2286
Marcello Pinti et al.
DOI: 10.1002/eji.201546178
Eur. J. Immunol. 2016. 46: 2286–2301
HIGHLIGHTS
REVIEW
Aging of the immune system: Focus on inflammation
and vaccination
Marcello Pinti1 , Victor Appay2 , Judith Campisi3 , Daniela Frasca4 ,
Tamas Fülöp5 , Delphine Sauce2 , Anis Larbi6 , Birgit Weinberger7
and Andrea Cossarizza8
1
Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
Sorbonne Universités, UPMC Univ. Paris 06, DHU FAST, CR7, Centre d’Immunologie et des
Maladies Infectieuses (CIMI-Paris), Paris, France
3
USA and Lawrence Berkeley National Laboratory, Buck Institute for Research on Aging,
Berkeley, CA USA
4
Department of Microbiology and Immunology, University of Miami Miller School of
Medicine, Miami, FL, USA
5
Division of Geriatrics, Department of Medicine, Research Center on Aging, University of
Sherbrooke, Canada
6
Singapore Immunology Network (SIgN), Aging and Immunity Program, A*STAR, Singapore
7
Institute for Biomedical Aging ResearchUniversity of Innsbruck, Innsbruck, Austria
8
Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of
Modena and Reggio Emilia School of Medicine, Modena, Italy
2
Major advances in preventing, delaying, or curing individual pathologies are responsible
for an increasingly long life span in the developed parts of our planet, and indeed reaching
eight to nine decades of life is nowadays extremely frequent. However, medical and
sanitary advances have not prevented or delayed the underlying cause of the disparate
pathologies occurring in the elderly: aging itself. The identification of the basis of the
aging processes that drives the multiple pathologies and loss of function typical of older
individuals is a major challenge in current aging research. Among the possible causes,
an impairment of the immune system plays a major role, and indeed numerous studies
have described immunological changes which occur with age. Far from the intention of
being exhaustive, this review will focus on recent advances and views on the role that
modifications of cell signalling and remodelling of the immune response play during
human aging and longevity, paying particular attention to phenomena which are linked
to the so called inflammaging process, such as dysregulation of innate immunity, altered
T-cell or B-cell maturation and differentiation, as well as to the implications of immune
aging for vaccination strategies in the elderly.
Keywords: Aging r B lymphocytes r Longevity r NK cells r Signaling r T lymphocytes r Vaccine
Correspondence: Prof. Andrea Cossarizza
e-mail:
[email protected]
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.eji-journal.eu
Eur. J. Immunol. 2016. 46: 2286–2301
Introduction
A long human life span—exceeding eight decades or more,
for example—is becoming increasingly attainable in the developed countries. Globally, the number of persons aged >60 is
expected to more than double by 2050, increasing from 901 million in 2015 to 2.1 billion in 2050, and that of persons aged
>80 is projected to increase from 125 million in 2015 to 434
million in 2050 (United Nations: World Population Prospects;
http://esa.un.org/unpd/wpp/). Unfortunately, an equally long
health span—years of healthy life, devoid of disease and diminished vigor—lags seriously behind the gains in lifespan [1]. Why
is the case? In large measure, the remarkable increase in lifespan
enjoyed by modern humans is due to major advances in preventing, delaying or curing individual pathologies such as infections,
hypertension, type 2 diabetes, and even some forms of cancer [2].
While modern biomedical interventions are successful at, for
example, reducing cardiovascular disease due to hypertension or
high blood cholesterol levels, basic aging processes continue to
impair, for example, the host response to lung function, cognition, muscle strength, and bone integrity. A major challenge in
current aging research, then, is to identify the basic aging processes driving the multiple pathologies and loss of function that
afflict older individuals, with the long-term goal of developing
effective interventions which ameliorate their effects.
What are these basic aging processes? For humans, this question remains formally unanswered. Nevertheless, a very large body
of experimental evidence from a wide variety of organisms ranging from yeast to primates strongly suggests there are at least nine
evolutionarily conserved hallmarks of aging that almost certainly
derive from a small handful of basic aging processes [3]. These
hallmarks of aging include stem cell exhaustion, altered intercellular communication, genomic instability and telomere attrition,
epigenetic alterations, loss of protein homeostasis (proteostasis),
altered nutrient and growth factor sensing, mitochondrial dysfunction, and cellular senescence [3]. There are still many open
questions regarding the prime causes and ultimate effects of these
hallmarks. However, emerging studies are beginning to identify
commonalities among the causes and effects of at least some of
these hallmarks. One of these commonalities has been linked to
the immune system: low levels of chronic inflammation, otherwise
known as inflammaging or inflamm-aging [4].
Inflammaging is a hallmark of virtually every major age-related
disease and phenotype and has been shown to be a defining pathological characteristic of aging tissues across multiple species [5].
Inflammaging is characterized by the low level persistent infiltration of immune cells, primarily but not exclusively cells of
the innate immune system, and elevated levels of several proinflammatory cytokines and chemokines [6], both within the tissue microenvironment and the systemic milieu. Because cells of
both the adaptive and innate immune systems change with age,
as discussed more extensively later in this review, the phenotypes
of these infiltrating immune cells remain to be thoroughly characterized.
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
HIGHLIGHTS
While a general consensus on a specific biomarker of inflammaging has never been reached, increased levels of circulating
inflammatory mediators such as pro-inflammatory cytokines and
acute phase proteins, e.g. interleukin-6 (IL-6) and C-reactive protein (CRP) are commonly used as indicators of inflammaging. In
particular, analysis on thousands of elderly subjects show that IL-6
and CRP levels systematically increase in an age-dependent manner, even in subjects never diagnosed with diseases commonly
associated with age, such as cardiovascular disease, myocardial
infarction, stroke, type 2 diabetes, or cancer [7].
The origin(s) of the cytokines and chemokines that attract
immune cells during inflammaging are incompletely understood.
One possible hypothesis–-based upon evidence observed in disease characterized by an accelerated aging of the immune system,
such as HIV infection [8]—is that microbial products translocated
from the gut might find their way into the circulation and ultimately into tissues more easily in elderly people, because of an
age-related increases in gut and/or vascular permeability [9].
The microbial composition and diversity of the gut ecosystem
changes with aging [10], as Bifidobacteria have been reported
to decrease [11], while facultative anaerobes, including Streptococci, Staphylococci, Enterococci, and Enterobacteria, increase with
age [12]; this alteration is associated with increased serum levels
of IL-6 and IL-8 [13].
Another origin might be the age-related accumulation of senescent cells—cells that have entered a state of irreversibly arrested
cell proliferation (growth) and altered function as a consequence
of many of the stresses that are known to increase with age [14].
These stresses include genome and epigenomic damage, activation of oncogenes, metabolic imbalances and mitochondrial dysfunction, among others. The senescence growth arrest almost
certainly evolved to suppress the development of cancer [15].
However, another hallmark of senescent cells is the acquisition of a
senescence-associated secretory phenotype (SASP), which entails
the chronic transcriptional induction and secretion of numerous
pro-inflammatory cytokines, chemokines, growth factors and proteases [16]. As senescent cells chronically release chemokines,
they may promote leukocyte recruitment, a well-known function
of chemokines and, as shown in an in vivo model with senescent
tumor cells, innate immune cells can migrate into the vicinity of
the senescent tumor area [17, 18]. The pro-inflammatory nature
of the SASP is generally considered deleterious [16, 19]. However,
using a mouse model in which senescent cells can be detected in
living animals, SASP was also recently shown to promote wound
healing and optimize the formation of certain embryonic structures. In this model, senescent fibroblasts appear at wound sites a
few days after skin injury, and these wound-associated senescent
cells promote optimal wound healing by secreting PDGF-A, a SASP
factor, which in turn promotes myofibroblast differentiation. [20].
Finally, SASP can optimize the formation of certain embryonic
structures [20, 21]. Thus, although the SASP can promote tissue
repair and remodelling, which also requires a controlled inflammatory response, but it can also become maladaptive and promote
aging phenotypes and pathologies when chronically present.
www.eji-journal.eu
2287
2288
Marcello Pinti et al.
The age-related accumulation of senescent cells was recently
shown to shorten both lifespan and health span in mice [22].
Senescent cells secrete a distinct suite of inflammatory cytokines
and chemokines depending upon whether senescence is induced
by genotoxic stress, [23] or mitochondrial dysfunction, as in
the form of deficiencies in mitochondrial sirtuins or damage to
mitochondrial DNA [24], each of which are considered distinct
hallmarks of aging [3]. Thus, activation of the immune system
by senescent cells might account for more than one hallmark of
aging. Likewise, activation of immune function by other hallmarks
of aging – loss of proteostasis, for example–might also account for
multiple aging phenotypes and pathologies.
Adipose tissue has been shown to play a major role in the
regulation of inflammatory status. Adiponectin, which is associated with lean states and insulin sensitivity, has been hailed as an
anti-inflammatory force in adipose tissue by regulating the production of anti-inflammatory cytokines, and polarizing macrophages
toward anti-inflammatory M2 phenotype [25, 26]. Leptin, on the
other hand, is produced in states of abundant adipose tissue and
systemic inflammatory distress, and has been shown to induce
the production of TNF-α, IL-6, and IL-12 in both human and
murine monocytes [27, 28]. The normal aging process often leads
to increased levels of visceral and subcutaneous adipose tissue.
Higher numbers of resident macrophages and T cells from adipose tissue have been found in aged mice, and their number is
correlated with higher inflammation. Concomitant with greater
body fat percentage, aged mice also have more adipose tissue T
cells (ATTs) than young mice, which can contribute to create a
proinflammatory environment in visceral fat [29].
