13] also reported two
German autopsied cases showing DLBD in 1979, which were the first DLBD cases
reported in Europe. In 1980, we [2] proposed the term “Lewy body disease” and
classified it into three types: brainstem type, transitional type, and diffuse type. The
brainstem type is the same as PD, and the diffuse type was later designated DLBD
DLB and PDD as Lewy Body Disease 257
[12]. Furthermore, we [14] reviewed all 37 DLBD cases reported in Japan and clas-
sified DLBD into two forms: a common form with more or less Alzheimer pathol-
ogy and a pure form without such findings. Since then, we have published several
papers concerning DLBD. The term “dementia with Lewy bodies” (DLB) was
proposed at the first international workshop in 1995 [1]. The CDLB guidelines
were published in 1996 [15], and the CDLB guidelines–revised were reported in
2005 [16].
The most important recent findings in this field were (1) alpha-synuclein gene
mutation in familial PD [17] and (2) alpha-synuclein as the main component of
Lewy bodies [18]. Thereafter, alpha-synuclein has received considerable attention
in the research on Lewy body disease.
At the first international workshop held in 1995, the difference between DLB and
PDD was discussed. In the CDLB guidelines [15], the “1-year rule” was adapted.
According to these guidelines, PDD is differentiated from DLB by the appearance
of dementia at least 1 year after the onset of PD symptoms. At the third international
workshop held in 2003, not a few researchers insisted that this 1-year rule should
be abolished. This rule was, however, maintained even in the CDLB guidelines–revised
reported in 2005 [16]. Considerable evidence has been reported that DLB and PDD
are almost the same not only clinically but also neuropathologically. Therefore, in
the revised guidelines, the term “Lewy body disease” was used as a generic term
that includes DLB, PD, and PDD [16]. In the report of the DLB and PDD working
group in 2007 [19], a similar description was also introduced.
Since our proposal of Lewy body disease in 1980 [2], we have insisted that
Lewy body disease is a generic term that includes PD, PDD, and DLBD [12, 14,
20–22].
Lewy body disease is now defined as follows: “Lewy body disease is a chronic
neuropsychiatric disease characterized clinically by idiopathic parkinsonism of
early- or late-onset, frequently followed by progressive dementia. In many cases,
progressive dementia is the main symptom, followed frequently by idiopathic
parkinsonism. It is neuropathologically characterized by the presence of numerous
Lewy bodies and Lewy neurites in the central and sympathetic nervous systems.”
As indicated above, we classified DLBD into two forms: a common form with
more or less ATD pathology and a pure form without it [14]. Then, we described
differences in the clinical features between the common form and the pure form. In
the common form, most cases showed later onset, and the initial symptom was
memory disturbance followed by progressive dementia, and about 70% of patients
later developed parkinsonism. However, the pure form was characterized by younger
onset, idiopathic parkinsonism as the initial symptom and later followed by progres-
sive dementia. Therefore, the pure form can be diagnosed as PDD. In 1992, when
the 150th anniversary congress of the German Psychiatry Association was held
258 K. Kosaka
References
1. Perry R, McKeith I, Perry E (1996) Dementia with Lewy bodies. Cambridge University Press,
Cambridge
2. Kosaka K, Matsushita M, Oyanagi S, Mehraein P (1980) A clinicopathological study of Lewy
body disease. Psychiat Neurol Jap 82:292–311
3. Parkinson J (1817) An essay on the shaking palsy. Sherwood, Neely and Jones, Wittingham
and Rowland, London
4. Charcot JM (1892) De la paralysie agitante Euvures completes, 6th edn. Progres Medicale, Paris
5. Lewy FH (1912) Paralysis agitans. I. Pathologische anatomie. In: Lewandowsky M (ed)
Handbuch der neurologie, vol 3. Berlin, Springer, pp 920–958
6. Tretiakoff C (1919) Contribution a l’etude de l’Anatomie pathologique du Locus Niger de
Soemmerung avec quelques deductions relatives a la pathogenie des troubles du tonus mus-
culaire et de la Parkinson. These de Paris
7. Hassler R (1938) Zur pathologie der paralysis agitans und des postenzephalitischen parkin-
sonismus. J Psychol Neurol 48:387–476
8. Greenfield JG, Bosanquet FD (1953) The brain-stem lesions in Parkinsonism. J Neurol
Neurosurg Psychiatry 16:213–226
9. Den Hartog Jager WA, Bethlem JU (1960) The distribution of Lewy bodies in the central and
autonomic nervous systems in idiopathic paralysis agitans. J Neurol Neurosurg Psychiatry
23:283–290
10. Kosaka K, Oyanagi S, Matsushita M et al (1976) Presenile dementia with Alzheimer-, Pick-
and Lewy body changes. Acta Neuropathol 36:221–233
DLB and PDD as Lewy Body Disease 259
11. Kosaka K (1978) Lewy bodies in cerebral cortex; report of three cases. Acta Neuropathol
42:127–134
12. Kosaka K, Yoshimura M, Ikeda K, Budka H (1984) Diffuse type of Lewy body disease. A
progressive dementia with numerous cortical Lewy bodies and senile changes of various
degree. A new disease? Clin Neuropathol 3:185–192
13. Kosaka K, Mehraein P (1979) Dementia-Parkinsonism syndrome with numerous Lewy bodies
and senile plaques in cerebral cortex. Arch Psychiatr Nervenkr 226:241–250
14. Kosaka K (1990) Diffuse Lewy body disease in Japan. J Neurol 237:197–204
15. McKeith I, Galasko D, Kosaka K et al (1996) Consensus guidelines for the clinical and patho-
logical diagnosis of dementia with Lewy bodies (DLB). Neurology 47:1113–1124
16. McKeith IG, Dickson DW, Lowe J et al (2005) Diagnosis and management of dementia with
Lewy bodies: third report of the DLB consortium. Neurology 65:1863–1872
17. Polymeropoulos MH, Lavedan C, Leroy E et al (1997) Mutation in the a-synuclein gene
identified in families with Parkinson’s disease. Science 276:2045–2047
18. Spillantini MG, Schmidt ML, Lee VMY et al (1997) a-Synuclein in Lewy bodies. Nature
(Lond) 388:839–840
19. Lippa CF, Duda JE, Grossman M et al (2007) DLB and PDD boundary issues. Diagnosis,
treatment, molecular pathology, and biomarkers. Neurology 68:812–818
20. Kosaka K, Iseki E (1996) Diffuse Lewy body disease within the spectrum of Lewy body
disease. In: Peery E, McKeith E, Perry E (eds) Dementia with Lewy bodies. Cambridge
University Press, Cambridge
21. Kosaka K, Iseki E, Odawara T et al (1996) Cerebral type of Lewy body disease.
Neuropathology 17:32–35
22. Kosaka K (2000) Diffuse Lewy body disease. Neuropathology 20 (suppl):73–78
23. Kosaka K (1992) Diffuse Lewy Koerperchen: vergleich klinisch- pathologischer Daten
zwischen Japanischen und Europaeischen/Amerikanischen Faellen. 150. Jahren Psychiatrie in
Deutschland, Koeln
24. Kosaka K (2004) Dementia with Lewy bodies and Parkinson’s disease with dementia. Do
most of the demented PD patients have DLBD? The 45th Annual Meeting of Japan Neurology
Association, Tokyo (panel discussion)
25. Braak H, Del Trediel K, Rub U et al (2003) Staging of brain pathology related to sporadic
Parkinson’s disease. Neurobiol Aging 24:197–211
Clinicopathological Characterization
of Frontotemporal Lobar Degeneration
Yoshio Mitsuyama
Keywords Frontotemporallobardementia•Motorneurondisease•Pickdisease
Introduction
Y. Mitsuyama (*)
Psychogeriatric Center of Daigo Hospital, 1270 Nagata, Mimata-cho,
Miyazaki 889-1911, Japan
e-mail: [email protected]
In the 1980s and 1990s, several research groups recognized that there were
many patients with clinical features similar to those of patients with Pick disease
who had neither the classical lobar atrophy nor characteristic neuronal changes.
These disorders came to be known by several different names, including FTLD-
LDH, which consists of frontotemporal lobar degeneration lacking distinct histol-
ogy, Pick disease, and FTLD with motor neuron disease (FTLD-MND), primary
progressive aphasia (PPA), and semantic dementia (SD) [3–10]. As expected, all
variants of FTLD have neuronal loss and gliosis affecting the frontal and temporal
cortices in keeping with a diagnosis of FTLD. The research criteria have been
focused on the need to have subgroups of patients for study during life and accurate
pathological diagnosis on autopsy.
Clinical criteria for diagnosing FTLD include the Lund and Manchester Criteria
and the more recent consensus criteria [2, 11, 12]. Patients with FTLD present
gradual and progressive changes in behavior, or gradual and progressive language
dysfunctions. The most common psychiatric symptom of FTLD is an early change
in social and personal conduct, characterized by difficulty in modulating behavior
to the social demands of a situation. FTLD patients are impaired in the regulation of
conduct. This dysfunction is often associated with a lack of inhibition, resembling
in impulsive or inappropriate behavior. Progression of the disease may lead to poor
financial judgement, and compulsive-like behaviors are common presenting symptoms
among FTLD patients [12, 13]. Complex compulsive acts may result from temporal
lobe involvement [14]. In some individuals, inappropriate sexual behavior occurs.
Perseverative and stereotyped behaviors encompass simple repetitive acts and
verbal expression or stereotypies such as lip smacking, habitual hand rubbing or
clapping, and humming that may result from frontostriatal circuit dysfunction or
involvement of the caudate nuclei. Psychotic symptoms such as delusions and
hallucinations are uncommon in FTLD. Nevertheless, there have been patients with
an initial schizophrenia-like psychosis or a psychotic affective disorder such as a
sign of FTLD [15, 16]. Dietary habits and personal hygiene may also change.
Patients become inactive with decreased behavioral motivation and spontaneity,
loss of interest in personal hygiene with failure to wash, bathe, groom, and so on.
In FTLD, fragments of the Klüver–Bucy syndrome can occur, particularly the
hyperorality that manifests as cramming and bingeing, altered food performance
especially for sweets or food fads. They may attempt to eat inedible items. FTLD
patients can be so hyperoral that require restraint to prevent suffocation or
aspiration. There is a loss of concern for one’s personal appearance, and patients
may be increasingly unkempt early in the disease. Patients show loss of personal
concern for their actions.
FTLD patients tend toward decreased verbal output progressively to complete
mutism. Early language disturbances of FTLD are empty speech, nonfluent anomia,
especially for words connecting action, and semantic anomia where the word loses
its meaning. They usually do not have a true amnestic syndrome. In the FTLD,
spatial difficulties are seen in patients with mild or moderate impairments. Many of
these patients present with troubles in the expression of language, problems using
the correct words, including the naming of persons and things, or expressing
Clinicopathological Characterization of Frontotemporal Lobar Degeneration 263
oneself. Some patients with FTLD have a progressive aphasia several years before
other clinical manifestations; in such cases, speech and language changes predomi-
nate (PPA and SD) [17].
FTLD patients have relatively preserved visuo-spatial abilities such as spatial local-
ization and orientation in familiar surroundings. FTLD patients lack sympathy, empa-
thy, emotional warmth, or awareness of the needs of others and appear emotionally
shallow and indifferent. FTLD patients have deficits of insight, abstraction, planning,
and problem solving, in keeping with a frontal “dysexecutive” syndrome. Judgement
is abnormal. FTLD patients are often very concrete on proverb interpretation and tests
assessing comprehension of similarities and differences. On neuropsychological test-
ing, frontal-executive functions are compromised early in FTLD, and most memory
and occipitoparietal functions are compromised early in most Alzheimer patients.
In the early and middle stages, neurological signs are usually absent or confined
to the presence of primitive reflexes such as grasp, snout, and sucking reflexes.
Dystonia, ideomotor apraxia, dysphagia, or fasciculations and muscle wasting are
occasionally observed.
There are no laboratory findings pathognomonic of FTLD. Routine investigations
of blood and urine yield unremarkable results. Conventional EEGs tends to remain
normal until late in the course, when they show diffuse slowing and occasional
focal frontal or temporal slow wave activity. Structural neuroimaging, particularly
magnetic resonance imaging (MRI), can help confirmation in the presence of
FTLD. Most FTLD patients show frontal (and anterior temporal) atrophy,
enlargement of the Sylvius fissures, anterior callosal atrophy, and eventual
hippocampal and entorhinal volume loss. Computed tomography (CT)/MRI may
show atrophy of the anterior temporal and frontal lobes. Some FTLD patients may
have additional MRI evidence of bilateral caudate nuclear atrophy. Functional
imaging is more sensitive than structural imaging for the diagnosis of FTLD.
Single-photon emission computed tomography/18F-fluorodeoxyglucose-positron
emission tomography (SPECT/FDG-PET) typically demonstrates decreased
perfusion and metabolism of the frontal and temporal lobes.
The usual clinical duration of FTLD is about 8–11 years. The clinical course of
FTLD can be divided into three stages. In the initial stages, there are prominent
personality changes, emotional alterations, and impaired insight and judgement.
Speech and language changes also may occur. In the second stage, aphasia and
other cognitive changes become pronounced, but there is at least partial preservation
of memory, visuo-spatial skills, and computational ability. The third and final stage
of the disease is often dominated by progressive muteness, and the patients become
profoundly demented.
At autopsy, the brains of patients with FTLD show a lobar distribution of
atrophy involving the frontal lobes, temporal lobes, or both. Coronal sections reveal
deep sulci and may show knife-edged gyri in the atrophic area, especially in Pick
disease. The orbitofrontal cortex and the anterior and medial temporal areas show
the most severe atrophic changes. The predominant neuropathological abnormalities
are frontotemporal neuronal loss and gliosis with ubiquitin-positive, tau-negative
inclusions and without detectable amounts of insoluble tau, with MND or without
264 Y. Mitsuyama
MND. The cortical degeneration involves mainly the grey matter, including the
insula and the anterior cingulate gyrus. In the past, clinicians have referred to FTLD
patients as having “Pick bodies,” but on neuropathological examination, most
FTLD patients lack the pathognomonic Pick bodies. There has been continuing
controversy concerning whether identifiable cellular changes of Pick bodies and
ballooned neurons are a requirement for diagnosis. Smaller number of FTLD
patients have involvement of substantia nigra, striato-pallidum, parietal cortex,
thalamus, and other structures [18, 19]. Most FTLD patients do not have senile
plaques or neurofibrillary tangles, although some have amyloid beta deposition,
particularly late in the course and in patients with an APO-E epsilon 4-allele [20].
In FTLD, about one-third of patients have tau pathology and about 10% have a tau
gene mutation [21, 22]. Pathological findings have, to date, not been associated
with specific clinical manifestations.
Pick Disease
Arnold Pick [23–25], in a series of articles based on gross examination of the brain,
reported cases of dementia with severe circumscribed atrophy of the frontotemporal
regions. The definition of the clinical entity of what became known as Pick disease
was presented in a series of articles in the 1920s [26–28]. The initial stage is char-
acterized by prominent personality changes and emotional alterations. Judgment is
impaired early, and insight is compromised. Social behavior deteriorates, and lan-
guage abnormalities are among the earliest intellectual alterations to occur. In the
second stage of disease, deterioration of mental status becomes evident and aphasia
is more prominent. Cognitive changes are more pronounced, but memory and
visuo-spatial skills may remain relatively intact. In the final stage of the disease,
progressive extrapyramidal syndrome usually appears and the patient becomes
mute and incontinent. Pick disease has a longer illness duration (more than 10
years). CT/MRI may provide supportive evidence for the diagnosis.