It has been suggested that this age-associated accumulation
of adipose tissue is the cause of elevated inflammatory cytokines
observed in obese individuals [30, 31], and indeed up to 30% of
circulating IL-6 could derive from adipose tissue in human healthy
subjects [32]. The contribution of adipose tissue to inflammaging is further supported by studies showing that elderly subjects
who exercise regularly and are leaner have fewer senescent T
cells and lower circulating pro-inflammatory cytokines [33], and
that healthy centenarians with low adipose mass and high insulin
sensitivity do not show elements of the proinflammatory profile
[34].
Innate immunity: still a complex question
Likely because of the clear and easily detectable effects of thymic
involution on naı̈ve and memory peripheral blood T lymphocytes
[35], studies on immunosenescence have been focused on adaptive immunity for decades. For a long time, innate immunity was
considered basically unaffected by aging but, especially after the
birth of the concept of inflammaging, several studies have demonstrated that crucial components of the innate immune system also
undergo profound changes, which are related to an increased risk
of infections and higher infection-related mortality. Moreover, the
importance of molecules containing damage-associated molecular patterns (DAMPs), such as mitochondrial DNA, in activating
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Eur. J. Immunol. 2016. 46: 2286–2301
innate immune cells and maintaining the status of inflammaging
is emerging [36].
The number of neutrophils does not change with age, but
profound functional alterations have been observed in this cell
type [37, 38]. In particular, neutrophils from elderly subjects are
characterized by a reduced capability to migrate towards a chemotactic signal, probably because of a constitutive activation of the
lipid kinase phosphoinositide 3-kinase [PI3 K] [39]; the same
phenomenon has been observed in mice, and has been attributed
to reduced expression of ICAM-1 [38]. It must be noted that
such reduced chemotactic capacity could also lead to a diminished egress of neutrophils from inflamed tissue, thereby contributing to higher local inflammation, as observed in aged mice
after burn-associated lung injury [40]. Concerning phagocytosis
and killing of ingested microorganisms, neutrophils from elderly
subjects show a well-preserved capability to ingest non-opsonized
particles [41], but a reduced capability to uptake opsonized particles, or pathogens such as Escherichia coli [37, 42]; in mice,
neutrophils from aged animals also display a reduced capability
to form neutrophil extracellular traps (NETs) in a model of severe
skin infection by Staphylococcus aureus [43]. This reduction could
be partially due to the lower expression of CD16 in neutrophils
from elderly subjects [37]. Interestingly, centenarians, the best
example of successful aging, show well-preserved neutrophil functions, such as bacterial phagocytosis, chemotaxis and superoxide
production, which are comparable to those of young subjects [42].
A crucial mechanism for activation of innate immune response
is the engagement of pattern recognition receptors by specific
agonists. Peripheral blood mononuclear cells (PBMCs) from old
individuals (ࣙ 65 years) have been shown to have a delayed
and altered transcriptional response to stimulation with TLR4,
TLR7/8, and RIG-I agonists; this altered response is accompanied
by a decreased production of the pro-inflammatory and antiviral cytokines TNF-α, IL-6, IL-1β, IFN-α, and IFN-γ, and of the
chemokines CCL2 and CCL7 [44].
Monocytes can be schematically divided in three main subsets on the basis of their phenotype: classical (CD14++ CD16− ),
non-classical (CD14+ CD16++ ) and intermediate (CD14++ CD16+ )
[45]. Aging has not been shown to significantly alter the absolute
number and the frequency of overall monocytes in humans [44],
but does determines significant changes in the relative distribution
of their subsets, with a marked reduction of the classical subset
and an increase in the number of intermediate and non-classical
monocytes [46]. As to functionality, significant age-related reduction of reactive oxygen species (ROS) production and phagocytosis
capability have been described [46, 47], along with profound dysregulation in the release of different cytokines after the activation
of monocytes through Toll-like receptors (TLR). The synthesis of
TNF-α and IL-6 after TLR1/2 engagement, for example, is severely
reduced in human monocytes, while release of TNF-α upon TLR4
stimulation is increased [48]. Furthermore, monocytes from aged
donors have been shown to release higher levels of IL-8 after
stimulation of TLR1/2, TLR2/6, TLR4, or TLR5 [49]. Such dysregulation appears to be caused by both alteration in surface TLR
expression and impairment of downstream signaling: while TLR2
www.eji-journal.eu
Eur. J. Immunol. 2016. 46: 2286–2301
expression is constant, TLR1 expression declines with age, and
activation of MAPK and ERK1/1 pathways after TLR1/2 triggering is severely reduced in cells from elderly subjects [49]. In contrast, signaling downstream of TLR5 has been shown to increase
with age [49]. It has to be underlined, however, that most of these
data concerning humans have been obtained in isolated monocytes
treated in vitro, and some of the contrasting results observed could
be due to enhanced responsiveness from cells with progressive
differentiation in vitro [50]. Similarly, some in vivo data have
been obtained on rodent models, and are often contrasting, probably because of different strains and experimental condition used.
In humans, the functional consequences of similar, possible alterations are less known. However, it has been shown recently that
there are no age-related differences in the capacity of the synthetic TLR4 agonist glucopyranosyl lipid A to induce expression
of co-stimulatory molecules or production of cytokines by human
antigen-presenting cells [51].
With regards to dendritic cells (DCs), age-related changes
in the frequency and absolute number of plasmacytoid DCs
(pDC) and myeloid DCs (mDC) were discordantly reported
[44, 52–55]. Conversely, it is well established that Langerhans
cells (LCs) markedly diminish with age [56, 57], and that such
a reduction could contribute to the higher risk of skin infection in elderly subjects [58]. Concerning the capability to secrete
cytokines upon stimulation, contrasting data exist for mDCs:
while some studies have indicated an increased secretion of proinflammatory cytokines in elderly subjects, others showed no
change or a decreased production [59, 60]. pDCs are characterized by a marked impairment of pro-inflammatory cytokine
release with aging: pDCs display a reduction in intracellular levels of TNF-α, IL-6 and IL-12, as well as IFN-α, IFN-β, and IFN-γ
upon viral or TLR stimulation [52, 53]; however, phagocytosis appears well preserved [ 41]. As the expression of TLRs in
pDCs is constant over the life [61], it is likely that this impairment is caused by defects in signal transduction, as discussed
below.
Data obtained in mice indicate that DC recruitment to lymphoid organs is also impaired with aging, probably because a
combination of both direct alterations in DC capacity to respond
to cytokine and chemokine stimulation, and indirect effects such
as age-related defects of lymphoid organ microenvironment. In
humans, a reduced mobilization of LCs from the skin to lymphoid organs upon TNF-alpha stimulation have been shown in
aged subjects [56], while an age-associated increase in TLR4- and
TLR8-dependent cytokine production has been observed in human
MDDCs [59]. The lower capacity of pDCs from elderly subjects
to release IFNs and pro-inflammatory cytokines has been associated with a reduced response to influenza vaccine [62]. However,
basal production of pro-inflammatory cytokines in the absence
of TLR engagement has been found to be higher in cells from
older compared with young individuals, suggesting a dysregulation of cytokine production that may limit further activation by
TLR engagement [44].
Finally, minimal attention has been paid to the effects of age
on mast cells, basophils and eosinophils. Concerning mast cells,
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
HIGHLIGHTS
mouse models have given inconsistent results, and data show no
age-related changes, or a reduced activity [63, 64]; degranulation was found increased in mast cells from aged mice, probably
because of a lower expression of FcγRIIB/III, a negative regulator
of their function [65]. No recent studies are available on basophils,
while eosinophils have been studied in young and elderly asthmatic patients, and indicate no age-related changed in either number or functions of these cells [66].
The general picture that emerges is that of a profound dysregulation of innate immune functions, with some functions downregulated, and others up-regulated or even enhanced. In particular, an increase in the basal production of proinflammatory
cytokines, observed in different cell types, could be a major contributor to the age-related increase of the levels of such molecules
observed in several cohorts of elderly subjects [19, 67, 68].
T lymphocytes in old humans: a matter of
quantity and quality
T cells are generated in the thymus, a primary lymphoid organ
that undergoes gradual decay with age. This process is referred to
as thymic involution, and is characterized by the progressive deterioration and disappearance of functional thymic compartments
(cortex and medulla) and accumulation of adipose tissue, so that
only traces of functional thymic tissues are found at age 70 or
more [69]. The exact causes of thymic involution are however not
fully understood. Age-related changes in the levels of thymostimulatory growth hormones (e.g., decreasing levels of GH and IGF-1),
or steroid hormones (increased during puberty), potentially thymosuppressive, and inflammatory cytokines (e.g., increasing levels of IL-6), as well as from oxidative stress-induced damages may
play a role in this process [69, 70]. However, the early-in-life
initiation of thymus decay in most vertebrates suggests a potential evolutionary role of this process, although this remains to be
understood [71]. Aging is also associated with apparent quantitative changes of naı̈ve and effector memory CD4+ or CD8+ αβ
T-cell subsets. The reduction in the frequency of naı̈ve T cells,
together with the increasing proportion of terminally differentiated T lymphocytes and the decrease in cells expressing T-cell
receptor rearrangement excision circles (the DNA molecules deriving from somatic recombination of TCR alpha chain, which are
present only in T cells recently egressed from thymus) have long
been hallmarks of immune aging [72]. These cellular changes are
commonly reported in the circulation (i.e., the blood) [73], but
can also be extended to lymphoid tissues and organs (e.g., spleen,
lymph nodes, lung, and gut) of old subjects [74].
Studies of young subjects having undergone thymectomy
during childhood have provided clear mechanistic insights into
the decline of the naı̈ve T-cell compartment in elderly humans
[75, 76]. These studies showed that, independently from age,
reduced thymic function and lower production of naı̈ve T cells,
together with the consumption of these cells (e.g., upon activation and differentiation during a viral infection), lead to
reduced numbers of naı̈ve T cells [75, 76]. This setting of partial
www.eji-journal.eu
2289
2290
Marcello Pinti et al.
lymphopenia results in increasing naive T-cell homeostatic proliferation, although this is not sufficient to maintain a constant
number of these cells [77, 78]. Reduced number of naı̈ve T cells is
associated with a less diverse TCR repertoire of the total T lymphocyte compartment overtime [79]. Nonetheless, despite their lower
frequency, the repertoire richness of the naı̈ve T cells declines only
modestly in healthy elderly adults, i.e., —two- to fivefold if compared with that of young adults [80]. Of note, the elderly naı̈ve
T-cell repertoire was characterized by large T-cell clones (distinct
from memory clones), suggesting that an uneven homeostatic proliferation occurs in the naı̈ve T-cell compartment.
In addition to quantitative changes, naı̈ve T cells display functional defects with advanced age. Altered TCR signaling and TCRinduced ERK phosphorylation, resulting in blunted activation,
have been reported in CD4+ and CD8+ naı̈ve T cells from old
people [81, 82]. In naı̈ve CD4+ T cells, the desensitization of
the TCR cascade was even associated with increased activity of
dual-specific phosphatase 6 [DUSP6], a negative regulator of the
ERK pathway [81]. Of note, the increased expression in T cells
of another dual specificity phosphatase, DUSP4, was also found
to cause defective TCR responses and aging markers reminiscent
of T-cell senescence [83]. Both these quantitative and qualitative defects of naı̈ve T cells result in a lower capacity of aged
individuals to induce de novo antigen-specific T-cell responses.
Using an in vitro model of T-cell priming, it was indeed shown
that CD8+ T cells from elderly individuals consistently mounted
impaired responses specific to a model neoantigen, such as MelanA/MART-1 [82]. Studies in old mice also revealed a decreased
trafficking capacity and reduced motility of naı̈ve CD4+ T cells
in lymph nodes prior to antigen encounter, thus indicative of
delayed immune cell recruitment and antigen recognition [84].
Reduced T-cell priming capacity (i.e., diminished CD4+ and CD8+
T-cell responses specific for neoantigens) with advanced age was
recently demonstrated in vivo, during a primary virus infection
experimentally induced in old humans by immunization with liveattenuated yellow fever vaccine [85]. Overall, these deficits likely
compromise priming of early adaptive immune responses in old
individuals, thus contributing to their susceptibility to infection or
cancer.
The increased proportion of terminally differentiated oligoclonal effector memory T-cell populations in the elderly has
been shown to be the consequence of recurrent or chronic
immune activation [86]. The latter usually results from the
numerous and recurrent challenges that the immune system
faces over time, in particular related to infection with persistent
viruses, such as cytomegalovirus (CMV). Terminally differentiated
(CD28− CD57+ ) T lymphocytes are characterized by strong effector functions such as expression of cytotoxins (e.g., Perforin and
Granzyme B) and cytolytic capacity, and a marked inflammatory
profile by way of increase proinflammatory cytokine (e.g., IFNγ, TNF-α, MIP-1β) secretion [86]. As a consequence, the accumulation of these cells likely participates in the establishment
of the hyperinflammatory status, characteristic of the old person. However, terminally differentiated T cells are constrained by
limited proliferative capabilities in tandem with short telomeres,
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Eur. J. Immunol. 2016. 46: 2286–2301
and to a large extent, have been defined as approaching senescence [86, 87]. Of note, the intensity of the immune response
upon activation can be regulated through the action of various coinhibitory receptors or immune check points (e.g., PD-1), which
can, for instance, lower the degree of expansion and differentiation of activated cells. On the one hand, this type of regulation
enables the immune system to adapt to the levels of antigenic stimulation, which could be beneficial in terms of limiting the immune
alterations eventually associated with aging. On the other hand,
it may also potentially limit the efficacy of the cellular immune
response in the setting of infectious or malignant diseases. Moreover, a less efficient response to infections with increasing age
may be related to a narrowing of the repertoire, ensuing recurrent
antigen exposure, and resulting in either holes in the repertoire
or even loss of effective T cells. Detailed analyses of the human
TCR repertoires of influenza A virus-specific CD8+ T cells, for
instance, revealed a direct correlation between increasing age and
narrowing of the TCR repertoire [88]. This may be associated
with an age-associated increase in the proportion of low-avidity
virus-specific CD8+ T cells, as shown in the case of CMV infection
for instance [89]. Recent studies have also provided new insights
into the molecular defects of terminally differentiated “senescent”
cells, reporting defective mitochondrial function, and elevated levels of ROS as well as p38 MAPK, associated with alterations of
energetic metabolism as well as autophagy, a major cellular lysosomal degradation pathway [90, 91]. Of note, blocking the p38
signaling pathway has been shown to enhance the proliferation of
such terminally differentiated T cells without compromising their
capacity for cytokine secretion [92]. Moreover, the use of spermidine, which enhances autophagy, could help rejuvenate CD8+
T-cell responses in old mice infected with influenza or murine
CMV [93]. These studies therefore identify potential intervention
targets for restoring responsiveness for these T cells and improving
aged immunity.
Similarly to the changes observed on αβ T cells, the γδ T-cell
compartment is also significantly affected by age. It is characterized by a decline in total γδ T-cell frequency along with phenotypic (accumulation of highly differentiated cells) and TCR repertoire changes (inflation of Vδ2-γδ T cells), phenomena which are
accentuated by CMV infection [94, 95]. Functional analyses of γδ
T cells from old individuals are nonetheless required to uncover
potential qualitative defects with aging. Altogether, these findings
provide further insights into the quantitative and qualitative cellular immune alterations and insufficiencies that accompany human
aging.
Natural killer cells: changes in their subsets
Based on the expression of the surface markers CD56 and
CD16, three NK-cell subsets have been characterized in humans.
CD56dim CD16+ cells represent approximately 90% of circulating
NK cells and are considered as mature mainly cytotoxic subset; CD56bright CD16neg/dim cells constitute approximately 10% of
the NK-cell population, and are considered immature with a
www.eji-journal.eu
HIGHLIGHTS
Eur. J. Immunol. 2016. 46: 2286–2301
cytokine-mediated immune-modulatory role [96]. Furthermore, a
scarce subset of NK cells, devoid of CD56 expression and displaying a reduced functional capacity, has been identified in healthy
controls and in chronic viral infections such HIV and hepatitis C
virus (HCV) [97].
Reports of changes in NK-cell phenotype and function with
old age have been inconsistent. The proportion of CD56bright NK
cells appears to be reduced in the elderly, which is likely the
consequence of an impaired production of new NK cells with
advanced age, due to a lower output from the bone marrow [98].
Instead, percentages and absolute numbers of CD56dim CD16+ NK
cells have been described to be either maintained, increased, or
decreased in the elderly population [99, 100]. NK cells from
elderly subjects exhibit generally normal IFN-γ production capacity, but a defective capacity to secrete chemokines post-stimulation
[101], accompanied by a reduced cytotoxic potential against MHC
class I molecule negative target cell lines [99].
The impact of aging on the expression of NK-cell receptors has
been recently reviewed [102] revealing that CD16 expression and
function, which is a key receptor for antibody-dependent cellular
cytotoxicity (ADCC), is not altered in the elderly, whereas the
expression of the activating natural cytotoxicity receptors (NCRs),
NKp30 and NKp46 and DNAM-1 are diminished [103]. There seem
to be limited age-related changes in KIR and NKG2 repertoires of
NK cells. A decrease in NKG2A expression occurs from young to
elderly adults [104, 105]. In contrast, an increased frequency of
KIR expression was observed in NK cells from cord blood to adults
without any further increases in the elderly [98].
Similarly to T cells, CD57 can be a marker of replicative
senescence for NK cells which have high expression of KIR, low
expression of NKG2A, decreased sensitivity to cytokines, reduced
replicative potential and high cytotoxicity properties [106]. Downregulation of NKG2A, acquisition and high expression of KIR and
expression of CD57 have been shown to correlate independently
with terminal differentiation, as shown by reduction in proliferation capacity, homing molecules, response to cytokines, and
expression of activation markers [107].
With a half-life estimated about 12 days in healthy young individuals [100], NK cells have been classically considered shortlived effector cells. However, the analysis of NK-cell homeostasis
in old donors, showing a decreased de novo production of NK cells
despite a relatively well preserved number of peripheral NK cells,
suggests the persistence of a high proportion of long-lived NK cells
in the elderly [108]. Little is known on the factors involved in the
generation of long-lived NK cells in humans. Recent evidence has
demonstrated that CMV infection could be a parameter associated
with this expansion of long-lived ‘memory-like’ NK cells, which
are characterized by the expression of CD94/NKG2C and CD57
[105, 109]. Moreover, this chronic viral infection leads to imprints
in the human KIR repertoires [110, 111]. Lately, this unusual
clonal expansion of NKG2C+ CD57+ NK cells has been found
in elderly CMV-seronegative donors, indicating that parameters
related to aging other than CMV could influence the peripheral
repertoire [105]. Mechanisms involved could include common factors between CMV infection and immune aging such as immune
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
senescence, pro-inflammatory environment and increased homeostatic turnover.
In conclusion, aging is associated with a gradual loss of the
CD56bright NK-cell subset, probably due to limited production of its
precursors, and with the expansion of highly differentiated mature
CD57+ CD56dim CD16+ and dysfunctional CD56− CD16+ NK cells.
Even if the majority of elderly people exhibit a normal NK-cell compartment, a minority of individuals show a breakdown of NK-cell
repertoire diversity which might influence immune surveillance.
This is particularly relevant in the context of cancer development,
which is negatively associated with the level of NK-cell-mediated
cytotoxicity [112]. Both the failure to replenish the naı̈ve NK-cell
pool, due to either inefficient NK-cell differentiation or a highly
skewed NK-cell repertoire caused by the selective expansion of
virus-specific NK cells, might impair NK-cell function in the elderly.