Neuropathology shows severe frontotemporal atrophy, often with “knife-
edged” gyri (Fig. 1), and extensive neuronal loss with gliosis. An abrupt transi-
tion is sometimes evident between involved and uninvolved cortical regions.
There is a tendency for selective sparing of the precentral gyrus and the posterior
one-third of the superior temporal gyrus. The concept of Pick disease emphasized
the importance of Pick bodies and ballooned neurons in the pathological diagno-
sis and the clinical distinction of this disorder from Alzheimer disease. The diag-
nosis of Pick disease occurs when there are Pick bodies with or without Pick
cells. Pick bodies are spherical, silver-staining (argyrophilic), ubiquitin- and tau-
positive intraneuronal inclusions. Pick bodies are concentrated in frontotemporal
neocortical layers II–VI, and the hippocampal formation in the granular layer of
the dentate gyrus and sector CA1. Pick bodies do not occur in normal aging.
There are pathognomonic microscopic findings showing ballooned neurons or
Pick cells and definite positive tau and ubiquitin bodies in neurons of the
Clinicopathological Characterization of Frontotemporal Lobar Degeneration 265
FTLD-LDH
FTLD is a clinical term applied to patients who present with progressive dementia
with an insidious onset, prominent behavioral or language dysfunction, or both. We
have recognized that this term includes groups of patients whose pathological con-
ditions and genetic mechanisms are heterogeneous. The clinical characteristics of
FTLD-LDH are almost similar to those of Pick disease. However, the severity of
intellectual and personality deterioration is less than those seen in Pick disease.
MRI shows frontotemporal atrophy, and SPECT shows selective hypoperfusion of
frontotemporal lobes. The most common pathology of FTLD-LDH is a nonspecific
frontotemporal atrophy, and FTLD-LDH is the usual FTLD pathology.
Microscopic study shows mild to slight neuronal loss and astrogliosis with
sponginess (minute cavities or microvacuolation) of the outer supragranular (II–III)
layers of the frontotemporal cortex with ubiquitin- and TDP-43-positive inclusions
266 Y. Mitsuyama
(Fig. 2). There is also variable involvement of subcortical and limbic structures.
The anterior hippocampal regions are also affected. FTLD-LDH has no Pick bod-
ies, plus depigmentation in the substantia nigra and striato-pallidum.
FTLD-MND
A subset of patients with FTLD develops symptoms suggestive of MND. The mean
age at onset and disease duration are 52 years and 2.3 years, respectively. FTLD-
MND is a clinicopathological entity characterized by the combination of FTLD and
MND. The link between FTLD and MND was suggested by the autopsy in 1984
[29], and the term FTLD-MND was subsequently proposed [30]. The development
of MND in patients presenting with a progressive behavioral disorder would strongly
support a clinical diagnosis of FTLD-MND. MND is also a clinical term, but it is
applied to patients with clinical evidence of corticospinal tract involvement, evi-
dence of brainstem or spinal cord anterior horn cell involvement, or both. FTLD-
MND have symptoms of mild forgetfulness and language output impairment, in
addition to the more prominent behavioral disorders. These symptoms include
weakness and muscle wasting. Symptoms of parkinsonism, such as rigidity, are
occasionally seen. Signs of MND may not always be present early. Most of the cases
of MND developed approximately 6–24 months after the onset of symptoms of
FTLD [29, 31]. Only features of dementia are noted early in the disease course, and
both initially carried a diagnosis of FTLD. MRI/SPECT reveals slight to moderate
atrophy and selective hypoperfusion in the frontotemporal regions. The patient
shows a relatively rapid clinical course, less than 5 years.
Clinicopathological Characterization of Frontotemporal Lobar Degeneration 267
degeneration does not correlate with clinical signs of MND or with severity of
neuronal loss and gliosis in the hypoglossal nucleus and spinal anterior horn cells.
Extramotor ubiquitin-positive inclusions are absent to sparse in all cortical regions.
Comments
Large clinicopathological series have been published that have clearly demon-
strated an overlap between the clinical syndromes subsumed under the term fronto-
temporal dementia and the progressive supranuclear palsy syndrome, corticobasal
degeneration syndrome, and MND. There have also been significant advancements
in brain imaging, neuropathology, and molecular genetics that have led to different
approaches to the classification.
FTLD and FTLD-MND represents a distinct clinicopathological entity that
shows essentially ubiquitin and TDP-43 proteinopathy. There are some cases of
FTLD with TDP-43-positive and ubiquitin-negative pathology. TDP-43 proteinop-
athy has been reported in many neurodegenerative diseases (filament inclusion
disease). TDP-43 might be more susceptible to neuronal damage. From these find-
ings, different pathoetiologies could lead to the varied clinicopathological presenta-
tions of FTLD.
The pathological features of FTLD-MND are variable. Most cases have a mix-
ture of lower motor neuron degeneration, occasionally associated with corticospi-
nal tract degeneration, similar to progressive spinal muscular atrophy or ALS, and
the majority have Bunina bodies, which are a histological hallmark of ALS.
The field is complicated by a barrage of overlapping clinical syndromes, and
neuropathological diagnosis that does not always respond to clinical presentations
and underlying neuropathology.
It is difficult to distinguish between cases on the basis of extramotor ubiquitin-posi-
tive pathological features or on the basis of predominant involvement of upper or lower
motor neurons. Hippocampal sclerosis with ubiquitin-positive inclusions in the dentate
fascia is a common feature of FTLD with ubiquitin-only immunoreactive changes.
Lower motor neuron involvement including spinal cord anterior horn cell and
hypoglossal nucleus degeneration is of the utmost importance for future clinico-
pathological studies with combined dementia and MND.
It is unclear how neurodegenerative diseases cause dysfunction and death of
brain cells or specific neuropsychiatric symptoms. Clinical manifestations of the
FTLD cannot accurately predict the nature of the underlying neurodegenerative
disease. The neuropathological findings alone cannot establish that a patient had
FTLD in the absence of documented clinical information. Although there is no
specific treatment for FTLD, many symptomatic therapies can be very helpful.
Many of the behavioral psychological symptoms of FTLD may respond to selective
serotonin uptake inhibitors (SSRI) [35]. Marked disinhibition, aggressive behavior,
or verbal outbursts may respond to small doses of major tranquilizers such as ris-
peridone, olanzapine, or quetiapine. FTLD is very stressful to the caregiver, and
support of the family is critically important.
Clinicopathological Characterization of Frontotemporal Lobar Degeneration 269
References
25. Oick A (1905) Zur Symptomatologie der linksseitigen Schlafen-lappenn Atrophie. Monatsschr
Psychiatr Neurol 16:378–388
26. Gans A (1922) Betrachtungen über Art und Ausbreitung des krankhaften Prozesses in einem
Fall von Pickscher Atrophie des Stirnhirns. Z Gesamte Neurol Psychiatr 80:10–28
27. Onari K, Spatz H (1926) Anatomische Beitrage zur Lehre von Pickschen umschriebenen
Grosshirnrinden-Atophie (“Picksche Krankheit”). Z Gesamte Neurol Psychiatr 101:470–511
28. Schneider C (1927) Über Picksche Krankheit. Monatsschr Psychiatr Neurol 65:230–275
29. Mitsuyama Y (1984) Presenile dementia with motor neuron disease in Japan: clinico-
pathological review of 26 cases. J Neurol Neurosurg Psychiatry 47:53–959
30. Neary D, Snowden JS, Mann DM et al (1990) Frontal lobe dementia and motor neuron dis-
ease. J Neurol Neurosurg Psychiatry 53:23–32
31. Mitsuyama Y (2000) Dementia with motor neuron disease. Neuropathology 20(Suppl):S79–S81
32. Mendez MF, Selwood A, Mastri AR et al (1993) Pick’s disease versus Alzheimer’s disease: a
comparison of clinical characteristics. Neurology 43:289–292
33. Schmitt HP, Yang Y, Fortstl H (1995) Frontal lobe degeneration of non-Alzheimer type and
Pick’s atrophy: lumping or splitting? Eur Arch Psychiatry Clin Neurosci 245:299–305
34. Josephs KA, Parisi JE, Knopman DS et al (2006) Clinically undetected motor neuron disease
in pathologically proven frontotemporal lobar degeneration with motor neuron disease. Arch
Neurol 63:506–512
35. Swartz JR, Miller BL, Lesser IM et al (2001) Frontotemporal dementia: treatment response to
serotonin selective reuptake inhibitors. J Clin Psychiatry 56:41–45
Diffuse Neurofibrillary Tangles
with Calcification
Keywords Dementia•DNTC•Neurofibrillarytangles•Calcification
Introduction
Case Presentation
Fig. 1 Brain computed tomography (CT) at the age of 74 (case 21). Cerebral atrophy of temporal
and frontal lobes and calcification in the basal ganglia and cerebellum
changes. Microscopically, neuronal loss and gliosis in the atrophic foci of the
cortices were observed in the temporal pole in particular. The amygdala,
hippocampus, and temporal lobe showed marked gliosis, and the white matter of the
frontal and occipital lobed had slight to moderate gliosis. Numerous NFTs were
observed in the cerebral cortex involving the frontal to occipital lobes. NFTs
contained both 3 repeat and 4 repeat tau. A few SPs were detected in the temporal
cortex; however, they were absent in other areas. The caudate nucleus showed atrophy,
and there were only a small number of NFTs and neuropil threads. There were a few
NFTs in the neurons of the substantia nigra and locus ceruleus. Calcification was
present in the pallidum, putamen, granular layer, and the dentate nucleus of the
cerebellum.
Case 2. A 48-year-old woman. There was no family history of dementia. At the
age of 45, she became unable to do housework. Memory disturbance and
disorientation gradually progressed, requiring caregiving for meals and excretion.
Neurologically, there were no abnormal findings other than dementia. The blood
pressure was normal. Personal contacts were maintained. Euphoria was noted.
Neither aggression nor negativism was observed. Wandering and yelling made keep-
ing her at home difficult, and the patient was admitted to a psychiatric hospital.
Dementia progressed, and she died of pneumonia, with a dementia follow-up of
3 years. Autopsy was performed. The brain weighed 1,000 g. The frontal lobes
showed atrophy bilaterally. On the cut surface, temporal lobe atrophy was noted. In
274 S. Kuroda et al.
the temporal lobe, the myelin sheath was markedly lightened, and gliosis was
observed at the same site. In the frontal lobe, similar changes were also noted,
although the grade was lower. Numerous NFTs were observed in the amygdala,
hippocampus and inferior temporal gyrus. Calcification was observed in the vascular
wall and parenchyma of the basal ganglia and granular layer of the cerebellar vermian
cortex. In the substantia nigra, a small number of NFTs and Lewy bodies were present.
A SP-like structure was observed, but amyloid staining showed no amyloid
deposition in this structure.
Epidemiology of DNTC
Clinical and pathological reports of more than 30 patients with DNTC have been
reported in Japan. However, in Western countries, not much interest in this disorder
has been shown. Calcification was observed in the basal ganglia, dentate nucleus of
the cerebellum, and cerebral white matter in severe cases (Fahr type). Brain CT
scanning facilitates detection of atrophy and calcification at these sites. In our sur-
vey using the criteria proposed by Kosaka [4], DNTC was suspected in 4 of 3,053
patients (male:female = 1:2) with dementia residing in Okayama Prefecture. All
these cases were female. In one case, autopsy led to a definitive diagnosis of
DNTC. The incidence of DNTC was 0.13% in patients with dementia in Okayama.
However, it may be lower in the general population. In Japan, several patients have
been reported in Tokyo, Nagoya, Okayama, and Kochi, suggesting regional differ-
ences. In 21 autopsied patients, the male-to-female ratio was 1:3. Familial DNTC
has not been reported. The mean age at onset was 53.1 years. A presenile onset
(40–69 years of age) was frequent. The mean time of illness duration was 10.5
years (range, 3–24 years). In patients without complications, the clinical course
may be prolonged. Generally speaking, the longer the duration, the lower the brain
weight. The brain weight had decreased to approximately 800 g in patients with a
prolonged course. In some patients, however, cerebral atrophy was evident even in
the early stage.
Pathological Characteristics
NFTs
distributed in the cerebral isocortex. They were also found in Meynert’s nucleus
and the locus ceruleus/raphe nuclei of the brainstem. These sites were similar
to those frequently observed in patients with AD. On the other hand, there were no
NFTs in the basal ganglia or cerebellum. Differing from AD, SPs were absent, or
only a small number were detected.
There are six isoforms of tau protein. Among these, tau protein involving exon-10,
which exists in the microtubule-binding site, is termed “4-repeat tau protein,” and
exon-10-free tau protein is called “3-repeat tau protein.” In patients with AD, all
six isoforms are detected. In those with progressive supranuclear palsy or cortical
basal ganglia degeneration, 4-repeat tau protein is found. In those with Pick body
disease, 3-repeat tau protein is present. In patients with DNTC, 60-, 64-, and
68-kDa major and 72-kDa minor bands were recognized in the presence of various
anti-tau antibodies. On Western blotting of sarkosyl-insoluble tau protein after
dephosphorylation, six isoforms of tau protein were observed [6], indicating that
the biochemical features of degenerative tau protein in patients with DNTC were
similar to those in AD.
Calcification
Cerebrovascular Lesions
Conclusion
References
1. Ando J, Okaniwa T, Tachibana K (1965) An autopsy case of Pick’s disease. Shinkei Kenkyu-no-
Shinpo 9:181–182 (In Japanese)
2. Shibayama H, Kobayashi H, Nagasawa MI et al (1992) Non-Alzheimer non-Pick dementia
with Fahr’s syndrome. Clin Neuropathol 11:237–250
3. Kosaka K (1994) Diffuse neurofibrillary tangles with calcification: a new presenile dementia.
J Neurol Neurosurg Psychiatry 57:594–596
278 S. Kuroda et al.
Eduardo Gastelumendi
Keywords Ayahuasca•Consciousness•Depression•Ethnopsychiatry•Psychotropic
Introduction
Brazil, Bolivia, Colombia, Ecuador, the Guyanas, Peru, and Venezuela share the
Amazon rainforest. Hundreds of different tribes inhabit this huge area. At least 70
of those indigenous groups, primarily in the Upper Amazon and Orinoco basins,
E.Gastelumendi(*)
PeruvianPsychiatricSociety,Ave.AngamosOeste387/203,Miraflores,Lima18,Perú
and
PeruvianPsychoanalyticSociety,JulioBecerra235,Miraflores,Lima18,Perú
and
InternationalCommitteeoftheINA,INASecretariatOffice,NeuropsychiatricInstitute,
EuroaCentre,ThePrinceofWalesHospital,Randwick,NSW2031,Australia
and
CalleParqueArmendariz119/4-E,Miraflores,Lima18,Perú
e-mail: [email protected]
have used for millennia the blend known as ayahuasca for divination, healing, and
other cosmogonic and shamanic purposes [1]. In its original milieu, the ritual,
which has as its main vehicle the ayahuasca, with strong psychotropic activity in
virtue of its serotoninergic properties, allows the participants to contact the “other”
reality, or the “real aspect of the world,” where the spirits of plants, animals, and
dead people exist. It is also considered as a preparation for life after death [2]. The
shaman of the tribe usually conducts the rite. The ingestion of ayahuasca produces
intense visions. At first these are of geometric patterns (which can be seen as part
of the Amazon people’s crafts, in clothes, paintings, and pottery), and then the
visions become gradually more complex, with emotional, cognitive, and somatic
components, as we will see later.