Aging of B-cell function
The number of circulating B cells has been shown to significantly
decrease with age, and changes in the relative frequencies of the
different B-cell subsets have been reported. The evaluation of specific subsets is complicated not only by variations between individuals but also by the use of different phenotyping protocols. There is
general consensus, however, that by using anti-CD19, -CD27 and
-IgD antibodies, it is possible to identify four major circulating
B-cell subsets: naı̈ve [IgD+ CD27− ], IgM memory [IgD+ CD27+ ],
switched memory [IgD− CD27+ ], and late/exhausted memory
[IgD− CD27− ] {reviewed in [113]}. Using these markers, it has
been shown that the percentage of switched memory B cells, the
predictors of optimal antibody responses [114], decreases with
age [115, 116]. Conversely, the percentage of late/exhausted
memory B cells, the antigen-experienced and pro-inflammatory Bcell subset, increases with age [117, 118]. The term “exhausted”
indicates terminally differentiated, senescent cells expressing the
cell cycle regulator p16INK4 , which decelerates cell progression
from G1 to S and induces cell cycle arrest. Late/exhausted memory B cells also have shorter telomeres [117, 118]. In addition,
they secrete pro-inflammatory cytokines before stimulation and
for this reason they are pre-activated and “refractory” to undergo
in vitro class switch when stimulated with antigens and mitogens,
as explained below. Moreover, it has been shown that bone marrow from old patients contains a low numebr of plasma cells [119].
Aging decreases antibody responses to exogenous antigens and
vaccines, leading to greater susceptibility of elderly individuals
to infectious diseases such as influenza. Functional alterations in
T cells have been considered the most significant contributors to
immunosenescence and sufficient per se to explain the age-related
decrease in antibody responses of elderly individuals. However,
some studies have managed to analyze defects in a variety of
components of the innate and adaptive immune systems which
occur with age. For example, in the case of influenza vaccination,
the following defects have been characterized: decreased T-cell
function and loss of CD28 expression, reduced specificity and class
of antibody produced and decreased memory B cells, reduced
www.eji-journal.eu
2291
2292
Marcello Pinti et al.
natural killer cell cytotoxicity on a per cell basis, and reduced
number and/or function of circulating dendritic cells (reviewed
in [120]).
Age-related intrinsic B-cell defects, responsible for sub-optimal
antibody responses in elderly individuals to infections and vaccines have been identified [95, 121–123], and include decreases
in expression and function of the key transcription factor E47
(see below), along with a reduction in activation-induced cytidine deaminase (AID), the enzyme of class switch recombination
and somatic hypermutation. AID is a measure of optimal B-cell
responses and its decreased expression in B cells from elderly
individuals has been shown to lead to a reduced ability to generate higher affinity protective antibodies [124]. For example, the
serum antibody response to both seasonal and pandemic influenza
vaccines, as well as the in vitro B-cell response after vaccination,
are both decreased with increasing age and are significantly correlated [114, 124, 125]. AID expression is also significantly correlated with antibody affinity maturation for the HA1 globular
domain of the pandemic (p)H1N1 HA, as measured by antibodyantigen complex dissociation rates and Surface Plasmon Resonance [125].
AID is transcriptionally regulated by E47, a class I basic helixloop-helix protein encoded by the E2A gene. E47 mRNA expression
has also been shown to be decreased in B cells from elderly individuals [115]. The reduced E47 and AID mRNA expression levels in B
cells from elderly individuals are due to reduced mRNA stability,
which is in turn due to the higher expression of the inflammatory
micro-RNAs (miRs) 16 and 155, which bind to the 3’-untranslated
region of E47 and AID mRNA, respectively, inducing mRNA degradation [115]. Inflammaging not only induces higher expression
of inflammatory miRs, but has also been shown to drive TNF-α
expression in B cells from elderly individuals, and these levels are
positively correlated with serum TNF-α and negatively correlated
with the response of the same B cells after in vitro stimulation,
which is measured by AID [121].
The higher levels of serum and B-cell-intrinsic TNF-α observed
in elderly individuals have been associated with the age-related
increase in CMV seropositivity. TNF-α activates the immediateearly promoter/enhancer of CMV, creating a “vicious cycle” in
which the production of pro-inflammatory cytokines is increased.
CMV may down-regulate B-cell responses either directly through
TNF-α [118] or indirectly through the induction of terminally differentiated T cells and senescent T cells [126], and reduced generation of memory T cells [127].
Elderly individuals have a significant reduction in B-cell
repertoire diversity and this correlates with their health status
[128, 129]. Influenza and pneumococcal vaccine-induced expansion of B cells with short and hydrophilic IgH CDR3 regions is
lower in older individuals, and the impaired anti-pneumococcal
IgM and IgA responses correlates with the spectratypes for their
IgM- and IgA-expressing B cells [130]. Moreover, elderly individuals have decreased numbers of B-cell lineages but increased prevaccination mutation load in their repertoire, resulting in a less
efficient response, and the diversity of the lineages is thus greatly
reduced as compared with that in young individuals [131].
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Eur. J. Immunol. 2016. 46: 2286–2301
Altered signaling modifies cell functions
In almost all immune cells, one of the most prominent changes
occurring with aging regards signaling, that is integrating all the
molecular events converging from the surface receptors to an
adequate cellular response [132]. The modifications in the numbers of the most common immune receptors, i.e., TLR, Fcγ, and
chemokine receptors, are controversial, even if, with some exceptions, the number itself is not changing [102]. Studies show that
the signaling pathways are generally altered either in their proximal events (such as those including MyD88, PI3K, Lyn) or at
the distal events (such as those including NFkB) [133]. JAK, Erk
and PI3K represent the three most important pathways which are
strictly interconnected. Their dysregulation can lead to altered
chemotaxis, free radical production, killing in neutrophils and
monocytes/macrophages, and reduced chemotaxis and antigen
presentation in DCs [reviewed in [102]). For example, it has been
recently demonstrated that the inhibition of PI3K, whose signaling
in resting human neutrophils is constitutively increased in elderly
subjects, significantly improves their functions by restoring neutrophil migratory accuracy [39]. The causes of such alterations are
numerous, ranging from the hostile inflammatory milieu, leading
to an increased basal level of cell activation, to intrinsic reasons,
such as membrane alterations, or the disequilibrium between feed
forward pathways and the inhibitory feedback loops [134].
In order to achieve an optimal T-cell response, the coordinated action of surface receptors and various signaling pathways,
including the metabolic ones, is required. Several studies found
age-related alterations in T-cell signaling pathways [135], including impairment of PTK phosphorylation, decreased Ca2+ mobilization, and lowered PKC, PI3K and MAPK activation [136]. TCR
density remains unchanged, but CD28 decreases by 20%-30% during aging, likely due to increased plasma levels of TNF-α. These
alterations lead to decreased activity of the transcription factors
NF-κB and NF-AT [137].
PTK Lck is obligatory for initiation of TCR signaling. Its activity is finely tuned by a multiple component module, comprising PTPase CD45 and PTK Csk bound to scaffold protein PAG
(CBP). Lck activity cycles between primed, active and inactive
states. Dysregulation of the Csk/PAG/CD45 loop in aged T cells
favors the inactive form of Lck [138], providing a molecular clue
to altered T-cell responses in aging. Negative feedback inhibitory
events are also compromised during aging. For instance, SHP1 activity has been shown to be higher in healthy elderly subjects than in young individuals, an observation consistent with the
decreased T-cell response. Importantly, pharmacological inhibition of SHP-1 resulted in recovery of TCR/CD28-dependent lymphocyte proliferation and IL-2 production, suggesting the possibility of improving T-cell responses in the healthy elderly [138].
Furthermore, Li et al. [81] identified an age-associated defect
in T-cell receptor (TCR)-induced ERK phosphorylation in naive
CD4+ T cells. The defective ERK signaling was caused by the
dual specific phosphatase 6 (DUSP6), whose protein expression
increased with age due to a decline in repression by miR-181a.
Reconstitution of miR-181a lowered DUSP6 expression in naive
www.eji-journal.eu
Eur. J. Immunol. 2016. 46: 2286–2301
CD4+ T cells in elderly individuals [81]. DUSP6 repression using
miR-181a or specific siRNA and DUSP6 improved CD4+ T-cell
responses, such as increased expression of activation markers,
improved proliferation and supported preferential T helper type 1
cell differentiation. Intrinsic alterations have been demonstrated
at the level the T-cell membrane, as the cholesterol content in the
membrane was found to be increased, interfering with the coalescence of the lipid rafts that are necessary for adequate signaling
[139].
The redox state of the cell also strongly influences T-cell signaling. Activation of CD28 has been shown to result in decreased
levels of reduced glutathione (GSH) and increased levels in cytosolic ROS [140]. In T cells from aged individuals, ROS remain
high [141]. High ROS levels in T cells can inhibit TCR signaling through lowered expression of TCR/CD3, diminished phosphorylation of ZAP70 and altered Ca2+ mobilization [136]. The
persistence of low amounts of pro-inflammatory cytokines, concomitant with increased production of ROS, both of which are
features of inflammaging, converge to diminish T-cell function in
older persons, in the form of reduced IL-2 production and clonal
expansion/proliferation [142].
Alterations in T-cell activation in the healthy elderly may also
result from accumulation of memory T cells [143, 144]. Recently
progresses have been made in linking of the development of
the memory phenotype signaling, and the concomitant cellular
metabolism orchestrated by the mTOR pathways [145]. It is now
accepted that the memory phenotype is emerging because of persistent activation of the MAPK p38. The fundamental metabolic
requirements of senescent primary human CD8+ T cells were elucidated in a recent study, where it was shown that p38 MAPK
blockade could reverse CD8+ T-cell senescence via a mTORindependent pathway, i.e., via the autophagy pathway [92]. Inhibition of mTOR has been shown to increase the general immune
response to vaccination in the elderly [145], and might be relevant
in designing new therapeutic strategies. Finally, T-cell metabolism
also drives the differentiation of TH1 cells to various other subsets [146], but data are lacking on the behavior of these cells from
aged individuals.