The ayahuasca brew is a blend of two different plants cooked together. Ayahuasca
proper (Baninsteropsis caapi) is a vine native to the Amazon – currently it grows in
different parts of the world – that contains beta-carbolines such as harmine, harmaline,
and tetrahydroharmine, all of which are potent monoamine oxidase (MAO) inhibitors.
The other plant is the Chacruna (Psychoatria viridis), a bush that contains the
tryptamine alkaloid N,N-dimethyltriptamine (DMT). DMT is not orally active itself,
because it is inactivated by peripheral MAO of the liver and gut.
It is the blend that works: the beta-carbolines present in the ayahuasca vine
inhibittheMAOandallowstheDMTtoentertheinternalmilieu.Thequestionof
how the indigenous people found the right combination remains unanswered, and
this is definitely a result of many years of investigation and combinations. Sometimes
the brew may include other plants, as tobacco, Toé (Brugmansia suaveolens), or
coca leaves.
In the last decades, this ceremony has come out from the rainforest and has
made its appearance in the main cities of many countries not only of South
America but abroad. Conducted also by a leader – a shaman, Maestro, or
Curandero – the aim is to have an inner experience with intense visions,
somatic and emotional effects, and occasional key insights. During the ritual, in
both Amazon and urban settings, the participants drink the brew from a cup
(50–100 ml) and sit in silence waiting for the effects of the substance to occur.
In an urban setting, the conductor has previously evaluated the participants, by
overtquestionsorsubtleobservation,todeterminetheamountofbreweachone
will have, if any. This evaluation is important to reduce the risks of a bad expe-
rience for the participant for reasons of psychic frailties or other health condi-
tions. In not rare occasions the shaman decides not to give the brew to a
participant.
This ritual would be of little importance to our field, or perhaps be taken as a
folkloric and curious issue, if it were not that some of our patients in psychotherapy
have participated in one or more of these ayahuasca sessions and have talked about
their experiences in their psychoanalysis and psychotherapies.
For historical and affective reasons, I would like to mention Carlos Alberto
Seguin, who in the decade of 1960 was among the first psychiatrists to study the
brewinPeru.Thesestudiesvaryfromaphenomenologicalapproachtoethnographic
psychiatry.
Ayahuasca:CurrentInterestinanAncientRitual 283
At the same time, neuropsychiatric research studies are been made on the effect
of ayahuasca in refractory depression [3] and on its effects in visualization,
memory, and consciousness [4].
I would like to suggest that the ayahuasca experience, because of its peculiarities,
hasauniquevalueforresearchinbothneuralandmentalfields.Letmesharewith
you a glimpse of what possibilities are open to research and theorization.
The ceremony usually takes place at night. The participants and the shaman sit in
a circle. The shaman gives a small amount of the brew to each person and then
asks all to sit in silence. Shortly, about half an hour later, the visions begin. At first,
they are colorful geometric patterns, and then gradually become more complex.
Songs sung by the shaman during the session lead the participants to different
visions, to changes in mood and in the level and quality of consciousness. The
participants describe themselves as entering another state of mind, with keen
awareness of the body, thoughts, and inner images. Very elaborate images may
also appear.1
During the 4 or 5 hours that the experience lasts, old memories may appear, many
of them painful and formerly repressed, as well as images of plants, animals,
landscapes, and buildings. A numinous feeling or sometimes a feeling of awe may
occur. Issues regarding the meaning and direction we give to our lives are not rare.
The shaman closes the ceremony with a brief rite as the participants gradually
come back to a “normal” state. The following day there is a feeling of renovation.
Insights may still be very clear.
It is interesting to consider that in Brazil there are two organized religions that
have the ayahuasca as the core of their inner experience and their beliefs. Medical
membersofoneofthesegroups,theUniãodoVegetal(www.udv.org.br; the other
one is the “Igreja de Santo Daime”), have participated in research and publication
of their data [5].AlsoinBrazil,intheUniversityofRibeirãoPreto,researchhas
been conducted on animals as well as in treating refractory depression in humans
in a strict medical setting [3]. Brazilian researchers have also published studies on
the nature of the visual imagery so vividly enhanced in the participants, using
functional magnetic resonance imaging (fMRI) [4].
From the psychodynamic perspective, what our patients bring to their sessions
isveryimpressiveandrequiresanopenattitudeonourpartastherapiststoavoid
considering the experience as mainly an acting-out or a risk-seeking attitude.
1
There are many artists that have depicted their visual experiences, with more or less skill.
AmongthemostinterestingofthemisPabloAmaringo(Peru,1943–2009),founderoftheArt
schoolUsko-Ayar,inPucallpa.
284 E.Gastelumendi
Let me share with you two experiences of patients who brought their ayahuasca
experience to their psychotherapeutic sessions.
In the first vignette, a woman in her sixties comes to analytical therapy because
she still feels somatic and psychic pains after a car accident suffered years ago. In
that accident, her teenaged nephew was killed and she was seriously injured.
Whenshecametoanalyticaltherapy,2yearsaftertheaccident,shewassomewhat
recovered, but still felt deep sadness, guilt, and constant pain in her head, side, and
legs. One of the issues that appeared during our work was the sadomasochistic
bond with her very old but still lucid mother, who lived nearby. It was in the sec-
ondyearoftherapywhenshedecidedtoparticipateinanayahuascasession.Even
though some symptoms had become lighter (less guilt and a better disposition
toward life), the pain in her body remained, and the relationship with her mother
and daughters was still complicated. She talked for some sessions about her inter-
est in drinking the brew and finally decided to participate in one group session
with a shaman.
This is a summary of her experience:
After an hour or so after she drank the brew, the visions started and she entered into a calm
mood and heightened sense of awareness. After a while she found herself directing her
attention to her body and started exploring every part of it inch by inch. She felt she could
distinguish the different kinds of pain and, at the same time, the distinct emotions “tied” to
each part. She cried very deeply. After a while a sort of oneiric vision appeared: she saw
herself sitting in a chair in a dark room, with her mother behind her, and her grandmother
further behind. She could almost guess (or see?) the mother’s grandmother further back. In
frontofher,herdaughtersweresitting.Allthesewomenformedasilentandquietrow.In
the same way that my patient had been able to observe every detail of her sore body, now
she could see the expressions and emotions of the women of her family. She noticed there
was a common pain that passed through all of them, a pain related to a way of being a
woman that had been transmitted through generations. Suddenly she had an insight: she
recognized the same style of hardening the hearts and controlling the emotions shared by
all the women in her family. They had lived doomed, so to say, to live that way.
Whathappenedafterherayahuascaceremonywasasurpriseforus.Inaverynaturalway,
the next time she approached her mother, some days after the ceremony, her attitude was
quitedifferent.Whentheymet,shelistenedtothequerulousoldwomanwithher“heart
opened,” and then she hugged her with a tenderness she could not remember feeling or
having with her. This was the beginning of a new relationship, more kind and affectionate,
that lasted until her mother passed away. The relationship with her daughters also gained
new life and dimensions, as my patient felt free to express more openly her emotions and
to tolerate, and appreciate, the daughters’ own characteristics and decisions.
There are also other kinds of insights produced by the experience, as the
following vignette can show:
A man in his late thirties comes for reanalysis. Some years before he had started
participating in ayahuasca sessions in Lima and later went to the Amazon. He had his first
ayahuasca experience moved by curiosity. He thought he would “enjoy” a psychedelic trip
but found something else. Some of the visions he had were in a way similar to the one
already described. These visions consisted in seeing himself in relationship with people he
Ayahuasca:CurrentInterestinanAncientRitual 285
loved, and he felt he could understand the feelings of each one of them. He also saw himself
as a baby and mourned deeply – he felt it was the first time – for the loss of his grandfather,
which had occurred when he was 3 years old. He was also able to remember his childhood
and adolescent dreams, what he wanted for his life, and could see where he was now
professionally and personally. From this perspective he felt he could see what decisions
and changes he had to make and take in his present life to redirect it in consistency with
his desires, which also became clearer to his eyes. These could be considered important
biographical visions and insights.
But during the sessions he also had another kind of experience: he sensed very strongly
and deeply something he had known intuitively and by his studies and work: being an
ecology activist, he was convinced that the only path to a better life in society passes
through the development of a new way of relating to nature and to others. During the most
intense part of the experience, between the second and third hour, the patient had a sense
of awe of being alive. He felt he could perceive emotionally, not only understand rationally,
the interconnectedness of all things. He could feel that the air he inhaled had been just
exhaled by the surrounding trees, and that the moon in the sky and his retina receiving that
light were intimately connected. And that he shared an intimate “brotherhood” with all
forms of existence. He came out from the experience with a sense of profound gratitude.
Conclusions
References
1. Luna LE, White SF (eds) (2000) Ayahuasca reader: encounters with the Amazon’s sacred
vine.Synergetic,NewMexico
2. LuzP(2004)TheAmerican-Indianuseofcaapi(Ousoameríndiodocaapi).In:LabateBC,
AraújoWS(eds)Theritualuseofayahuasca(Ousoritualdaayahuasca).MercadodasLetras,
Campinas,SãoPaulo
3. FortunatoJ,RéuxG,KirschTetal(2009)Acuteharmineadministrationinducesantidepres-
sive-likeeffectsandincreasesBDNFlevelsintherathippocampus.ProgNeuropsychopharmacol
BiolPsychiatry33(8):1425–1430
4. DeAraujoDB,RibeiroSTG,CecchiGetal(2009)Neuralbasisofenhancedvisualimagery
following Ayahuasca ingestion (Manuscript submitted for publication)
5. LabateBC,AraújoWS(eds)(2004)Theritualuseofayahuasca(Ousoritualdaayahuasca).
MercadodasLetras,Campinas,SãoPaulo
Additional Reading
6. AlmendroM(2008)Chamanismo:Lavíadelamentenativa.EditorialKairos,Barcelona
7. ChiappeM(1974)Curanderismo:Psiquiatríafolklóricaperuana.Tesisparaobtenereldoc-
toradoenmedicinaparalaUniversidadNacionalMayordeSanMarcos,Perú
8. ChiappeM,LemlijM,MillonesL(1985)AlucinógenosyshamanismoenelPerúcontem-
poráteno.EdicionesElVirrey,Lima
9. GastelumendiE(2001)MadreayahuascayEdipo.In:MabitJ(ed)MemoriadelSegundoforo
sobreEspiritualidadIndígena:ética,malytransgresion.CISEI,Lima
10. NarbyJ(1998)Thecosmicserpent:DNAandtheoriginsofknowledge.JeremyP.Tarcher/
Putnam,NewYork
11. SeguínCA(1988)Medicinastradicionalesymedicinafolklórica.FondoeditorialdelBanco
CentraldeReservadelPerú,Lima
12. SeguínCA(1979)Psiquiatríafolklórica:shamanesycuranderos.EdicionesErmar,Lima
13. DobkindeRiosM,GrobCh(2005)ThemeEds.AyahuascaUseinCross-CulturalPerspective.
JournalofPsychoactiveDrugs37(2).Haight-AshburyPublications.SanFrancisco
14. Shanon B (2002) The antipodes of the mind: charting the phenomenology of the ayahuasca
experience.OxfordUniversityPress,NewYork
The Molecular Genetics of Suicide
Abstract The increasing number of suicidal victims all over the world is a major
concern. There are three neurobiological systems involved in the pathophysiology
of suicidal behavior: dysfunction of the serotonergic system, hyperactivity of the
noradrenergic system, and increased activity of the hypothalamic–pituitary–adrenal
(HPA) axis appear to be involved. Increasing evidence points to an overlap between
neurobiological and cognitive psychological approaches to understanding suicidal
behavior. The authors reviewed the molecular genetics of suicidal behavior. A better
understanding of the neurobiology of suicide can help detect at-risk populations
and help develop better treatment interventions. Because suicide continues to be a
major public health problem, further studies are necessary, including research on
the effects of combined medical and psychosocial approaches.
Keywords Dopamine•Moleculargenetics•Neurobiology•Noradrenalin•Serotonin
•Suicide
Introduction
K. Maeda (*)
Department of Occupational Therapy, Kobe Gakuin University School of Rehabilitation,
Ikawadani-cho518,Nishi-ku,Kobe651-2180,Japan
e-mail: [email protected]
O.ShirakawaandN.Nishiguchi
DepartmentofNeuropsychiatry,KinkiUniversitySchoolofMedicine,
7-5-2,Kusunoki-Cho,Chuo-Ku,Kobe651-0017,Japan
M. Fukutake
Department of Psychiatry, Kobe University School of Medicine,
7-5-2,Kusunoki-Cho,Chuo-Ku,Kobe651-0017,Japan
this number is the fourth highest in the world. Suicide is one of the most
important public concerns and a major cause of death among young people
throughout the world. No remarkable reduction in the number of suicide
victims has yet been achieved, although there has been recent progress in the
treatment and management of psychiatric disorders. During the past two decades,
there has been a considerable accumulation of findings in the neurobiology of
suicide [1].
Although many factors such as biological, psychological, and social factors are
involved in suicide behavior, the presence of mental disorder is thought to play an
important role in suicide [2]. More than 90% of suicide victims or suicide attempters
are diagnosed having mental disorders such as depression, schizophrenia, or alcohol
dependence (Fig. 1). However, the underlying mental disorders are diverse, and
the severity of the mental disorders does not necessarily correlate with increased
risk of suicide.
Even in the psychiatric groups at the highest risk, most patients never attempt
suicide. These findings indicate the importance of a diathesis or predisposition to
suicidal behavior that is independent of the main psychiatric disorder.
[Suicide and Mental Disorders (n=15,629) ]
Other Diagnosis on Axis 5.5% No Diagnosis 2.0%
Adjustment Disorders 2.3%
Anxiety Disorders/
Somatoform Disorders 4.8%
Mood disorders
Other Mental Disorders
4.1% (Depression)
30.2%
Organic Mental Disorder
6.3%
Personality Disorders Substance-Related
13.0% Disorders
(Alcohol-Related
Schizophrenia Disorders)
14.1% 17.6%
Preventing Suicide; A Resource for General Physicians.
WHO/WNH/MBD/00.1, World Health Organization, Geneva, 2000.