Immune aging and vaccinating the elderly
The fact that vaccines are the most effective measure to prevent infectious disease is widely accepted in the pediatric setting, and tremendous progress has been achieved in developing
novel and improved vaccines for children over the last years.
There is still a great need for vaccines tailored to optimally stimulate the aged immune system, as the elderly suffer more frequently from severe infections and experience poorer outcomes
from these infections as compared to younger adults [147]. However, vaccine recommendations for the elderly vary from country
to country and include vaccination against influenza, Streptococcus pneumoniae and Herpes zoster as well as booster vaccinations
against tetanus/diphtheria, and in some cases pertussis and polio
(Table 1).
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
HIGHLIGHTS
Vaccine-induced immune responses are frequently lower in
the elderly compared to younger adults. In most studies, antibody concentrations are measured to determine immunogenicity
of vaccines, but lower antibody responses cannot be attributed
solely to defects in B-cell function. Age-related changes in antigen
uptake, processing and presentation, as well as functional defects
of T cells, also lead to reduced antibody responses [148, 149]. In
addition to cell-intrinsic defects, inflammaging can also contribute
to impaired vaccine responses, as measured by antibody production.
The immunogenicity of subunit and split influenza vaccines is
usually measured by the hemagglutination inhibition assay (HAI),
and has been shown to generate lower results in the elderly compared with responses from younger adults. A meta-analysis of
more than 30 studies demonstrated unadjusted odds ratios (OR) of
0.48 (95% CI: 0.41-0.55 for H1N1 Ag); 0.63 (0.55-0.73; H3N2 Ag)
and 0.38 (0.33-0.44; B Ag) for seroconversion (HAI titer increase
ࣙ 4-fold) and 0.47 (0.40-0.55; H1N1 Ag), 0.53 (0.45-0.63; H2N3
Ag), and 0.58 (0.50-0.67; B Ag) for seroprotection (HAI ࣙ40) in
a comparison of elderly versus young adults [150].
In elderly patients, frailty, a multifactorial syndrome characterized by reduced stress resistance and physiological reserve
[151, 152], and associated with increased serum levels of IL6 [153], has been shown to impact susceptibility to influenza
and responsiveness to influenza vaccine [154]. Expression profiles predicting vaccination responses have been investigated prior
to and in the early phase after influenza vaccination [155]. Prevaccination expression of genes associated with T-cell and Bcell function were positively correlated with influenza-specific
antibody responses, while monocyte- and inflammation-related
genes were negatively correlated with influenza-specific antibody
responses, supporting the concept that inflammatory responses
at baseline might be detrimental to vaccine-induced antibody
responses [155]. It has been shown in mice that the inflammatory
condition associated with obesity limits the antibody response to,
and efficacy of, influenza vaccination [156]. Decreased influenzaspecific antibody levels and B-cell function have also been
described for obese humans [157]. It has been suggested that
elevated baseline inflammation may aggravate or cause intrinsic
defects in T cells and B cells, hampering their responses to antigenic stimulation (summarized in [158]). Cell-mediated immunity, namely the cytolytic activity of CD8+ T cells after vaccination, is also lower in the elderly [159]. Various strategies have been
pursued in order to improve vaccine-elicited antibody responses
in the elderly, leading to the licensing of several novel vaccines
against influenza. These include an intradermal vaccine [160],
a high-dose vaccine [161], and a vaccine adjuvanted with the
oil-in-water emulsion MF59, containing a synthetic muramyl peptide which has been shown to possess low toxicity and significant
immunostimulatory activity in humans [162]. All of the above
formulations show slightly higher immunogenicity in the elderly
compared to the standard trivalent inactivated vaccine. The MF59adjuvanted vaccine also induces more antibodies against heterologous viral strains compared to the standard influenza vaccine
[163].
www.eji-journal.eu
2293
2294
Marcello Pinti et al.
Eur. J. Immunol. 2016. 46: 2286–2301
Table 1. Recommendations for vaccination of adults and older adults in selected countries for 2015
Country
USA
Germany
Austria
UK
Italy
Guideline
Influenza
[a]
Annually
[b]
Annually >60
[c]
Annually,
particularly
>50
[d]
Annually >65
S. pneumoniaea)
Once >50,
PCV13, after 1
year PPV23
Once >60
PPV23
Once >50
PCV13, after 1
year PPV23
Once >65
PPV23
Herpes zoster
Diphtheriab)
Once >60
Every 10 years
−
Every 10 years
once >70
−
Tetanusb)
Every 10 years
Every 10 years
−
every 10 years
Pertussis
(acellular)b)
Once during
adulthood
Once during
adulthood
−
every 10 years
Polio
(inactivated)b)
−
−
Once >50
Every 10
years, >60
every 5 years
Every 10
years, >60
every 5 years
Every 10
years, >60
every 5 years
Every 10
years, >60
every 5 years
[e]
Annually for
all adults,
particularly
>65
Once >65
PCV13,
followed by
PPV23
once >60
every 10 years
−
a)
for persons without prior vaccination with PCV13 or PPV23.
for persons with adequate primary vaccination earlier in life.
a. Recommended Adult Immunization Schedule United States. 2015 http://www.cdc.gov/vaccines/schedules/downloads/adult/adult-schedule.pdf.
Accessed 15-1-2016
b. Empfehlungen der Ständigen Impfkommision (STIKO) am Robert-Koch-Institut. 2015 http://www.rki.de/DE/Content/Infekt/EpidBull/Archiv/2015/
Ausgaben/34_15.pdf?__blob = publicationFile. Accessed 15-1-2016
c. Impfplan Österreich. 2015 http://bmg.gv.at/cms/home/attachments/8/9/4/CH1100/CMS1389365860013/impfplan.pdf. Accessed 15-1-2016
d. Complete Immunisation Schedule UK. 2015 https://www.gov.uk/government/uploads/system/uploads/attachment_data/file/473570/9406_PHE_
2015_Complete_Immunisation_Schedule_A4_21.pdf. Accessed 15-1-2016
e. Piano Nazionale Prevenzione Vaccinale. 2015 http://www.quotidianosanita.it/allegati/allegato1955037.pdf. Accessed 15-1-2016
General recommendations for age groups are shown, while additional recommendations for specific risk groups, e.g. persons with underlying
diseases, are not included here and can be found in the cited documents.
b)
Studies analyzing clinical efficacy or effectiveness against
influenza are difficult to compare as their outcome depends heavily on the study population, on read-out parameters, and on epidemiological parameters (such as prevalence and virulence of the
virus, the degree of mismatch between the vaccine strains and
circulating virus strains, among others). Meta-analyses have estimated clinical efficacy and/or effectiveness of influenza vaccine
and it can be concluded that protection is lower in the elderly
than in young adults [164, 165]. Substantial research has been
performed in order to develop a universal influenza vaccine which
would be able to protect from all strains of influenza by inducing
broad, long-lasting immune responses, and which could solve the
issue of annual re-vaccination. Several viral proteins have been
suggested as candidate antigens and a variety of delivery platforms, such as viral vectors, adjuvants or DNA vaccines have been
tested [166].
A 23-valent polysaccharide vaccine against Streptococcus pneumoniae has been used for many years in the older population.
Meta-analyses reported efficacy against invasive disease, but the
efficacy against pneumonia is frequently in doubt, as results from
clinical studies are inconclusive [167]. Recently, a 13-valent con
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
jugate vaccine has been licensed for adults and its clinical efficacy was 45.6% (95.2% CI: 21.8–62.5; p < 0.001) for confirmed vaccine-type community-acquired pneumonia and 75.0%
(95% CI: 41.4–90.8; p < 0.001) for vaccine-type invasive disease in a large randomized placebo-controlled study enrolling
more than 84 000 elderly persons [168]. Data from this study
were used to analyze the effect of age on vaccine efficacy using a
statistical model, and a decrease of vaccine efficacy for vaccinetype community acquired pneumonia and invasive disease from
65% (95% CI: 38–81) in 65-year-old subjects, to 40% (95% CI: 17–
56) in 75-year-old subjects was determined [169].
The incidence of herpes zoster, caused by reactivation of the
varicella zoster virus, increases with age. A live-attenuated vaccine is available, which has been shown to reduce the incidence
of herpes zoster by 51.3% (95% CI: 44.2–57.6) and of postherpetic neuralgia, a severe complication occurring frequently in
the elderly, by 66.5% (95% CI: 44.5–79.2) in the vaccinated population compared to placebo [170]. The protective effect against
post-herpetic neuralgia was independent of age, whereas clinical
efficacy against herpes zoster declined to only 27.6% in persons
older than 69 years. The current live-attenuated vaccine cannot
www.eji-journal.eu
HIGHLIGHTS
Eur. J. Immunol. 2016. 46: 2286–2301
be used to vaccinate immunocompromised persons, who are at
great risk of herpes zoster also at a younger age. A novel, inactivated vaccine containing the viral glycoprotein E, adjuvanted
with the liposome-based AS01B system (MPL and QS21), has
recently been developed [171]. In a phase III randomized placebocontrolled trial clinical efficacy against herpes zoster was 97.2%
(95% CI: 93.7–99.0; p < 0.001) for persons over the age of 50
and did not decrease for older age groups (>70 years) [171].
The AS01B adjuvant system efficiently induces IgE-specific CD4+
T cells [172] and cell-mediated immunity; antibody responses are
also higher compared with those induced by the live-attenuated
vaccine [173]. Experiments in mice showed that adjuvants containing MPL and QS21 rapidly induce chemokines and cytokines at
the intramuscular injection site, attracting monocytes and granulocytes [174]. Increased numbers of neutrophils, monocytes, and
DCs were also observed in the draining lymph node and it has
been hypothesized that the adjuvant-mediated recruitment and
activation of APCs, particularly of MHCIIhigh DCs, is responsible
for the efficient stimulation of adaptive immune responses. However, these results have been obtained in young mice, and it is
still not known if this mechanism is preserved in aged animals
[174].