Fig. 1 Mental disorders in suicide. In completed suicide, about 30% of all suicide victims occur
in relation to mood disorders, and the rest are related to various other psychiatric disorders, includ-
ing substance-related disorders, such as substance abuse and alcoholism, schizophrenia and per-
sonality disorders. Source: World Health Organization: preventing suicide; a resource for general
physicians.WHO/WNH/MBD/00.1,WorldHealthOrganization,Geneva,2000
The Molecular Genetics of Suicide 289
Numerous studies on neurochemical abnormalities have been done; the results
obtained from those studies are summarized in Table 1. Recently, studies on the
biological aspect of suicide have been shifting from neurochemical to molecular
genetic approaches. Genetic studies on suicidal behavior have suggested that the
heritability of such behavior is independent of psychiatric diagnosis. Adoption
studies have shown a higher rate of suicide in the biological parents of adoptees
who commit suicide compared with biological relatives of control adoptees, even
after controlling for rates of psychosis and mood disorders (Table 2). Concordance
rates for suicide and suicide attempts are higher in monozygotic than dizygotic
twins, and the heritability of suicidal behavior was estimated to be approximately
55% based on twin studies in people with serious suicide attempts [3]. People who
commit suicide or make suicide attempts are those with a higher rate of familial
suicidal acts. These findings suggest the presence of hereditary and biological
factors that determine the threshold to commit suicide and which are independent
of specific psychiatric disorders. Thus, genetic factors in suicide have been a recent
focus, and recognition has been growing that suicide and suicidal behaviors are
highly familial and that genetics contributes to suicide and suicidal behaviors
(Table 3). In addition, completed suicides are thought to be more homogeneous
than suicide attempters in terms of inheritance of suicidal behavior [4].
Table 7 GenotypeandallelefrequenciesoftheCOMT158Val/Metpolymorphism
Genotype Allele
AA (%) AG (%) GG (%) A (%) G (%)
Suicides (n=163) 16(10) 79 (48) 68(42) 111 (34) 215(66)
Controls (n=169) 18 (11) 61(36) 90 (53) 97 (29) 241 (71)
Male suicides (n = 112) 9 (8) 60 (54) 43 (38) 78 (35) 146(65)
Male controls (n = 114) 10 (9) 42 (37) 62 (54) 62(27) 166(73)
Female suicides (n = 51) 7 (14) 19 (37) 25 (49) 33 (32) 69(68)
Female controls (n = 55) 8 (15) 19 (35) 28 (50) 35 (32) 75(68)
Total: genotype c2 = 5.4, df = 2, P=0.068;allelec2 = 2.2, df = 1, P = 0.14
Male: genotype c2=6.7,df = 2, P = 0.036; allele c2 = 3.1, df = 1, p = 0.080
Female: genotype c2=0.86,df = 2, P=0.96;allelec2 = 0.07, df = 2, P = 0.93
OnoH,ShirakawaO,NishiguchiNetal.(2004)[15]
commonA118GSNP.ThegenotypicandallelicdistributionsoftheA118GSNPwere
significantly different between the completed suicide and control groups (P = 0.014 and
0.039, respectively; Table 9). Moreover, the dominant model of genotype (AA vs. AG +
GG) analysis showed an enhanced association with suicide (P = 0.0041, OR = 0.575).
This finding means that individuals with one or two copies of the G allele of the A118G
SNPoftheOPRM1genearelessvulnerabletosuicide.Theseresultsraisethepossibility
thattheA118GSNPoftheOPRM1geneisassociatedwithsuicide.
We have conducted another association study examining the functional gene
polymorphisms of angiotensin-converting enzyme (insertion/deletion), prostaglan-
din E receptor subtype EP1 [17], and neuronal nitric oxide synthase (nNOS or
NOSI), which affect the HPA system, and demonstrated significant associations
with suicide. These findings have suggested that the disturbance of the HPA system
plays an important role in suicide (Table 9).
Recently, we found an association between regulators of G-protein signaling
(RGS) 2 gene polymorphisms and suicide and observed increased RGS2 immu-
noreactivity in the postmortem brains of suicide victims (Tables 10, and 11) [18].
RGSs are a family of proteins that negatively regulate intracellular signaling
of G protein-coupled receptors (GPCRs), such as the serotonin receptor. RGS2 is
thought to play an important role in anxiety and/or aggressive behavior.
To explore the involvement of the RGS2 gene in vulnerability to suicide, we
screenedJapanesesuicidevictimsforsequencevariationsintheRGS2geneandcar-
ried out an association study of RGS2 gene polymorphisms with suicide victims. In
the eight identified polymorphisms that were identified by mutation screening, we
genotyped four common single-nucleotide polymorphisms in the RGS2 gene and
foundsignificantdifferencesinthedistributionoftheSNP3genotypesandallelesof
theSNP2andtheSNP3betweencompletedsuicidesandthecontrols.Thedistribution
of the haplotype was also significantly different between the two groups (Table 11;
global < 0.0001). Furthermore, RGS2 immunoreactivity significantly increased in the
amygdala and the prefrontal cortex [Brodmann area 9] of the postmortem brain of the
suicide subjects (Fig. 2). These findings suggest that RGS2 is genetically involved in
the biological susceptibility to suicide in the Japanese population.
Table 8 Distributionofthesingle-nucleotidepolymorphisms(SNPs)intheOPRM1 gene
Statistics Statistics allele
Minor allele Minor allele genotype
Marker Controls frequency Completed suicides frequency P value c2 P value
rs1074287 AA AG GG AA AG GG
(n = 373) 74.8% 23.9% 1.3% 0.132 (n = 179) 71.5% 26.8% 1.7% 0.151 0.706 0.666 0.414 (0.927)
rs12205732 GG GA AA GG GA AA
(n = 372) 78.0% 21.2% 0.8% 0.114 (n = 170) 80.6% 17.6% 1.8% 0.106 0.403 0.127 0.722 (0.998)
A118G AA AG GG AA AG GG
(n = 367) 26.2% 57.2% 16.6% 0.452 (n = 181) 38.1% 46.4% 15.5% 0.387 0.014 4.252 0.039 (0.198)*
IVS2+G691C GG GC CC GG GC CC
(n = 370) 4.1% 37.3% 58.6% 0.227 (n = 170) 6.4% 37.4% 56.2% 0.251 0.471 0.762 0.383 (0.840)
HishimotoA,CuiH,MouriKetal.(2008)[16]
* P < 0.05
Table 9 Susceptible polymorphisms for suicide associated with HPA system
Functional variants associated with suicide
COMTVal158MetSNP
OPRM1Asn40AspSNP
ACE insertion/deletion polymorphism
ADRA2ApromoterC-1291GSNP
ProstaglandinEreceptorEP1geneSNPs
NeuronalnitricoxidesynthaseSNP
Functional polymorphisms not associated with completed suicide
FKBP5rs1360780T/Cpolymorphism
Peripheral benzodiazepine receptor gene polymorphism
Glucocorticoid receptor (BclIRFLP,N363S,ER22/23EK)polymorphism
HishimotoA,ShirakawasO,NishiguchiNetal.(2006)[17]
Table 10 GenotypedistributionsandallelefrequenciesofRGS2genepolymorphisms
Genotypefrequency Allelefrequency
SNPs Genotype Control Suicide P value Allele Control Suicide P value
SNP1 A/A 64(0.30) 39 (0.21) 0.068 A 228 (0.54) 176(0.47) 0.0293*
A/G 100 (0.48) 98 (0.52) df = 2 df = 1
G/G 46(0.22) 52 (0.27) c2=5.365 G 192(0.46) 202 (0.53) c2 = 4.749
SNP2 G/G 85 (0.40) 52 (0.28) 0.0383* G 260(0.62) 197 (0.53) 0.0175*
C/G 90 (0.43) 93 (0.51) df = 2 df = 1
C/C 35 (0.17) 39 (0.21) c2=6.527 C 160(0.38) 171 (0.47) c2=5.644
SNP3 C/C 68(0.32) 37 (0.20) 0.0105* C 243 (0.58) 179 (0.48) 0.0048**
C/G 107 (0.51) 105(0.56) df = 2 df = 1
G/G 35 (0.17) 45 (0.24) c2 = 9.119 G 177 (0.42) 195 (0.52) c2 = 7.939
SNP4 T/T 80 (0.38) 47(0.26) 0.0528 T 257(0.61) 189 (0.53) 0.0287*
C/T 97(0.46) 95 (0.54) df = 2 df = 1
C/C 33(0.16) 35 (0.20) c2 = 5.883 C 163(0.39) 165(0.47) c2 = 4.787
SNP1:−638A/G(rs2746071);SNP2:−395C/G(rs2746072);SNP3:2,971C/G(rs4606);SNP4:
3,438C/T(rs3767488)(dbSNPID)
CuiH,NishiguchiN,IvlevaEetal.(2008)[18]
* P < 0.05, ** P < 0.01
Table 11 HaplotypeanalysisofRGS2geneSNP1–4
Frequency Global Pvalue=0.000046
HaplotypeofSNP1–2–3–4 Control Suicide c2 P value
1 A-G-C-T 0.5183 0.4382 4.8671 0.0274*
2 G-C-G-C 0.3479 0.427 5.0199 0.0251*
3 G-G-G-T 0.0565 0.0285 3.5473 0.0596
4 G-G-C-T 0.0201 0.0266 0.3563 0.5506
5 G-G-G-C <0.01 0.0295 12.5573 <0.01**
6 G-C-G-T <0.01 0.0144 1.9983 0.1575
7 G-C-C-C 0.0161 <0.01 5.6202 0.0178*
8 A-C-C-T <0.01 0.0121 1.2136 0.2706
9 A-G-G-T <0.01 0.012 1.1609 0.2813
10 G-G-C-C 0.0119 <0.01 4.1444 0.0418*
… … <0.01 <0.01 … …
SNP1:–638A/G(rs2746071);SNP2:–395C/G(rs2746072);SNP3:2,971C/G(rs4606);SNP4:
3,438C/T(rs3767488)
*P < 0.05, **P < 0.01
CuiH,NishiguchiN,IvlevaEetal.(2008)[18]
296 K. Maeda et al.
Controls
Suicides
AMY
Suicides: 142.4 37.9 %
Controls: 100.0 37.9 %
BA9
Suicides: 133.2 34.1 %
Controls: 100.0 35.1 %
Conclusion
References
1. MannJJ(2003)Neurobiologyofsuicidalbehavior.NatRevNeurosci4:819–828
2. Brent DA, Mann JJ (2005) Family genetic studies, suicide, and suicidal behavior. Am J Med
Genet C Semin Med Genet 133:13–24
3. RoyA,SegalNL(2001)Suicidalbehaviorintwins:areplication.JAffectDisord66:71–74
4. BondyB,BuettnerA,ZillP(2006)Geneticsofsuicide.MolPsychiatry11:336–351
5. OnoH,ShirakawaO,NishiguchiNetal(2000)Tryptophanhydroxylasegenepolymorphisms
arenotassociatedwithsuicide.AmJMedGenet96:861–863
6. OnoH,ShirakawaO,NishiguchiNetal(2001)Serotonin2Areceptorgenepolymorphismis
notassociatedwithcompletedsuicide.JPsychiatrRes35:173–176
7. Nishiguchi N, Shirakawa O, Ono H et al (2001) No evidence of an association between
5HT1B receptor gene polymorphism and suicide victims in a Japanese population. Am J Med
Genet 105:343–345
The Molecular Genetics of Suicide 297
8. OnoH,ShirakawaO,KitamuraNetal(2002)Tryptophanhydroxylaseimmunoreactivityis
altered by the genetic variation in postmortem brain samples of both suicide victims and
controls. Mol Psychiatry 7:1127–1132
9. NishiguchiN,ShirakawaO,OnoHetal(2002)Lackofanassociationbetween5-HT1A
receptor gene structural polymorphisms and suicide victims. Am J Med Genet 114:
423–425
10. Ono H, Shirakawa O, Nishiguchi N et al (2002) No evidence of an association between a
functional monoamine oxidase a gene polymorphism and completed suicides. Am J Med
Genet 114:340–342
11. OkamuraK,ShirakawaO,NishiguchiNetal(2005)Lackofanassociationbetween5-HT6
receptorgenepolymorphismsandsuicidevictims.PsychiatryClinNeurosci59:345–349
12. Anguelova M, Benkelfat C, Turecki G (2003) A systematic review of association studies
investigating genes coding for serotonin receptors and the serotonin transporter: II. Suicidal
behavior.MolPsychiatry8:646–653
13. HattoriH,ShirakawaO,NishiguchiNetal(2006)Noevidenceofanassociationbetween
tyrosine hydroxylase gene polymorphisms and suicide victims. Kobe J Med Sci 52:
195–200
14. FukutakeM,HishimotoA,NishiguchiNetal(2008)Associationofa2A-aderenergic receptor
gene polymorphism with susceptibility to suicide in Japanese females. Prog
NeuropsychopharmacolBiolPsychiatry32:1428–1433
15. OnoH,ShirakawaO,NushidaHetal(2004)Associationbetweencatechol-O-methyltrans-
ferase functional polymorphism and male suicide completers. Neuropsychopharmacology
29:1374–1377
16. HishimotoA,CuiH,MouriKetal(2008)Afunctionalpolymorphismofthem-opioid receptor
geneisassociatedwithcompletedsuicides.JNeuralTransm115:531–536
17. HishimotoA,ShirakawaO,NishiguchiNetal(2006)Associationbetweenafunctionalpolymor-
phismintherenin-angiotensinsystemandcompletedsuicide.JNeuralTransm113:1915–1920
18. CuiH,NishiguchiN,IvlevaEetal(2008)AssociationofRGS2genepolymorphismswith
suicide and increased RGS2 immunoreactivity in the postmortem brain of suicide victims.
Neuropsychopharmacology33:1537–1544
Brief History and Current Status
of the International Neuropsychiatric
Association
Koho Miyoshi
Introduction
K. Miyoshi (*)
Jinmeikai Research Institute for Mental Health Kabutoyama-cho, Nishinomiya, Hyogo, Japan
e-mail: [email protected]
Dear Colleague:
Neuropsychiatry is a burgeoning discipline not only in the United States, but
worldwide. Recent binational meetings sponsored by the American
Neuropsychiatric Association and the British Neuropsychiatric Association
not only attest to widespread interest in neuropsychiatry but also to meaning-
ful and interesting differences in neuropsychiatric training, practice, and
thinking among countries.
This summer’s World Congress of Psychiatry in Madrid, Spain, offers an
excellent opportunity to convene neuropsychiatrists from around the world to
encourage the development of a world neuropsychiatric community that could
subsequently maintain communications over the Internet. The American
Neuropsychiatric Association (ANPA), The University of Illinois at Chicago
Department of Psychiatry, and The University of Seville Department of
Psychiatry have undertaken to organize an International Congress of Neuro-
psychiatry, to be held in Seville, Spain, for three days immediately following the
World Congress. The program includes plenary presentations by major
American, British, European and Latin-American scientists and clinicians, poster
sessions, and round table discussions. All major speakers have agreed to be avail-
able on the day of their talk for informal interchanges with participants, and all
major speakers will participate in lunch round tables on a variety of topics.
Interpreters will be available to facilitate cross-national communications.
The Hotel Alfonso XIII, site for the meeting, is one of the finest hotels in
the world, and Seville offers a picturesque venue that is a short flight from
other European cities.
We look forward to seeing you at this meeting, the first of many such
international gatherings.
Sincerely,
The International Congress of Neuropsychiatry
Organizing Committee
Brief History and Current Status of the International Neuropsychiatric Association 303
The First Congress was held successfully at the Hotel Alfonso XIII in Seville,
August 29–31, 1996. Large numbers of neuropsychiatrists, approximately 400,
from Europe, North and South America, and Asia-Oceania gathered there and
discussed enthusiastically the new organization of neuropsychiatry (Fig. 1).