Regular booster vaccination against tetanus and diphtheria
throughout life is recommended in many countries. However, the
levels of tetanus- and to an even greater extent diphtheria-specific
antibodies in the elderly are frequently below that considered to
be protective [175]. Single booster shots late in life were shown
not to elicit long-lasting antibody responses against diphtheria in
a substantial proportion of the elderly [176]. Appropriate vaccination documentation is crucial to timely deliver booster vaccinations, but is often poorly documented in the elderly. Epidemiological data show that pertussis is relevant for older age groups and
does not solely affect infants [177]. Some countries recommend
regular booster vaccination against pertussis in combination with
the tetanus/diphtheria vaccine or at least one booster shot during
adulthood. Immunogenicity of the vaccine is lower in the elderly
compared with that in younger adults [177].
Substantial effort is put into the development of vaccines
against several pathogens that are of relevance for the elderly. Particularly, persons with underlying diseases and frail elderly have a
risk of severe disease caused by respiratory syncytial virus (RSV).
Estimations in the United Kingdom reported up to 18 000 hospitalizations and 8400 deaths caused by RSV per season with 79%
of hospitalizations and 93% of deaths in persons older than 65
years [178]. Several vaccine candidates against RSV are currently
in early clinical development [179], and these vaccine candidates
should also be tested in adults and the elderly. Vaccines against
nosocomial pathogens such as Staphylococcus aureus, Clostridium
difficile, Escherichia coli, Klebsiella pneumoniae, and Candida ssp.
could provide great benefit for the elderly, as this age group has
a high risk of hospitalization and nosocomial infections. Clinical development is ongoing for several vaccine candidates [180],
and successful vaccination against nosocomial pathogens has the
potential to save many lives and to substantially reduce healthcare
costs.
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Conclusion
The increase in human lifespan poses several new questions and
complex challenges to the medical and scientific community,
including for immunologists. Today, the immune system has to
defend the organism for several decades, and thus has to work
effectively for a substantial number of years; this is a reality that
was not considered when Jenner developed the smallpox vaccine. Moreover, every day immune cells have to cope with external insults (such as oxygen, UV light, chronic infection), personal
and social behaviors (nutrition, obesity, psychological stress, lack
of exercise, hyper-training, pollution, smoking, economic status)
and unavoidable internal changes (cell metabolism, turnover and
production of DAMPs). Our community is well aware of this challenge, and indeed an unprecedented attention is now paid to aging
and longevity, that includes the search for new strategies for an
optimal maintenance of immunological performances in the long,
last part of our life.
Acknowledgements: M.P. is supported by Fondazione Cassa
di Risparmio di Vignola (Italy); V.A. and D.S. are supported
by the French Agence Nationale de la Recherche (ANR; project
ANR-14-CE14-0030-01) and the Fondation Recherche Médicale
(project DEQ20120323690); D.F. is supported by NIH grants
R21 AI096446, R21 AG042826, and R56 AG032576; T.F. has
received grants from by the Canadian Institutes of Health Research
(CIHR) (No. 106634 and 106701), the Université de Sherbrooke,
and the Research Center on Aging; A.L. is supported by SIgN
and the Agency for Science Technology and Research (JCO DP
#1434m00115 and SRIS SRG/14018); B.W. has received funding from the European Union’s Seventh Framework Programme
[FP7/2007-2013] under Grant Agreement No: 280873 ADITEC;
A.C. has been supported by Ministero dell’Istruzione, Università,
Ricerca (MIUR grant RBAP11S8C3).
Conflict of interest: The authors declare no financial or commercial conflict of interest.
References
1 Olshansky, S. J., Perry, D., Miller, R. A. and Butler, R. N., Pursuing the
longevity dividend: scientific goals for an aging world. Ann. N.Y. Acad.
Sci. 2007. 1114: 11–13.
2 The Silver Book. Chronic disease and medical innovation in an aging
nation. Published by the Alliance for Aging Research, Updated May,
2011 (http://www.silverbook.org).
3 López-Otı́n, C., Blasco, M. A., Partridge, L., Serrano, M. and Kroemer, G.,
The hallmarks of aging. Cell 2013. 153:1194–1217.
4 Cevenini, E., Monti, D. and Franceschi, C., Inflamm-ageing. Curr. Opin.
Clin. Nutr. Metab. Care 2013. 18: 14–20.
www.eji-journal.eu
2295
2296
Marcello Pinti et al.
Eur. J. Immunol. 2016. 46: 2286–2301
5 Finch, C. E., Morgan, T. E., Longo, V. D. and de Magalhaes, J. P., Cell
23 Rodier, F., Coppé, J. P., Patil, C. K., Hoeijmakers, W. A., Munoz, D. P.,
resilience in species lifespans: a link to inflammation? Aging Cell 2010.
Raza, S. R., Freund, A. et al., Persistent DNA damage signalling triggers
9:519–526.
senescence-associated inflammatory cytokine secretion. Nat. Cell Biol.
6 Fagiolo, U., Cossarizza, A., Scala, E., Fanales-Belasio, E., Ortolani, C.,
Cozzi, E., Monti, D. et al., Increased cytokine production in mononuclear
cells of healthy elderly people. Eur. J. Immunol. 1993. 23: 2375–2378.
7 Puzianowska-Kuźnicka M., Owczarz M., Wieczorowska-Tobis, K.,
2009. 11: 973–979.
24 Wiley, C. D., Velarde, M. C., Lecot, P., Liu, S., Sarnoski, E. A., Freund,
A., Shirakawa, K. et al., Mitochondrial dysfunction induces senescence
with a distinct secretory phenotype. Cell Metab. 2016. 23:303–314.
Nadrowski, P., Chudek, J., Slusarczyk, P., Skalska, A. et al., Interleukin-
25 Arita, Y., Kihara, S., Ouchi, N., Takahashi, M., Maeda, K., Miyagawa,
6 and C-reactive protein, successful aging, and mortality: the PolSenior
J., Hotta, K. et al., Paradoxical decrease of an adipose-specific protein,
study. Immun Ageing. 2016. 3: 13–21.
adiponectin, in obesity. Biochem Biophys Res Commun. 1999. 257: 79–83.
8 Brenchley, J. M., Price, D. A., Schacker, T. W., Asher, T. E., Silvestri, G.,
26 Ohashi, K., Parker, J. L., Ouchi, N., Higuchi, A., Vita, J. A., Gokce, N.,
Rao, S., Kazzaz, Z. et al., Microbial translocation is a cause of systemic
Pedersen, A. A. et al., Adiponectin promotes macrophage polarization
immune activation in chronic HIV infection. Nat Med. 2006. 12: 1365–71.
9 Biagi, E., Candela, M., Fairweather-Tait, S., Franceschi, C. and Brigidi,
P., Ageing of the human metaorganism: the microbial counterpart. Age
2012. 34: 247–267.
10 Heintz, C. and Mair, W., You are what you host: microbiome modulation
of the aging process. Cell 2014. 156: 408–411.
11 Mueller, S., Saunier, K., Hanisch, C., Norin, E., Alm, L., Midtvedt, T.,
Cresci, A. et al., Differences in fecal microbiota in different European
study populations in relation to age, gender, and country: a crosssectional study. Appl Environ Microbiol. 2006. 72: 1027–33.
12 Woodmansey E. J., McMurdo, M. E., Macfarlane, G. T. and Macfarlane,
S., Comparison of compositions and metabolic activities of fecal microbiotas in young adults and in antibiotic-treated and non-antibiotictreated elderly subjects.) Appl Environ Microbiol 2004. 70: 6113–6122
13 Biagi, E., Nylund, L., Candela, M., Ostan, R., Bucci, L., Pini, E., Nikkı̈la,
J. et al., Through ageing, and beyond: gut microbiota and inflammatory
status in seniors and centenarians. PLoS One 2010. 5: e10667.
14 Campisi, J. and d’Adda di Fagagna, F., Cellular senescence: when bad
things happen to good cells. Nature Rev. Molec. Cell Biol. 2007. 8:729–740.
15 Campisi, J., Senescent cells, tumor suppression and organismal aging:
good citizens, bad neighbors. Cell 2005. 120: 513–522.
16 Coppé, J. P., Patil, C. K., Rodier, F., Sun, Y., Munoz, D., Goldstein, J., Nelson, P. S. et al., Senescence-associated secretory phenotypes reveal cell
non-automous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008. 6: 2853–2868.
17 Mantovani, A., Chemokines in neoplastic progression. Semin. Cancer Biol.
2004. 14: 147–148.
18 Xue, W., Zender, L., Miething, C., Dickins, R. A., Hernando, E.,
Krizhanovsky, V., Cordon-Cardo, C. and Lowe, S. W., Senescence and
tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007. 445: 656–660.
19 Coppé, J. P., Desprez, P. Y., Krtolica, A. and Campisi, J., The senescenceassociated secretory phenotype: the dark side of tumor suppression.
Annu. Rev. Pathol. 2010. 5: 99–118.
20 Demaria, M., Ohtani, B., Youssef, S. A., Rodier, F., Toussaint, W.,
Mitchell, J. R., Laberge, R. M. et al., An essential role for senescent cells
in optimal wound healing through secretion of PDGF-AA. Dev. Cell 2014.
31:722–733.
21 Storer, M., Mas, A., Robert-Moreno, A., Pecoraro, M., Ortells, M. C., Di
Giacomo, V., Yosef, R. et al., Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 2013.
155:1119–1130.
22 Baker, D. J., Childs, B. G., Durik, M., Wijers, M. E., Sieben, C. J., Zhong, J.,
Saltness, R. et al., Naturally occurring p16Ink4a-positive cells shorten
healthy lifespan. Nature 2016. 530: 184–189.
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
toward an anti-inflammatory phenotype. J Biol Chem. 2010. 285: 6153–
6160.
27 La Cava, A. and Matarese, G., The weight of leptin in immunity. Nat Rev
Immunol. 2004. 4: 371–379.
28 Sarraf, P., Frederich, R. C., Turner, E. M., Ma, G., Jaskowiak, N. T., Rivet,
D. J., 3rd, Flier, J. S. et al., Multiple cytokines and acute inflammation
raise mouse leptin levels: potential role in inflammatory anorexia. J Exp
Med. 1997. 185: 171–175.
29 Lumeng, C. N., Liu, J., Geletka, L., Delaney, C., Delproposto, J., Desai,
A., Oatmen, K. et al., Aging is associated with an increase in T cells and
inflammatory macrophages in visceral adipose tissue. J Immunol. 2011.
187: 6208–6216.
30 Sewter, C. P., Digby, J. E., Blows, F., Prins, J. and O’Rahilly S., Regulation
of tumour necrosis factor-alpha release from human adipose tissue in
vitro. J Endocrinol. 1999. 163: 33–38.
31 Park, H. S., Park, J. Y. and Yu, R., Relationship of obesity and visceral adiposity with serum concentrations of CRP, TNF-alpha and IL-6. Diabetes
Res Clin Pract. 2005. 69: 29–35.
32 Mohamed-Ali, V., Goodrick, S., Rawesh, A., Katz, D. R., Miles, J. M.,
Yudkin, J. S., Klein, S. et al., Subcutaneous adipose tissue releases
interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin
Endocrinol Metab. 1997. 82: 4196–4200.
33 Simpson, R. J., Lowder, T. W., Spielmann, G., Bigley, A. B., LaVoy, E. C.,
Kunz, H., Exercise and the aging immune system. Ageing Res Rev. 2012.
11: 404–420.
34 Tchkonia, T., Morbeck, D. E., Von Zglinicki, T., Van Deursen, J., Lustgarten, J., Scrable, H., Khosla, S. et al., Fat tissue, aging, and cellular
senescence. Aging Cell. 2010. 9: 667–684.
35 Cossarizza, A., Ortolani, C., Paganelli, R., Barbieri, D., Monti, D., Sansoni, P., Fagiolo, U. et al., CD45 isoforms expression on CD4+ and CD8+
T cells throughout life, from newborns to healthy centenarians: implications for T cell memory. Mech Ageing Develop. 1996. 86: 173–195.
36 Pinti, M., Cevenini, E., Nasi, M., De Biasi, S., Salvioli, S., Monti, D.,
Benatti, S. et al., Circulating mitochondrial DNA increases with age and
is a familiar trait: implications for "inflamm-aging". Eur. J. Immunol. 2014.
44: 1552–1562.
37 Butcher, S. K., Chahal, H., Nayak, L., Sinclair, A., Henriquez, N. V.,
Sapey, E., O’Mahony, D. et al., Senescence in innate immune responses:
reduced neutrophil phagocytic capacity and CD16 expression in elderly
humans. J. Leukocyte Biol. 2001. 70: 881–886.
38 Brubaker, A. L., Rendon, J. L., Ramirez, L., Choudhry, M. A. and Kovacs,
E. J., Reduced neutrophil chemotaxis and infiltration contributes to
delayed resolution of cutaneous wound infection with advanced age.
J. Immunol. 2013. 190: 1746–1757.
39 Sapey, E., Greenwood, H., Walton, G., Mann, E., Love, A., Aaronson,
N., Insall, R. H. et al., Phosphoinositide 3-kinase inhibition restores
www.eji-journal.eu
HIGHLIGHTS
Eur. J. Immunol. 2016. 46: 2286–2301
neutrophil accuracy in the elderly: toward targeted treatments for
immunosenescence. Blood 2014. 123: 239–248.
tion of human Langerhans cells: the influence of ageing. Brit.J. Dermatol.
2002. 146: 32–40.
40 Nomellini, V., Brubaker, A. L., Mahbub, S., Palmer, J. L., Gomez, C. R.
57 Zavala, W. D. and Cavicchia, J. C., Deterioration of the Langerhans cell
and Kovacs, E. J., Dysregulation of neutrophil CXCR2 and pulmonary
network of the human gingival epithelium with aging. Arch. Oral Biol.
endothelial ICAM-1 promotes age-related pulmonary inflammation.
2006. 51: 1150–1155.
Aging Dis. 2012. 3: 234–47.
41 Emanuelli, G., Lanzio, M., Anfossi, T., Romano, S., Anfossi, G. and Calcamuggi, G., Influence of age on polymorphonuclear leukocytes in vitro:
phagocytic activity in healthy human subjects. Gerontology 1986. 32: 308–
316.
42 Alonso-Fernandez, P., Puerto, M., Mate, I., Ribera, J. M. and de la Fuente,
M., Neutrophils of centenarians show function levels similar to those
of young adults. J. Am. Geriatr. Soc. 2008. 56: 2244–2251.
43 Tseng, C. W., Kyme, P. A., Arruda, A., Ramanujan, V. K., Tawackoli,
W. and Liu, G. Y., Innate immune dysfunctions in aged mice facilitate
the systemic dissemination of methicillin-resistant S. aureus. PLoS One
2012. 7: e41454.
44 Metcalf, T. U., Cubas, R. A., Ghneim, K., Cartwright, M. J., Grevenynghe,
J. V., Richner, J. M., Olagnier, D. P. et al., Global analyses revealed agerelated alterations in innate immune responses after stimulation of
pathogen recognition receptors. Aging Cell 2015. 14: 421–432.
45 Ziegler-Heitbrock, L., Blood monocytes and their subsets: established
features and open questions. Front. Immunol. 2015. 6: 423.
58 Laube, S., Skin infections and ageing. Ageing Res. Rev. 2004. 3: 69–89.
59 Agrawal A., Agrawal, S., Cao, J. N., Su, H. F., Osann, K. and Gupta, S.,
Altered innate immune functioning of dendritic cells in elderly humans:
a role of phosphoinositide 3-kinase-signaling pathway. J. Immunol. 2007.
178: 6912–6922.
60 Janssen, N., Derhovanessian, E., Demuth, I., Arnaout, F., SteinhagenThiessen, E. and Pawelec, G., Responses of dendritic cells to TLR-4
stimulation are maintained in the elderly and resist the effects of CMV
infection seen in the young. J Gerontol A Biol Sci Med Sci 2016. 71: 1117–
1123.
61 do Nascimento, M. P., Pinke, K. H., Penitenti, M., Ikoma, M. R. and Lara,
V. S., Aging does not affect the ability of human monocyte-derived
dendritic cells to phagocytose Candida albicans. Aging Clin Exp Res 2015.
27: 785–789.
62 Sridharan, A., Esposo, M., Kaushal, K., Tay, J., Osann, K., Agrawal, S.,
Gupta, S. et al., Age-associated impaired plasmacytoid dendritic cell
functions lead to decreased CD4 and CD8 T cell immunity. Age 2011. 33:
363–376.
46 Hearps, A. C., Martin, G. E., Angelovich, T. A., Cheng, W. J., Maisa, A.,
63 Hart, P. H., Grimbaldeston, M. A., Hosszu, E. K., Swift, G. J., Noonan,
Landay, A. L., Jaworowski, A. et al., Aging is associated with chronic
F. P. and Finlay-Jones, J. J., Age-related changes in dermal mast cell
innate immune activation and dysregulation of monocyte phenotype
prevalence in BALB/c mice: functional importance and correlation with
and function. Aging Cell 2012. 11: 867–875.
47 Mclachlan, J. A., Serkin, C. D., Morrey, K. M. and Bakouche, O., Antitumoral properties of aged human monocytes. J.Immunol. 1995. 154: 832–
843.
dermal mast cell expression of Kit. Immunology 1999. 98: 352–356.
64 Nguyen, M., Pace, A. J. and Koller, B. H., Age-induced reprogramming
of mast cell degranulation. J Immunol 2005. 175: 5701–5707.
65 Atsuta, R., Akiyama, K., Shirasawa, T., Okumura, K., Fukuchi, Y.
48 van Duin, D., Mohanty, S., Thomas, V., Ginter, S., Montgomery, R. R.,
and Ra, C., Atopic asthma is dominant in elderly onset asthmatics:
Fikrig, E., Allore, H. G. et al., Age-associated defect in human TLR-1/2
possibility for an alteration of mast cell function by aging through
function. J. Immunol. 2007. 178: 970–975.
Fc receptor expression. Int Arch Allergy Immunol 1999. 120 Suppl 1:
49 Qian, F., Wang, X. M., Zhang, L., Chen, S., Piecychna, M., Allore,
76–81.
H., Bockenstedt, L. et al., Age-associated elevation in TLR5 leads to
66 Mathur, S. K., Schwantes, E. A., Jarjour, N. N. and Busse, W. W., Age-
increased inflammatory responses in the elderly. Aging Cell 2012. 11:
related changes in eosinophil function in human subjects. Chest 2008.
104–110.
50 Mizel, S. B. and Bates, J. T., Flagellin as an adjuvant: cellular mechanisms and potential. J. Immunol. 2010 185: 5677–5682.
51 Weinberger, B., Joos, C., Reed, S. G., Coler, R. and Grubeck-Loebenstein,
133: 412–419.
67 Ferrucci, L., Harris T. B., Guralnik J. M., Tracy R. P., Corti M. C., Cohen H.
J., Penninx B. et al., Serum IL-6 level and the development of disability
in older persons. J. Am. Geriatr. Soc. 1999 47:639–646.