The Topics in the First Congress were as follows:
Creating a World Neuropsychiatric Community, New Developments in
Dementia, The Frontal Lobe System, Advances in Brain Imaging, Keynote
Lecture: The Past and the Future of Neuropsychiatry, Basic and Clinical Topics in
Neuropsychiatry, Round Table Discussion (Education and Training, Organization
and Financing of Health Care, Outcome Studies, AIDS Dementia Complex,
Aggression, Psychiatric Symptoms in Dementia), The Practice of Neuropsychiatry
and System Care, Clinical Perspectives in Neuropsychiatry (Neuropsychiatry in
Europe, SPECT and Depression, Schizophreniform Phenomena and the Temporal
Lobe, Biology of Melancholia, Laterality and Schizophrenia, Building a World
Neuropsychiatric Community)
Soon after the congress in Seville, Dr. Colin Shapiro (Toronto) and Dr. Koho
Miyoshi (Kyoto) expressed their intentions to the organizing committee to hold
the coming neuropsychiatric congress in their countries. The arrangement of the
future congress was discussed in Orlando, Florida, where the Eighth Annual
Meeting of American Neuropsychiatric Association was held, in 1997. Dr.
Moises Gaviria attended the meeting on behalf of the organizing committee of the
First Congress in Seville. It was decided to hold the Second Congress in
Toronto.
Dr. Shapiro, Dr. Gaviria, and Dr. Miyoshi gathered to discuss about the financial
issues of the INA in Hawaii, where the Ninth Annual Congress of American
Neuropsychiatric Association was held in January 1998, and decided to manage the
organization temporarily by their financial contributions.
The Second International Congress of Neuropsychiatry, which was called
formally the “International Neuropsychiatry Congress (INC) and American
Neuropsychiatric Association (ANPA) 1998 Joint Meeting,” was held May 31 in
Toronto, Canada. The INA sessions and the ANPA sessions were held in the
meeting. In conjunction with the Toronto meeting, the INA, dedicated to promoting
the study of the brain and behavior from a neuropsychiatric perspective, was
established to supersede the ION. Twenty-four countries from Europe, North and
South America, Asia, and Australia were represented. It was decided that the
association would hold a biennial meeting.
As the first president of the association, Dr. Colin Shapiro (Canada) was
elected. Dr. Koho Miyoshi (vice-president) and Dr. Moises Gaviria (secretary-
general) were elected as the officers. Drs. R.H. Belmaker (Israel), E.Y.H. Chen
(Hong Kong), D.D. Dikeos (Greece), M. Robertson (UK), P. Sachdev (Australia),
and P. Sandor (Canada) were elected as the members of the Executive Committee.
The office of the INA was decided to be located in the office of the president in
Toronto. Dr. Paul Sander (Canada) was elected as a newsletter editor, and Dr.
Sharon Chung (Canada) was appointed as the secretariat in the INA office
(Fig. 2).
Brief History and Current Status of the International Neuropsychiatric Association 305
Fig. 2 The Presidents of the INA: Dr. C. Shapiro (1998–2002), Dr. P. Sachdev (2004–2006),
Dr. K. Miyoshi (2008–), and Dr. M. Gaviria (2002–2004) in Buenos Aires, 2002 (from left)
The scientific topics and principal speakers in the Second Congress were:
Neuropsychiatry of Tourettes’ Syndrome [M. Robertson (UK), E.C. Miquel
(Brazil), B.J.M. van de Wetering (the Netherlands)]; The Epidemiology of
Hallucinations [M. Ohayon (Canada)], Neuropsychiatry, and Neuroimaging
[L. Farde (Sweden), P. Martin (US), R. Dolan (UK)]; Genetics and Psychobiology
of Personality (Keynote Lecture) [C.R. Cloninger (US)]; Genetics of Neuropsychiatry
[K. Kidd (US), D. Black (US), M. Lepper (US), B. Clementz (US), P. Propping
(Germany)], and Art and the Mind [H. Walton (UK)].
The executive committee was held in New Orleans, where the Tenth Annual Congress
of American Neuropsychiatric Association took place, on January 31, 1999. The pro-
posed by-laws of the INA were examined and accepted by the executive committee.
The Third Congress was held in Kyoto, Japan, April 9–13, 2000. The venue of
the Congress was The Kyoto International Conference Hall at Takaragaike, Kyoto.
At the Kyoto meeting, approximately 700 people participated. Thirty-one countries
and districts, including Australia, Belgium, Canada, Croatia, Egypt, France,
Germany, Greece, Hong Kong, India, Israel, Italy, Japan, Lithuania, Macedonia, the
Netherlands, the Philippines, Poland, Russia, Spain, Sweden, Switzerland, Taiwan,
Thailand, Turkey, the United Arab Emirates, the United Kingdom, the United
States, Uruguay, and Venezuela, sent delegates.
The topics of the plenary lectures in the Third Congress were:
Dementia with Lewy Body [I. McKeith (UK)]; Tourette Syndrome [M.M.
Robertson (UK)]; Neuropsychiatry in the Elderly [J.L. Cummings (US)]; Zen and
the Brain [J.H. Austin (US)]; Functional Neuroimaging [H. Shibazaki (Japan)]; Sex
Differences in Brain Aging [C.E. Coffey (US)]; Biological Substrate of Late Life
Depression [J. Schweitzer (Australia)]; Neuropsychiatry of Stroke [R.G. Robinson
(US)]; Neuropsychiatry of Limbic and Subcortical Disorders [S.P. Salloway (US)];
Neuroimaging and Neurobiology of Schizophrenia [N. Andreasen (US)]; Brain
Laterality and Psychopathology [G. Gainotti (Italy)]; Behavioral and Psychological
Symptoms of Dementia [S.I. Finkel (US)]; Neuropsychiatry in 21st Century [C.
Shapiro (Canada)]; Positron Emission Tomography Studies in Schizophrenia [J.J.
Lopez-Ibor (Spain)]; Neuropsychiatric Aspects of Sleep Disorders [C. Soldatos
(Greece)], Brain Pathology of Dementia [K. Kosaka (Japan)]; Non-Alzheimer Type
of Dementia [L. Gustafson (Sweden)]; New Trends of Pharmacological Treatment
of Dementia [K. Miyoshi (Japan)]; and Schizophrenia-like Psychoses and Epilepsy
[P. Sachdev (Australia)].
The topics of the symposia and workshop in the Third Congress were:
Worldwide Collaborations in Neuropsychiatry, Transcranial Magnetic
Stimulation, Advances in Psychooncology and Psychoimmunology, Behavioral
Genetics of Personality, Vulnerability Markers of Schizophrenia, Vascular Dementia,
Biology of Eating Disorders, Dementia in Asian Countries, Brain and Behavior,
Molecular Genetics of Stimulant-Induced Psychosis, Neuropsychiatric Disorders
Brief History and Current Status of the International Neuropsychiatric Association 307
The Fourth Congress, chaired by Dr. Marquez (Buenos Aires), was held in Buenos
Aires in 2002. The venue was Hotel Crowne Plaza Panamericano in Buenos Aires.
Approximately 1,500 people participated in the Congress. The topics of the meeting
covered the neuropsychiatric field almost completely. As the Second INA president,
Dr. Moises Gavira (US) was elected for a 2-year term. Treasurer [G. Tortora
(Argentina)] and the members of Executive Committee [R. Belmaker (Israel),
K. Miyoshi (Japan), A. Kanner (US), E.S. Krishnamoorthy (India), M. Marquez
(Artentina), S. Shi (China), J. Tellez (Columbia), D. Dikeos (Greece), and C. Soldatos
(Greece)] were elected in the Executive Meeting.
Issues of the official journal and committees, such as the education committee
and membership committee, were discussed in the Executive Meeting.
The topics of the lectures in the Fourth Congress were:
Psychopathology in Epilepsy [A. Kanner (US)]; G-Protein Signaling in
Anti-Depressants [M. Rasenick (US)]; Tau Changes and Deficit in Episodic
Memory [L. Binder (US)]; Concussion in Sports [R. Bornstein (US)]; New
Trends in the Treatment of Dementia [K. Miyoshi (Japan)]; Cognitive Impairment
in Parkinson’s Disease [O. Gershanik (Argentine)]; Organic Psychosis as a Model
for Schizophrenia [P. Sachdev (Australia)]; Thyroid Hormone and Affective
Disorders [R. Bunevicius (Lithuania)]; Neuropsychological and Neuropsychiatric
Aspects of Electric Trauma [N. Pliskin (US)]; Mechanism and Action of mu-Opioides
[J. Lemos (US)]; Cognitive Therapy [R. Baber (US)]; Towards an Early and
Presymptomatic Diagnosis of Primary Degenerative Dementia [C. Mangone
(Argentine)]; Prion Disease [A.L. Taratuto (Argentine)], Sleep Disorders
[C. Shapiro (Canada)], Cognitive-Motor Disorder in Psychiatric Diseases
[R. Leiguarda (Argentine)]; Functional MRI in Neuropsychiatry [G. Stebbens
(US)]; Neurogenesis in Adult Hippocampus [A. Schinder (Argentine)]; Current
Status and Future Directions in Psychiatric Neurosurgery [K. Slavin (US)];
Music Brain and Culture [M. Gaviria (US)]; A Genetic Approach to the
Conceptual Nosological Continuum of Schizophrenia and Mood Disorders
[D. Dikeos (Greece)]; Vascular Dementia: an Overview [G. Roman (US)];
Chronobiology and Affective Disorders [D. Cardinali (Argentine)]; Attention
Deficit [J. Gutierrez (Mexico)]; Brain Damage and Recovery [M. Gaviria (US)];
308 K. Miyoshi
Dr. M Trimble (UK), the awardee of the Alwyn Lishman Award, presented the
lecture “The Evolution of the Limbic System and Epilepsy as a Clinical Model of
Dissolution,” and Dr. V.S. Ramachandran (US), the awardee of the Ramon y Cajal
Award, gave a lecture titled “Art and Brain.”
The topics of the lectures in the Fifth Congress were:
Vascular Dementia [M. Gaviria (US)]; Schizophrenia as Disorder of
Consciousness [C.R. Hojaij (Australia)]; Ethical Issues in Biological Psyc-
hiatry Research [A. Okasha (Egypt)]; Long-Term Outcome of Schizophrenia
[H.J. Möller (Germany)]; The Psychopathology of Fatigue [C. Shapiro (Canada)];
The Evolution of the Limbic System and Epilepsy [M. Trimble (UK)]; Art and
Brain [V.S. Ramachandran (US)]; Bipolar I and II Disorders [L. Judd (US)]; What
Causes the Onset of Psychosis? [R.M. Murray (UK)]; Can Stress Cause Depression?
[H.M. van Praag (the Netherlands)]; Catatonia [M. Fink (US)]; Neuropsychiatry of
Traumatic Brain Injury [R.G. Robinson (US)]; Brain Mechanisms of Cognitive
Processes [A. Georgopoulos (US)]; Towards New International Classification and
Diagnostic Systems [J.E. Mezzich (US)]; Biological Correlates of Obsessive
Compulsive Disorders [J.J. López-Ibor (Spain)]; Genetics of Mood Disorders
[J. Mendlewicz (Belgium)]; Agitated Depression in Bipolar Disorder [M. Maj
(Italy)]; Biological Perspectives in Psychiatric Prevention [G. Christodoulou
(Greece)]; Neuropsychiatric Aspects of Mental Disorders in Old Age [K. Miyoshi
(Japan)]; Impulsivity and Aggression [J.L. Ayuso-Gutiérrez (Spain)]; and Whither
Neuropsychiatry? [P.S. Sachdev (Australia)].
The topics of the symposia in the Fifth Congress were:
Cognitive Declines and Behavioral Changes in Neuropsychiatric Disorders,
Methodological Approaches in Neuropsychiatric Disorders, Psychopharmacology
for Children and Adolescents, Basic and Clinical Aspects on Hippocampus in
Depression, Management of Post Stroke Depression, ECT, Emerging Pharmacological
and Neuroimaging Strategies in the Evaluation and Treatment of Dementia,
Neuroimaging, Clinical and Experimental Europsychopharmacology, Clinical
Responses and Ethnopsychopharmacology, Neuropathological Vistas in
Neuropsychiatry, New Developments in Event-Related Potential Methodology,
The Sixth Congress was held in Sydney in 2006. Approximately 600 people
attended the congress. Neurobiological investigations of the endogenous psychoses
as well as the neuropsychiatric disorders were discussed in the congress. In this
congress, the realm of the neuropsychiatry was clearly widened by the impressive
presentations of the neurobiological bases of endogenous psychoses as well as
neuropsychiatric disorders. Dr. Perminder Sachdev, chair of the meeting, publicized
the core curriculum for the training of neuropsychiatrists.
In this meeting, Dr. Soldatos was elected the fourth INA president. Professor
Alvaro Pascual-Leone had been awarded the Ramon y Cajal award for his
outstanding contribution to the understanding of higher cognitive functions and the
treatment of neuropsychiatric disorders using transcranial magnetic stimulation.
The Lishman Award was awarded to Professor C. Edward Coffey, an accomplished
physician and healthcare leader recognized for developing highly successful
integrated mental health care systems and for his important contributions to
our understanding of brain–behavior relationships. As the Award Lectures,
Dr. A. Pascual-Leone presented a lecture entitled “The Right Side in Sigmund
Freud,” and Dr. C.E. Coffey gave a lecture on “Dramatically Improving the Quality
of Care in Neuropsychiatry” (Fig. 3).
Brief History and Current Status of the International Neuropsychiatric Association 311
Instead of the sessions of the plenary lectures, the symposia and workshops were
planned as the main sessions of the congress.
The topics of the symposia in the Sixth Congress were:
Neuropsychiatry as a Discipline for the Future (Presidential Symposium),
Transcranial Magnetic Stimulation, Neurophilosophy, Psychiatric Aspects of Epilepsy,
Traumatic Brain Injury (TBI), Neurology of Schizophrenia, Newer Antidepressants,
Delusional Belief, Developmental Neuropsychiatry, Neuropsychopharmacology,
Neuroimaging, Neurobiology of Consciousness, Movement Disorders and Catatonia,
Neurobiology of Hallucinations, Neuropsychiatry of Bipolar Disorder, Old Age
Psychiatry, Vitamins, Homocysteine and Omega 3 in Neuropsychiatry, Investigative
Applications, Genetics of Childhood-Onset Psychiatric Disorders, Brain Stimulation,
Neurobiology of Eating Disorders, Neuroimaging, Neurobiology of Melancholia,
Unusual and Uncommon Neuropsychiatric Syndromes, New Horizons in Epilepsy
and Behaviour, Brain Changes in Early Psychosis, Current Status of Vascular
Cognitive Impairment, An Update on ECT, Depression in Old Age, ADHD Across
the Lifespan, Catatonia and Cycloid Psychoses, Parkinson’s Disease, The Genetics of
Neuropsychiatric Disorders, Frontotemporal Dementia, Dementia, Controversies
about Mild Cognitive Impairment (MCI), Neuropsychiatry of Sleep, Brain Reserve,
and Traumatic Brain Injury.
Principal participants of the meeting in the program of the Sixth Congress were:
R.H. Belmaker (Israel), M. Bennett (Australia), S. Berkovic (Australia), H.
Brodaty (Australia), G.A. Broe (Australia), C.E. Coffey (US), D. Copolov (Australia),
A. David (UK), M. Gaviria (US), P. Hay (Australia), A. Jablensky (Australia), D.
Jeste (US), P. Joyce (New Zealand), E.S. Krishnamoorthy (India), H.S. Mayberg
(Canada), R. Meares (Australia), P. Mitchell (Australia), K. Miyoshi (Japan), C.
Pantelis (Australia), G. Parker (Australia), A. Pascual-Leone (US), G.C. Román
(US), M Ron (US), I. Skoog (Sweden), C. Shapiro (Canada), A. Snyder (US), C.R.
Soldatos (Greece), S.E. Starkstein (Australia), M. Trimble (UK), etc.
The Seventh Congress took place in Hotel Fiesta Americano Condesa, Cancun,
Mexico, December 3–5, 2008. As a chairman of the Congress, Professor Ricardo
Colin-Piana and his colleagues organized the meeting very nicely. Scientific topics
covered the neuropsychiatric field widely. The awardee of Cajal’s award, Dr. M Marsel
Mesulam, presented a lecture on “Primary Progressive Aphasia.” The awardee of
Lishman’s award, Dr. German E. Berrios, gave a lecture on “Neuropsychiatry–
Clinical Epistemology and Hermeneutics”.
The topics of the plenary lectures in the Seventh Congress were:
Early Detection of Neurodegenerative and Vascular Dementia [A. Wallin (Sweden)];
Behavioral Disturbances in Dementia [M. Mendez (US)]; Thyroid Axis Functioning
and Psychosis [R. Bunevicius (Lithuania)]; Atypical Viral Brain Infections [J. Sotelo
(Mexico)]; The Mayan Heritage [J.D. Vos (Mexico)]; Atypical Antipsychotics and
312 K. Miyoshi
Regional Activities
Currently, Dr. K. Miyoshi holds the position of INA president. Dr. M. Kopelman
will succeed to the presidency in Chennai in the year 2011. Dr. J. Trollor is
Brief History and Current Status of the International Neuropsychiatric Association 313
secretary-general and treasurer until 2013. Ms. Angie Russell is the secretariat in
the INA office, located in Sydney. Dr. M. Mula edits the newsletter, and Mr. L.
Tortora is the webmaster of INA. The INA Secretariat Office, Neuropsychiatric
Institute, The Prince of Wales Hospital, Randwick, NSW 2031, Australia (email:
[email protected]).
Executive Committee
The names of the Members of the Executive Committee are as follows: Ignacio
Brusco (Argentina), Robertas Bunevicius (Lithuania), Ricardo Colin-Piana (Mexico),
Anthony David (UK), Valsamma Eapen (Australia), Ennapadam S. Krishnamoorthy
(India), Robert Belmaker (Israel), Kiyoshi Maeda (Japan), Edward Coffey (US),
Moises Gaviria (US), Ingmar Skoog (Sweden), and David Arciniegas (US).
Advisory Council
The past presidents, Colin M. Shapiro (Canada), Moises Gaviria (US), Constantin
R. Soldatos (Greece) and Perminder Sachdev (Australia), are the members of the
Advisory Council. The core business of this committee is to advise the Executive
Committee (EC) on future directions for INA to propose nominations for vacancies
of the EC. All members will continue to sit on the Executive.
International Committee
The members of the International Committee are expected to (1) contribute to the
life and direction of INA, (2) act as key contacts for local activities of INA,
including taking initiative to hold local or regional scientific meetings on behalf of
INA, (3) communicate closely and regularly with the INA secretariat, (4) contribute
articles regarding neuropsychiatry activities in their country to the newsletter every
2 years, and (5) recruit new INA members in their own countries by sharing e-mail
lists of individuals who may have an interest in joining INA.
The members of the International Committee are:
Ahmed Malalia Salah AlAnsari (Bahrain), Celso Arango (Spain), Gilberto
Brofman (Brazil), Alexandre Castro Caldas (Portugal), Vytenis Deltuva (Lithuania),
Greg Finucane (New Zealand), Simon Fleminger (UK), Eduardo Gastelumendi
(Peru), Andres M Kanner (US), Francis Krivoy (Venezuela), Mario Lipez-Gomez
(Mexico), Maximiliano Luna (Argentina), Olya Mikova (Bulgaria), Ming-Chyi Pai
(Taiwan), Kang Seob Oh (Korea), Ranan Rimon (Finland), Ilya Reznik (Israel),
Bernard Saletu (Austria), Surachai Kuasirkul (Thai), Thordur Sigmundsson
314 K. Miyoshi
Awards
The Lishman Award was established to honor William Alwyn Lishman of London,
UK, as the modern pioneer of neuropsychiatry. It is awarded to an individual who
has made a notable contribution to clinical neuropsychiatry at an international level.
By accepting the award, the recipient presents a lecture at the Biennial Congress.
The Cajal Award was established to honor the “Father of Neuroscience,”
Santiago Ramon Y Cajal, and is awarded to an individual who has made a
distinguished contribution to neuroscience with an application to neuropsychiatry.
Similarly to the Lishman award, the recipient presents a lecture at the INA Congress
held every 2 years. The inaugural Lishman and Cajal Awards were given at the
Congress held in Buenos Aires, Argentina, in 2002.
Journal
Future Direction
Keywords Behavioralneurology•Curriculum•Neuropsychiatry•Specialization
•Training
*TheCurriculumCommitteeoftheINAwascomposedofP.Sachdev(chair),M.Gaviria,
C.Soldatos,C.Shapiro,J.Trollor,andE.S.Krishnamoorthy.
P. Sachdev (*)
School of Psychiatry, University of New South Wales,
Sydney, NSW 2052, Australia
and
NeuropsychiatricInstitute,EuroaCentre,PrinceofWalesHospital,
Barker Street, Randwick, NSW 2031, Australia
e-mail: [email protected]
Background
There is no one model that will suit all training programs in NP. An attempt is made
here to outline the basic tenets of such a program.
1. An NP training program shall endeavor to create specialists in NP who function
as secondary and tertiary level specialists. They shall provide consultations to
general psychiatrists, neurologists, and general physicians on a range of neurop-
sychiatric disorders.
2. An NP training program will generally comprise a 2-year fellowship program
that focuses on the core competencies detailed below. In some situations, only a
1-year fellowship in NP may be practicable. Full competency should not be
assumed after 1 year of training. However, if the trainee works for a further
2yearsinalargelyorexclusivelyneuropsychiatricservice(butnotspeciically
as a trainee), it would be considered likely that the training requirements would
have been met in this period.
3. The NP Fellow will have previously received training in psychiatry and/or neu-
rology. In general, this would have been a 3-year training program in a center
thatofferstraininginbothspecialties.Itisexpectedthatthepsychiatrytrainee
would have received at least 6 months training in neurology, but the neurology
trainee would have at least 1 year of training in psychiatry. If this is not the case,
the Fellowship period would be used to remedy this with a clinical rotation in the
appropriate discipline.
320 P. Sachdev et al.
Objectives
Attitude Objectives
Knowledge Objectives
(d) A basic grasp of issues related to the mind–brain debate, the biology of
consciousness, and other neurophilosophical issues.
2. Basic neuroscience:
(a) The molecular biology of psychiatric disorders;
(b) The biochemical basis of neuropsychopharmacology;
(c) The basic principles of neurophysiology, and their application to diagnosis
and treatment of neuropsychiatric disorders;
(d) ThebasicprinciplesofgeneticsandimmunologyastheyapplytotheCNS;
(e) The basic principles of neuroimaging and their application to diagnosis and
assessment of neuropsychiatric disorders.
3. Neuropsychiatric disorders
By the completion of training, NP trainees should be knowledgeable about the
epidemiology, etiology, psychopathology, clinical features (including complica-
tions), and natural history of neuropsychiatric disorders, including concepts of
impairment, disability, and handicap. A sound knowledge of the assessment and
careoftheseconditionsisalsoexpected.
(a) The incidence and prevalence of neuropsychiatric illnesses in various
populations;
(b) The phenomenology of organic brain syndromes, including nonspecific and
atypical presentations of illness such as “pseudodementia,” “masked”
depression, “conversion” disorders, and behavioral disorders;
(c) The criteria on which neuropsychiatric diagnoses are based, within the
framework of one of the widely accepted classification systems;
(d) Possiblecausativeorexacerbatingfactorsinneuropsychiatricdisorders;
(e) The natural history of the disease process in neuropsychiatric disorders,
which enables identification of (1) the severity of the disease; (2) the
urgency of the need for treatment; (3) the stage of the illness; and (4)
the prognosis;
(f) The assessment of common neuropsychiatric disorders, including the
following:
(i) Cognitivedisorders
(ii) Dementias and predementia syndromes
(iii) Nondementing cognitive disorders
(iv) Seizure disorders
(v) Movement disorders
(vi) Traumatic brain injury
(vii) Secondary psychiatric disorders, that is, psychosis, depression, mania,
andanxietydisorderssecondaryto“organic”braindisease
(viii) Substance-induced psychiatric disorders: alcohol, drugs of abuse, etc.
(ix) A ttentional disorders [adult attention deficit hyperactivity disorder
(ADHD)andrelatedsyndromes]
(x) Generalhospitalliaisonneuropsychiatry
322 P. Sachdev et al.
(xi) Developmentaldisorders
(xii) Sleepdisorders
(g) Appropriate management plans for neuropsychiatric disorders including:
(i) Interpretation of medical, psychological, and neurodiagnostic investi-
gations and assessments
(ii) T
he use of psychopharmacology, electroconvulsive therapy (ECT),
and other physical treatments including the frequency and manage-
ment of side effects
(iii) Application of psychotherapies, including supportive, cognitive-behav-
ioral, group, and family therapies
(iv) The use of behavior modification, environmental adaptation, and pre-
ventive measures
(v) Situations in which referral to, or consultation with, colleagues in
psychiatry and other disciplines is appropriate
(vi) Programs involving changes in lifestyle
(vii) Rehabilitation programs
(viii) Management in forensic settings
(ix) Strategiesthatmeettheneedsofcarersincludingtheroleofself-help
groups, including Alzheimer’s Association, Tourette Syndrome
Association, etc.
(h) The influence of specific factors on assessment and care of neuropsychiatric
disorders, including:
(i) Age
(ii) Intellectual capacity including intellectual disability
(iii) Medical illness and disability
(iv) Sex
(v) Culture
(vi) Spiritual beliefs
(vii) Socioeconomic status
(viii) Psychiatric comorbidity
(ix) Polypharmacy
(x) Supportfactors
(i) The influence of factors that affect treatment outcome including other medi-
cal illnesses;
(j) The principles underlying the choice and integration of interventions
in neuropsychiatry, including the evidence base and relative cost-
effectiveness;
(k) The principles of medicolegal aspects to the practice of NP, with particular
emphasis on mental health and guardianship legislation, including its local
application, testamentary capacity, enduring power of attorney, informed
consent, assessment of older offenders, and fitness to plead;
(l) The community care system including the relevant welfare legislation that
affects the management of people with neuropsychiatric disorders, especially
dementia;
NeuropsychiatricCoreCurriculumoftheINA 323
(m) Issues of aging and mental health in older people with intellectual and other
disabilities;
(n) Prevention and health promotion in NP;
(o) Issues specific to mental health promotion in relationship to neuropsychiatry;
(p) Risk factors for neuropsychiatric disorders that become apparent earlier in life.
4. Medicine in relationship to NP
By the completion of training, NP trainees should be knowledgeable about
medical and surgical conditions in general. Higher levels of knowledge are
expectedinthoseareasofmedicinethatparticularlyrelatetopsychiatricprac-
tice, such as neurology, rehabilitation medicine, etc.
5. Research method
By the completion of training, NP trainees should be knowledgeable about the
principles of scientific method in their practice and the use of this knowledge to
evaluate developments in neuropsychiatric research.
6. Service issues
By the completion of training, NP trainees should be knowledgeable about the
organization and delivery of mental health care to neuropsychiatric patients,
including the ethical, economic, geographic, and political constraints within
which it operates.
7. Professional responsibility
By the completion of training, NP trainees should be knowledgeable about the
principles of medical ethics, the development of professional attitudes, and
mechanisms for the development and maintenance of clinical competence,
acknowledging the need for professional and public accountability.
Skills Objectives
1. Healthpromotion
• By the completion of training, the NP trainee should be able to apply specific
knowledge of the principles and processes of health promotion and illness
prevention:
– Recognize and address risk factors for common neuropsychiatric prob-
lems in the community, in hospitals, and in long-term care, such as falls,
confusion, and depression;
– Recognize and address the needs of carers of neuropsychiatric patients.
2. Assessment of neuropsychiatric patients
• By the completion of training, trainees should possess the skills necessary for
performingacomprehensiveneuropsychiatricexamination:
324 P. Sachdev et al.
The curriculum below identifies some core competencies in the skill base and spe-
cific modules of the specialist knowledge base; these shall be acquired over 2 years.
The competencies are described as modules, but they are not necessarily indepen-
dent of each other. The importance of the core skills module is highlighted. The
aims and objectives of this module will normally be covered within the specific
clinical modules undertaken but should represent an additional and specific focus
ofstudywithintheindividualclinicalmodules.Thelevelofexpertiseineachofthe
specific modules will vary, depending upon the facilities available, but a basic level
ofcompetenceineachmoduleisexpectedina2-yeartrainingprogram.
1. Coreskillsmodule
1.1. Knowledge base in clinical neuroscience
1.2. Clinicalskillsinneuropsychiatry
1.2.1. Neuropsychiatricdiagnosisincludinghistoryandexamination,neu-
rophysiological investigations, neuroimaging, neuropsychology, and
other investigations;
1.2.2. Treatment, including pharmacology and other physical treatments
[ECT,transcranialmagneticstimulation(TMS),surgicalinterven-
tions], without neglecting psychotherapeutic and rehabilitative
interventions.
1.3. Criticalthinkinginneuropsychiatry:researchandscholarship
2. Specific modules
2.1. Cognitivedisorders
NeuropsychiatricCoreCurriculumoftheINA 325
Specific Competencies
ClinicalSkillsinNeuropsychiatry
(b) Perform a neuropsychiatric assessment; this will again involve and encom-
passalltheroutineskillsrequiredtocarryoutapsychiatricexamination,but
in addition will include:
• Demonstration of the ability to elicit information relevant to possible
neuropsychiatricdisordersandneurologicalconditions,forexample,the
ability to list the history of stepwise cognitive decline or psychomotor
seizure activity.
(c) Performacognitiveexamination(simpleandextended):
• A core skill in NP is the ability to carry out simple tests “at the bedside”
to determine a patient’s level of orientation, attention, concentration,
memory,etc.,andtodosointhecontextofapsychiatricexamination.
• A neuropsychiatrist, and in particular one from a neurological back-
ground, would be competent in assessing deficits in language, praxis,
gnosis, visuo-spatial function, and other cognitive syndromes.
• This competency would not require the ability to administer formal neu-
ropsychological tests, but may involve carrying out paper-and-pencil
tests and the use of simple materials such as word lists or pictures.
• A neuropsychiatrist should have competency in interpreting results of
suchanexaminationtodeterminewhetherthepatientissufferingfroma
dementing illness, a confusional state, or a specific cognitive deficit as
well as competency in diagnosing the range of adult psychiatric condi-
tions.Partoftheskillwouldinvolveplacingtheresultsoftheexamina-
tioninthecontextofthepatient’seducationalandsocialbackgroundand
premorbid level of functioning.
(d) Performaneurologicalexamination:
• The trainee should be able to carry out a full and detailed neurological
examination, if necessary, with particular emphasis on the central ner-
vous system and higher cortical functioning.
• The trainee should be able to demonstrate the ability to interpret any
abnormal signs elicited and place them in the context of the patient’s
presentation and a differential diagnosis; this may include eliciting signs,
which requires further specialist investigation, either within the realm of
NP or neurology or electrophysiology.
(e) Constructaneuropsychiatricdifferentialdiagnosis:
• The trainee neuropsychiatrist should be able to demonstrate familiarity
withmulti-axialformsofclassification.
• The trainee should be able to arrange multiple diagnoses into a rational
hierarchy and be able to summarize the key elements of the history and
examinationwhichsupportthatdifferentialdiagnosis.
• The trainee should be able to evaluate the extent to which patterns of
psychiatric symptomatology and presentation may be the result of under-
lying organic brain disease.
NeuropsychiatricCoreCurriculumoftheINA 327
• The trainee should be familiar with the range of organic disorders that
may account for particular presentations.
• The trainee should be able to communicate this in a clear and concise
way to other health professionals as well as to patients and their carers.
2. Undertake and plan investigation of a patient with apparent or possible neuro-
psychiatric problems:
(a) Trainees should be familiar with the relevant hematological, metabolic,
bacteriological,virological,immunological,andtoxicologicalinvestigations
of relevance to NP. This requirement includes:
• Demonstrating knowledge and judgment that the relevant parameter is of
central importance to the neuropsychiatric presentation.
• Knowing which investigations need to be pursued with further tests and
knowing which may be incidental or within normal limits.
• Interpretationofexaminationofcerebrospinalfluid,nerve,muscle,andbrain
biopsy will also be required, although detailed knowledge is not necessary.
(b) In contrast to many other specialities within psychiatry, NP requires familiarity
withEEGandotherneurophysiologicalinvestigationsandtheirinterpretation:
• The trainee should be able to discuss the advantages and limitations of
theroutineEEG,sleepEEG,andlonger-termEEGtelemetryinpatients
with possible neuropsychiatric problems.
• Although the trainee is not expected to be competent in reading EEGs
independently, she or he should have a working knowledge of the profiles
ofnormalandabnormalEEGs.
• In addition, the trainee should understand the use and application of sen-
sory evoked potentials and nerve conduction studies and EMG as they
occur in neurological disorders with neuropsychiatric complications, and
alsoasatooltoexcludeneurologicalcausesofabnormalfunctionthat
may in fact have a psychological basis.
• The trainee should be familiar with the settings in which these investiga-
tions are carried out, should be able to query the interpretation with a
consultantorexperiencedtechnicianinthearea,andtoconveythisinfor-
mation to members of the multidiscipline team, carers, and patients alike.
(c) NP requires sound understanding of the indications for, and interpretations
of, the various forms of brain imaging, both structural and functional,
includingmagneticresonanceimaging(MRI),computedtomography(CT),
single-photonemissioncomputedtomography(SPECT),andpositronemis-
sion tomography (PET):
• The trainee should have sufficient familiarity with these techniques to be
able to describe them to a patient and their family/carer and to be able to
interpret the results.
• The trainee should know when such investigations are likely to alter
management or treatment decisions and should have some understanding
of their theoretical importance.
328 P. Sachdev et al.
• Thetraineeshouldhavesufficientfirst-handknowledgeofCTandMRI
brain scans to be able to detect salient abnormalities and critically assess
anexpertreport.
3. Prescribe and oversee treatment of patients with neuropsychiatric disorders such
asthosewithpsychiatricandbehavioralsymptomsandcoexistingneurological
disorder. Be familiar with social, psychological, and biological interventions for
neuropsychiatric disorders:
(a) Thetraineeshouldhavesufficientskilltoexplainthemodeofaction,ben-
efits, and side effects of these treatments to fellow health professionals,
patients, and their families;
• Be familiar with the principles of treatment of major neurological disorders
and be familiar with neuropsychiatric complications of such treatment.
• The neuropsychiatrist should also be aware of the neurological manifes-
tations and complications of psychiatric treatment and advise patients
and professionals on evaluating the importance of these and in minimiz-
ing their occurrence and severity.
(b) Be familiar with potential drug interactions between psychiatric and neuro-
logical medications and other treatments;
• This requirement will include the awareness of the risks associated with
prescribing psychotropic drugs to patients with neurological and neuro-
surgical diseases.
(c) Be familiar with nonpharmacological treatments in neurological and
neuropsychiatric disorders;
• The trainee will have competence in the assessment for and the adminis-
trationofECTinitscurrentform.
• The trainee should have some understanding of the newer physical
treatments such as transcranial magnetic stimulation (TMS), vagus nerve
stimulation (VNS), deep brain stimulation (DBS), and other physical
treatments.
• The trainee should also acquire knowledge of the principles of neurore-
habilitation and familiarity with the concepts of disability and handicap.
4. Todiagnoseandtreatpatientswithmedicallyunexplainedsymptomsthatpresent
as neurological and neuropsychiatric problems; this includes working with col-
leagues in other disciplines to determine which further tests and investigations
are necessary or not as the case may be;
(a) NP should involve competence in understanding the possible social, cultural,
andfamilyinluencesonunexplainedneurologicalsymptoms.
(b) The trainee should be able to develop a grasp of the principles behind cogni-
tive-behavioral treatments for such patients and be able to plan and oversee
such treatments carried out by another professional such as a trained nurse
or clinical psychologist.
NeuropsychiatricCoreCurriculumoftheINA 329
(c) The trainee should be aware of the relationship between NP and allied psy-
chiatric subspecialties such as old age, child and learning disability psychiatry,
and which service patients might most appropriately be served by.
CriticalThinkinginNeuropsychiatry:ResearchandScholarship
A specialist training in NP will equip the trainee to think critically in the field. The
trainee should be able to critically assess the empirical evidence in support of any
clinical practice, including the ability to criticize published material. This skill can
be developed by means of journal clubs, attendance at research meetings, research
presentations, short-term courses, etc.
It is expected that in the second year of training, the trainee will undertake a
research project. This work should ideally involve all the steps in an empirical
project (background review, design of study, applying for ethics clearance, data
gathering, analysis, and report preparation). However, it may take the form of a
critical review of a current topic, or a case series. The trainee will produce a report
of a publishable standard, as judged by the supervisors, and will be encouraged to
publish in a peer-reviewed journal. The research report will be a mandatory com-
ponent of the second year of training.
Specific Modules
SpecificCompetencies
Diagnostic Techniques
Be familiar with the main principles involved in the management and treatment of
cognitive disorders and of dementias
HowTaught
HowAssessed
1. Clinicalsupervision
2. Direct observation
3. Clinicallogbook
4. Clinicalaudit
5. Casepresentations,etc.
SpecificCompetencies
(b) Be familiar with the indications for and interpretation of structural and func-
tional neuroimaging in people with epilepsy.
4. Prescribetreatmenttopatientswithcoexistingneurologicaldisorder:
(a) Be familiar with social and psychological interventions for the treatment of
epilepsy including relaxation techniques and other behavioral methods of
controlling/inhibiting seizures;
(b) Be familiar with the principles of the medical treatment of the different
seizure and syndrome types;
(c) Be familiar with potential drug interactions between psychiatric medications
and anticonvulsants;
(d) Be aware of the risks associated with prescribing psychotropic agents to
patients with epilepsy;
(e) Be familiar with the surgical treatment of epilepsy including vagal nerve
stimulation.
5. Assess and manage special patient groups with epilepsy:
(a) Be familiar with the difficulties in assessing and managing seizure disor-
ders in children and adolescents with epilepsy, including issues around
puberty;
(b) Be familiar with the difficulties in assessing and managing seizure disor-
ders in women with epilepsy, including catamenial epilepsy, contracep-
tion, pregnancy, teratogenicity, polycystic ovarian syndrome, and
menopause;
(c) Be familiar with the difficulties in assessing and managing seizure disorders
in older age patients, including cognition and issues regarding concomitant
physical illnesses and medication;
(d) Be familiar with the difficulties in assessing and managing seizure disorders
in patients with learning disability including etiology, difficulty eliciting a
history, and cognitive and treatment issues.
6. Assess and manage psychiatric comorbidity in people with epilepsy: pre-ictal,
ctal, post-ictal, inter-ictal, and iatrogenic:
(a) Be familiar with the diagnosis and management of depression in people with
epilepsy, including the risk of suicide;
(b) Befamiliarwiththediagnosisandmanagementofanxiety/panicattacksin
people with epilepsy, including the difficulties in differentiating between
panic attacks and ictal panic;
(c) Be familiar with the diagnosis and management of psychosis (post-ictal
psychosis, chronic inter-ictal psychosis, and forced normalization) in people
with epilepsy;
(d) Be familiar with the diagnosis and management of cognitive dysfunction in
people with epilepsy, resulting from seizures and anticonvulsant medication,
including the role of neuropsychological assessments;
(e) Be familiar with the diagnosis and management of sexual dysfunction in
people with epilepsy;
NeuropsychiatricCoreCurriculumoftheINA 333
SpecificCompetencies
1. Clinicalassessment
(a) Take a history of movement disorder
(b) Assess psychiatric history
(c) Assess neurological history
(d) Performpsychiatricexamination
(e) Performneurologicalexamination
(f) Constructdifferentialdiagnosisofmovementdisorder
2. Investigation
(a) Reviewpreviousneurologicalexaminations
(b) Review previous neurological treatment
(c) Review previous psychiatric treatment
(d) Order further relevant investigations
3. Treatment
(a) Review previous psychiatric treatment
(b) Review previous neurological treatment
(c) Recommend alterations to current treatment
(d) Prescribe new appropriate treatment
(e) Review effects of treatment
SuggestedAssessmentMethod:ClinicLogbook
1. Parkinson’s disease
2. Tourette’s syndrome: tics
3. Tremor
4. Dystonia
5. Catatonia
6. Neuroleptic-induced movement disorders: tardive dyskinesia, tardive dystonia,
akathisia, NMS, drug-induced parkinsonism, etc.
7. Hystericalconversion/somatizationdisorders
ClinicalSettings
SpecificCompetencies
ClinicalSettings
1. Psychiatric wards
2. Neuropsychiatric outpatient clinics
3. Medical and surgical wards
SpecificCompetencies
ClinicalSettings
SpecificCompetencies
2. Competencyintheinvestigationofthesesyndromes,includingbiologicaland
psychosocial investigations.
3. Experienceinthetreatmentofsuchsyndromes,includingtheuseofpsychotro-
pic drugs and psychosocial and rehabilitative interventions.
4. Knowledge of the pathophysiological mechanisms underlying the development
of these syndromes.
ClinicalSettings
1. SpecializedadultADHDclinic
2. Psychiatric wards
3. Neuropsychiatric outpatient clinics
SpecificCompetencies
1. FamiliaritywithcommonpresentationsofADHDinadults.
2. Competency in the investigation of attentional and frontal dysexecutive syn-
dromes, including biological and psychosocial investigations.
3. Experienceinthetreatmentofsuchsyndromes,includingtheuseofpsychotro-
pic drugs and psychosocial and rehabilitative interventions.
4. Knowledge of the pathophysiological mechanisms underlying the development
of these syndromes.
KeyCompetencies
Undertakeassessmentofpatientswithunexplainedneurologicalsymptoms:
1. Take an appropriate neuropsychiatric history.
2. Interpret previously performed investigations.
3. Performexaminationofmentalandphysicalstatus.
4. Assessthepatients’functioninthecontextoftheirdisability.
5. Understand the concepts of conversion, somatization, and dissociation in a neu-
rologicalcontext.
6. Formulate appropriate management plans.
7. Communicateinformationtotheneurologicalteam.
(b) Assesspatients’functioninthecontextoftheirdisability
Suggested learning methods Suggested assessment methods
Observation/modeling Validated self-assessment
Supervised clinical practice Clinicalsupervision
Specific teaching from relevant health Clinicallogbook
professionals (e.g., occupational Casepresentation
therapist)
(e) Communicateinformationtoneurologyteam
Suggested learning methods Suggested assessment methods
Obervation/modeling Clinicalsupervision
Supervised clinical practice Direct observation
(c) Constructanappropriatedifferentialdiagnosis(deliriumvs.depressionvs.
dementia)
Suggested learning methods Suggested assessment methods
Supervised clinical practice Clinicalsupervision
Appropriate reading Casepresentation
Clinicallogbook
Validated self-assessment
Preamble
Developmental NP is that branch of psychiatry concerned with the diagnosis and man-
agement of emotional, behavioral, and learning disorders that are associated with
demonstrable or suspected organic brain dysfunction, and which manifest during
childhood. Because these disorders are primarily disruptive to normal developmental
attainments or adjustment, they are known as neurodevelopmental disorders. The prac-
tice of developmental NP requires skills and knowledge that encompass not only child
psychiatry, in broad terms, but also pediatric neurology and learning disabilities.
Currently,thereisnoformaltrainingprogramleadingtoaspecificaccreditation
in developmental NP. In this respect, the subspecialty is in the same category as
NeuropsychiatricCoreCurriculumoftheINA 339
adult NP. Few child psychiatric training programs explicitly include training in
developmental NP. However, it is arguable that within the clinical field of child
psychiatry, neurodevelopmental disorders are now the predominant reason for
specialist referral.
The competencies outlined below describe the minimum range of skills in devel-
opmental NP that should be acquired by consultant child psychiatrists in training.
Werecommendthatalltraineeshaveatleast1yearofexperienceinthisspecialty,
but that those who intend to become specialists in this area may choose to spend
additional time gaining particular skills.
SpecificCompetencies
1. Undertake clinical assessment of patients with apparent neurodevelopmental
disorders
(a) Take a developmental neuropsychiatric history
Suggested learning methods Suggested assessment methods
Observation/modeling Validated self-assessment
Working as a team member In-training assessment
Supervised clinical practice Clinicalsupervision
Focused training courses Direct observation of clinical work
Peer review
Clinicallogbook
Clinicalaudit
Casepresentations
Review of case notes and other records
Chart-stimulatedrecall
2. Advise staff about the interpretation and management of abnormal mental states
and behaviors
Suggested learning methods Suggested assessment methods
Observation/modeling Clinicalsupervision
Supervised clinical practice Direct observation
(b) Produce a differential diagnosis and formulation for patients with mental
disorder in this setting
Suggested learning methods Suggested assessment methods
Supervised clinical practice Clinicalsupervision
Appropriate reading Casepresentation
Clinicallogbook
Corecompetenciesinassessmentandmanagementofpatientswithsleepdisorders
SpecificCompetencies
Haveknowledgeofetiology,prevalence,diagnosis,categorization,andtreatment
of sleep disorders:
1. Primary insomnia
2. Secondary insomnia
3. Hypersomnias
4. Parasomnias
5. Neuropsychiatric consequences of sleep apnoea syndrome
342 P. Sachdev et al.
Diagnostic Techniques
Management
1. Observation/modeling
2. Supervised clinical practice
3. Readingrelevanttexts
4. Peer group discussion
5. Multidisciplinary case conferences, journal clubs
6. Speciic teaching from relevant health professionals (e.g., EEG, respiratory,
neurology)
7. Primary responsibility for diagnosis and treatment of a reasonable number and
adequate variety of patients with acute and chronic sleep disorders (e.g., at least
five hypersomnia, five parasomnia, ten insomnia, of a range of ages)
8. Attendance at a respiratory sleep disorder clinic for the diagnosis of sleep
apnea
9. Attendance at multidisciplinary national conferences
NeuropsychiatricCoreCurriculumoftheINA 343
1. Validated self-assessment
2. Clinicalsupervisionandfeedback
3. Casepresentation
4. Clinicallogbook
ClinicalSettings
Knowledge
Skills
4. To work with the MDT, including psychologists and other therapists, to produce
an overall treatment strategy for symptoms.
5. To interpret neuropsychometric test results sufficiently to produce a neuropsy-
chiatric formulation.
6. To set up, in collaboration with the MDT, a program of therapy based on goal
planning.
7. To work alongside psychologists, behavioral nurse therapists, and others to
implement cognitive-behavioral treatments and behavioral treatments.
8. To set up effective aftercare following inpatient rehabilitation, based on good
communication across health services, social services, statutory services, and
voluntary sector.
9. To undertake a risk assessment for all commonly occurring risks following
acquired brain injury, and ensure that there are procedures in place to offer a
reasonable risk management strategy.
10. To understand the symptoms and signs of the post-concussion syndrome and pro-
vide advice to patients following a brain injury to minimize the risk of problems on
returning to work, and/or return to living in the community with family and/or carers.
11. To appreciate the psychodynamic processes that follow brain injury and other
forms of disability and provide appropriate psychotherapeutic support.
12. To manage the common sequelae of brain injury, including disturbances of
mood, psychotic disorders, personality change (especially associated with anti-
social behavior), and reduced initiation and motivation.
KeyCompetencies
Acknowledgements WearegratefultotheBritishNeuropsychiatricAssociation(H.Ring)and
the American Neuropsychiatric Association (D. Arciniegas) for making their curricula available
for use during the preparation of this document. Members of the Neuropsychiatric Section of the
RANZCPmadeusefulcontributions(P.Hay,S.Starkstein,M.Hopwood,D.Velakoulis).Lauren
Norton and Angie Russell assisted with manuscript preparation.
Recommended Reading
Neuropsychiatry Textbooks
1. YudofskySC,HalesRE(2008)Textbookofneuropsychiatryandclinicalneurosciences,5th
edn. American Psychiatric Publishing, Arlington, VA
2. David A, Fleminger S, Kopelman M, Lovestone S (2009) Lishman’s organic psychiatry: a
textbookofneuropsychiatry.Wiley-Blackwell,London
3. Cummings JL, Mega MS (2003) Neuropsychiatry and behavioral neuroscience. Oxford
University Press, New York
4. SchifferRB,RaoSM,FogelBS(2003)Neuropsychiatry:acomprehensivetextbook,2ndedn.
Lippincott Williams & Wilkins, Baltimore
5. Coffey CE, Brumback RA (1998) Textbook of pediatric neuropsychiatry. American
PsychiatricPublishing,Washington,DC
6. CoffeyCE,CummingsJL(2000)Textbookofgeriatricneuropsychiatry,2ndedn.American
PsychiatricPublishing,Washington,DC
7. ArciniegasDB,BeresfordTP(2001)Neuropsychiatry:anintroductoryapproach.Cambridge
UniversityPress,Cambridge
8. SachdevP(withKeshavanM)(2010)Secondaryschizophrenia.CambridgeUniversityPress,
Cambridge
Behavioral Neurology
9. M-Marsel M (2000) Principles of behavioral and cognitive neurology, 2nd edn. Oxford
University Press, New York
10. Pincus JH, Tucker GJ (2003) Behavioral neurology, 4th edn. Oxford University Press,
New York
11. Feinberg TE, Farah MJ (1997) Behavioral neurology and neuropsychology. McGraw-Hill,
New York
346 P. Sachdev et al.
12. Kirshner HS (2002) Behavioral neurology: practical science of mind and brain, 2nd edn.
Butterworth-Heinemann,Boston
13. Leon-CarrionJ,GianniniMJ(2001)Behavioralneurologyintheelderly.CRC,BocaRaton
14. Strub RL, Black FW (1981) Neurobehavioral disorders: a clinical approach, 2nd edn. Davis,
Philadelphia
15. Cummings JL, Trimble MR (2002) Concise guide to neuropsychiatry and behavioral
neurology, 2nd edn. American Psychiatric Publishing, Arlington, VA
16. TrimbleMR,CummingsJL(1997)Contemporarybehavioralneurology(bluebooksofprac-
ticalneurology),vol16.Butterworth-Heinemann,Boston
Neuropsychology
17. Lezak MD, Howieson DB, Loring DW (with Hannay HJ, Fischer JS) (2004) Neuro-
psychologicalassessment,4thedn.OxfordUniversityPress,NewYork
18. WalshK,DarbyD(1999)Neuropsychology:aclinicalapproach,4thedn.ChurchillLiving-
stone, Edinburgh
Neuroscience
19. KandelER,SchwartzJH,JessellTM(2000)Principlesofneuralsciences,4thedn.McGraw-
Hill,NewYork
20. SquireLR,BloomFE,SpitzerNC,duLacS,GhoshA,BergD(2008)Fundamentalneurosci-
ence, 3rd edn. Academic, Burlington
21. SachdevP(2002)Neuropsychiatry:adisciplineforthefuture.JPsychosomRes53:625–627
(Editorial)
22. SachdevP(2005)Whitherneuropsychiatry?JNeuropsychiatryClinNeurosci17:140–144
23. PriceBH,AdamsRD,CoyleJT(2000)Neurologyandpsychiatry:closingthegreatdivide.
Neurology54:8–14
24. ANPA Standards for Fellowship Training in Neuropsychiatry (2001) I. Definition of neurop-
sychiatry. http://www.neuropsychiatry.com/ANPA
25. Accreditation Council on Graduate Medical Education (2001) Program requirements for
training in psychiatry. http://www.acgme.org/RRC/Psy_Req2.asp
26. Accreditation Council on Graduate Medical Education (2002) Program requirements for
training in neurology. http://www.acgme.org/RRC/Psy_Req2.asp
27. American Board of Psychiatry and Neurology, Inc (2003) Information for applicants for
certification in the subspecialties of addiction psychiatry, clinical neurophysiology, forensic
psychiatry, geriatric psychiatry, and neurodevelopmental disabilities. http://www.abpn.com/
Downloads/2003subspec_ifa.pdf
28. Academy of Psychosomatic Medicine (1998) Standards for fellowship training in consulta-
tion-liaison psychiatry. http://www.apm.org/fellow.html
Index
A Body consciousness, 72
Activities of daily living (ADL), 39, 165 Body image, 100
Adoption studies, 289 Body schema, 72
Adult guardianship law, 232 Body-scheme disorders, 95
Age Gene/Environment Susceptibility- BPRS, 43
Reykjavik Study, 170 Brain-derived neurotrophic factor, 174
Alcohol dependence, 288 Brain Impairment Behavior
Allan–Herndon–Dudley syndrome, 24 Bother Scale, 172
Alpha synuclein, 248 Brain Impairment Behavior
Alzheimer’s dementia, 229 Inventory, 172
Alzheimer’s disease, 25, 26, 56, 150, Brain injury, 38
214, 236 Burden, 38
Alzheimer type pathology, 248
Amygdala, 113, 132
Angiotensin-converting enzyme, 293 C
Anosognosia, 34, 72 Capgras syndrome, 70
a1-Antichymotrypsin, 178, 194 Catechol-O-methyltransferase
Antidepressants, 28 (COMT), 291
Antiepileptic drugs, 104, 108, 109 CBT, 37
Anxiety, 10, 25 Cerebral white matter, 132
Apathy, 10, 132, 206 Chalder Fatigue Scale, 169
Apathy Scale, 166 Cingulate cortex, 132
Apolipoprotein E, 237 Cingulated, 36
Asomatognosia, 100 Clinical epileptology, 95
Attention-deficit disorder, 22 Cognitive impairment, 149–157, 170
Autoimmune thyroid disease, 20, 26 Complex partial seizure, 96
Ayahuasca, 281–285 Confabulation, 84
Confusion Assessment Protocol, 135–136
Consciousness, 283–285
B Consultations, 317–319, 333–336
BADS, 40 Contralateral release phenomenon, 165
BDI, 43 Copy number variant, 240
Behavioral and psychological symptoms of Corticobasal degeneration, 13
dementia, 233 C-reactive protein (CRP), 191, 192
Behavioral change, 12 Cretinism, 23
Behavioral dyscontrol, 132 Creutzfeldt-Jakob disease (CJD), 59
Behavioral neurology, 317, 318, 345 CT, 273, 274
Benzodiazepines, 166 Curriculum, 317, 318, 324
Bipolar disorder, 35, 109 Cytokines, 178
347
348 Index
D Frontal lobe, 36
Deiodinase, 20, 25, 26, 28 Frontal lobe syndrome, 173
Delirium, 12, 26 Frontotemporal lobar degeneration (FTLD),
Delirium Rating Scale-Revised-98, 135–136 236, 261
Delusion, 11 Fusiform area, 113
Delusional misidentification syndrome
(DMS), 70
Delusional perception, 66 G
Dementia, 12, 38, 149–157, 203, 235 Galveston Orientation and Amnesia Test
Dementia of the Alzheimer type, 13 (GOAT), 128
Dementias of non-Alzheimer type, 13 General physicians, 317–319
Dementia with Lewy bodies (DLB), 14, 52, Genetic biomarkers, 216
247–251, 256 Glasgow Coma Scale, 128
clinical features, 249–250 G-protein signaling 2 (RGS2), 293
Demyelination, 154 Grey matter, 36
Denial, 34 Guidelines for diagnoses and treatment of
Depression, 25–27, 103–110, 151, 153–154, dementia, 234
156–157, 202, 281, 283, 285, 288
Depressive disorder, 35
DEX, 34 H
Diagnosis, 317–319, 321, 324, 326, 329, 330, Hallucination, 11
332, 333, 338–342 Hashimoto’s encephalopathy, 27
Diffuse Lewy body disease, 256 Herpes encephalitis, 83–84
Diffuse neurofibrillary tangles with calcifica- Hippocampus, 132
tion, 271–277 Human rights, 233
clinical features, 274–275 Huntington’s disease, 58
pathological features, 275–276 Hypertension, 152–155
Dignity, 232 Hyperthyroidism, 21, 25
Discrepancy, 40 Hypothalamic-pituitary-adrenal
Dopamine transporter imaging, 251 (HPA) system, 290
Dorsolateral prefrontal cortex, 37, 132 Hypothyroidism, 21, 25
Dyssomnia, 167
I
E Ictal autoscopy hallucination, 99
Early detection, 228, 230 Ictal visual spatial distortions, 98
Early diagnosis, 215 Idiosyncratic auras, 97
Edelman’s Model, 73 Inferior (inferolatera) prefrontal cortex, 132
Emotional disinhibition syndrome, 171 Inflammation, 178, 179
Emotional Intensity Scale (EIS), 67 Informant, 40
Entorhinal-hippocampal complex, 132 Interictal dysphoric disorder, 103–110
Epilepsy, 96, 113 Interleukins, 178, 191–193
Epilepsy addendum for psychiatric International Index of Erectile Function, 169
assessment, 108 International Neuropsychiatric Association
Evaluation, 317–320, 342 (INA), 301–315, 317, 318
Executive function impairments, 132, 134 Advisory Council, 312
Alwyn Lishman Award, 314
executive committee, 313
F history, 301–312
Fahr-type calcification, 271, 273, 274, 277 INA Office, 312
Fluctuation, 249, 250 International Committee, 313
Forgetfulness, 229 Ramon Y. Cajal Award, 314
Frontal assessment battery, 136–137 The Fifth Congress, 308
Index 349
PCRS, 34 R
Peri-ictal symptoms, 106 Rancho Los Amigo Scale (RLAS), 135–136
Persistent cognitive impairment, 12 Rankin Scale, 168
Persistent neurological memory disorders, REM sleep, 167
81–84 REM sleep behaviour disorder, 250, 251
Persistent psychogenic amnesia, 87–88 3-Repeat tau protein, 276
Personality change, 12 4-Repeat tau protein, 276
Perth Community Stroke Project, 164 Research, 317–320, 323, 324, 329
Pick bodies, 264 Restless leg syndrome, 168
Polymorphism, 290 Reticulocortical system, 132
Possible neuropsychiatric symptoms, 7 Reticulothalamic system, 132
Poststroke Risk factors of suicide, 289
agitation, 173
anxiety, 165
apathy, 166 S
cognitive performance, 170 SAI-E, 34
depression, 173 Schizophrenia, 26, 27, 34, 288
disturbance of the sleep-wake cycle, 167 Serotonergic system, 287
fatigue, 168 Serum amyloid A, 193
involuntary emotional expression disorder, Silent infarcts, 154
171 Single nucleotide polymorphism (SNP), 291
irritability, 172 Single-photon emission computed tomography
mania, 164 (SPECT), 165
psychosis, 172 Situation-specific amnesia, 86–87
sexual dysfunction, 169 Skin conductance responses (SCRs), 70
Posttraumatic amnesia (PTA) Somatoparaphrenia, 72
aggression, 132 Specialization, 317
definition, 132 Stroke, 152–154
Galveston Orientation and Amnesia Test Subcortical dementia, 13
(GOAT), 128, 136 Subtle neurological signs, 136
Orientation Log (O-Log), 128, 136 Suicide, 287
outcome prediction using PTA, 129 SUMD, 34
Posttraumatic coma, 132, 133 Symptomatic epilepsy, 96
Posttraumatic dysexecutive syndrome,
132–134
Posttraumatic encephalopathy, 132–134 T
Posttraumatic seizures, 139 TDP-43, 239, 266
Presenilin, 237 Teaching, 317, 318, 320, 337, 338, 342, 344
Prevalence, 248 Temporal cortices, anterior, 132
Primary care physicians, 229–231 Temporal lobe, 36
Probable neuropsychiatric symptoms, 6 Temporal lobe epilepsy, 96, 105
Programs, 317–319, 322, 333, 339, 344 Thyroid and brain development, 23, 28
Progranulin, 239 Thyroid and mood disorders, 27
Progressive supranuclear palsy, 13, 57 Thyroid and schizophrenia, 27
Prosopagnosia, 70 Thyroid hormones, 20, 28
Prostaglandin E receptor, 293 receptors, 21
Psychiatrists, 317–319, 339 transporters, 21, 25, 29
Psychiatry, 113 Thyrotropin-releasing hormone (TRH), 26, 28
Psychogenic fugue, 85–86 Training, 317–321, 323, 324, 329, 338–340
Psychotic disorders following traumatic brain Transcranial magnetic stimulation, 175
injury (PDFTBI), 66 Transient epileptic amnesia, 80
Psychotic symptoms, 151 Transient global amnesia, 79–80
Psychotropic, 282 Transient ischemic attack (TIA), 168
Index 351