B., The stimulatory effect of the TLR4-mediated adjuvant glucopyra-
68 Cohen, H. J., Harris, T. and Pieper, C. F., Coagulation and activation of
nosyl lipid A is well preserved in old age. Biogerontology 2016. 17: 177–
inflammatory pathways in the development of functional decline and
187.
52 Panda, A., Qian, F., Mohanty, S., van Duin, D., Newman, F. K., Zhang,
L., Chen, S. et al., Age-associated decrease in TLR function in primary
human dendritic cells predicts influenza vaccine response. J Immunol
2010. 184: 2518–2527.
53 Jing, Y., Shaheen, E., Drake, R. R., Chen, N., Gravenstein, S. and Deng,
Y., Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively
unaltered in human peripheral blood. Hum Immunol 2009. 70: 777–784.
54 Perez-Cabezas, B., Naranjo-Gomez, M., Fernandez, M. A., Grifols, J. R.,
Pujol-Borrell, R. and Borras, F. E., Reduced numbers of plasmacytoid
dendritic cells in aged blood donors. Exp Gerontol 2007. 42: 1033–1038.
55 Della Bella, S., Bierti, L., Presicce, P., Arienti, R., Valenti, M., Saresella,
M., Vergani, C. et al., Peripheral blood dendritic cells and monocytes
are differently regulated in the elderly. Clin Immunol 2007. 122: 220–228.
56 Bhushan, M., Cumberbatch, M., Dearman, R. J., Andrew, S. M., Kimber,
I. and Griffiths, C. E. M., Tumour necrosis factor-alpha-induced migra-
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
mortality in the elderly. Am. J. Med. 2003 114: 180–187.
69 Gruver, A. L., Hudson, L. L. and Sempowski, G. D., Immunosenescence
of ageing. J Pathol. 2007. 211: 144–156.
70 Pinti, M., Nasi, M., Lugli, E., Gibellini, L., Bertoncelli, L., Roat, E., De
Biasi, S. et al., T cell homeostasis in centenarians: from the thymus to
the periphery. Curr Pharm Des. 2010. 16:597–603.
71 Shanley, D. P., Aw, D., Manley, N. R. and Palmer D. B., An evolutionary
perspective on the mechanisms of immunosenescence. Trends Immunol.
2009. 30: 374–381
72 Nasi, M., Troiano, L., Lugli, E., Pinti, M., Ferraresi, R., Monterastelli, E.,
Mussi, C. et al., Thymic output and functionality of the IL-7/IL-7 receptor
system in centenarians: implications for the neolymphogenesis at the
extreme limit of human life. Aging Cell 2006. 5: 167–175.
73 Lugli, E., Pinti, M., Nasi, M., Troiano, L., Ferarresi, R., Mussi, C., Salvioli,
G. et al., Subject classification obtained by cluster analysis and principal
component analysis applied to flow cytometric data. Cytometry (part A)
2007. 71A: 334–344.
www.eji-journal.eu
2297
2298
Marcello Pinti et al.
74 Thome, J. J., Yudanin, N., Ohmura, Y., Kubota, M., Grinshpun, B.,
Sathaliyawala, T., Kato, T. et al., Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell 2014. 159:
814–828.
75 Sauce, D., Larsen, M., Fastenackels, S., Duperrier, A., Keller, M.,
Grubeck-Loebenstein, B., Ferrand, C. et al., Evidence of premature
immune aging in patients thymectomized during early childhood. J Clin
Invest 2009. 119: 3070–3078.
76 Zlamy, M., Almanzar, G., Parson, W., Schmidt, C., Leierer, J., Weinberger, B., Jeller, V. et al., Efforts of the human immune system to maintain the peripheral CD8+ T cell compartment after childhood thymectomy. Immun Ageing 2016. 13: 3.
77 Kohler, S. and Thiel, A., Life after the thymus: CD31+ and CD31- human
naive CD4+ T-cell subsets. Blood 2009. 113: 769–774.
Eur. J. Immunol. 2016. 46: 2286–2301
drives the senescence of human T cells. Nat. Immunol. 2014. 15: 965–
972.
91 Henson, S. M., Lanna, A., Riddell, N. E., Franzese, O., Macaulay, R.,
Griffiths, S. J., Puleston, D. J. et al., p38 signaling inhibits mTORC1independent autophagy in senescent human CD8(+) T cells. J. Clin.
Invest. 2014. 124: 4004–4016.
92 Henson, S. M., Macaulay, R., Riddell, N. E., Nunn, C. J. and Akbar, A.
N., Blockade of PD-1 or p38 MAP kinase signaling enhances senescent
human CD8(+) T-cell proliferation by distinct pathways. Eur. J. Immunol.
2015. 45: 1441–1451.
93 Puleston, D. J., Zhang, H., Powell, T. J., Lipina, E., Sims, S., Panse, I.,
Watson, A. S. et al., Autophagy is a critical regulator of memory CD8(+)
T cell formation. Elife 2014. 3: e03706.
94 Roux, A., Mourin, G., Larsen, M., Fastenackels, S., Urrutia, A., Gorochov,
78 Sauce, D., Larsen, M., Fastenackels, S., Roux, A., Gorochov, G., Katlama,
G., Autran, B. et al., Differential impact of age and cytomegalovirus
C., Sidi, D. et al., Lymphopenia-driven homeostatic regulation of naive
infection on the gammadelta T cell compartment. J. Immunol. 2013. 191:
T cells in elderly and thymectomized young adults. J. Immunol 2012. 189:
1300–1306.
5541–5548.
95 Wistuba-Hamprecht, K., Frasca, D., Blomberg, B., Pawelec, G. and Der-
79 Britanova, O. V., Putintseva, E. V., Shugay, M., Merzlyak, E. M., Tur-
hovanessian, E., Age-associated alterations in gammadelta T-cells are
chaninova, M. A., Staroverov, D. B., Bolotin, D. A. et al., Age-related
present predominantly in individuals infected with cytomegalovirus.
decrease in TCR repertoire diversity measured with deep and normal-
Immun. Ageing 2013. 10: 26.
ized sequence profiling. J. Immunol 2014. 192: 2689–2698.
80 Qi, Q., Liu, Y., Cheng, Y., Glanville, J., Zhang, D., Lee, J. Y., Olshen, R. A.
et al., Diversity and clonal selection in the human T-cell repertoire. Proc
Natl Acad Sci U S A. 2014. 111: 13139–13144.
81 Li, G., Yu, M., Lee, W. W., Tsang, M., Krishnan, E., Weyand, C. M. and
Goronzy, J. J., Decline in miR-181a expression with age impairs T cell
receptor sensitivity by increasing DUSP6 activity. Nat Med 2012. 18: 1518–
1524.
82 Briceno, O., Lissina, A., Wanke, K., Afonso, G., von Braun, A., Ragon, K.,
Miquel, T. et al., Reduced naive CD8(+) T-cell priming efficacy in elderly
adults. Aging Cell 2016. 15: 14–21.
83 Bignon, A., Regent, A., Klipfel, L., Desnoyer, A., de la Grange, P., Martinez, V., Lortholary, O. et al., DUSP4-mediated accelerated T-cell senescence in idiopathic CD4 lymphopenia. Blood 2015. 125: 2507–2518.
84 Richner, J. M., Gmyrek G. B., Govero, J., Tu, Y., van der Windt, G. J.,
Metcalf, T. U., Haddad, E. K. et al., Age-dependent cell trafficking defects
in draining lymph nodes impair adaptive immunity and control of West
Nile virus infection. PLoS Pathog. 2016. 11: e1005027.
85 Schulz, A. R., Malzer, J. N., Domingo, C., Jurchott, K., Grutzkau, A., Babel,
N., Nienen, M. et al., Low thymic activity and dendritic cell numbers
are associated with the immune response to primary viral infection in
elderly humans. J. Immunol 2015. 195: 4699–4711.
86 Papagno, L., Spina, C. A., Marchant, A., Salio, M., Rufer, N., Little, S.,
Dong, T. et al., Immune activation and CD8(+) T-cell differentiation
towards senescence in HIV-1 infection. PLoS Biol. 2004. 2: E20.
87 Brenchley, J. M., Karandikar, N. J., Betts, M. R., Ambrozak, D. R., Hill, B.
J., Crotty, L. E., Casazza, J. P. et al., Expression of CD57 defines replicative
senescence and antigen-induced apoptotic death of CD8+ T cells. Blood
2003. 101: 2711–2720.
88 Gil, A., Yassai, M. B., Naumov, Y. N. and Selin, L. K., Narrowing of
human influenza A virus-specific T cell receptor alpha and beta repertoires with increasing age. J. Virol. 2015. 89: 4102–4116.
89 Griffiths, S. J., Riddell, N. E., Masters, J., Libri, V., Henson, S. M.,
Wertheimer, A., Wallace, D. et al., Age-associated increase of lowavidity cytomegalovirus-specific CD8+ T cells that re-express CD45RA.
J. Immunol. 2013. 190: 5363–5372.
90 Lanna, A., Henson, S. M., Escors, D. and Akbar, A. N., The kinase
p38 activated by the metabolic regulator AMPK and scaffold TAB1
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
96 Cooper, M. A., Fehniger, T. A., Fuchs, A., Colonna, M. and Caligiuri, M.
A., NK cell and DC interactions. Trends Immunol. 2004. 25: 47–52.
97 Bjorkstrom, N. K., Ljunggren, H. G. and Sandberg, J. K., CD56 negative NK cells: origin, function, and role in chronic viral disease. Trends
Immunol. 2010. 31: 401–406.
98 Wang, J. W., Geiger, H. and Rudolph, K. L., Immunoaging induced by
hematopoietic stem cell aging. Curr. Opin. Immunol. 2011. 23: 532–536.
99 Le Garff-Tavernier, M., Beziat, V., Decocq, J., Siguret, V., Gandjbakhch,
F., Pautas, E., Debre, P. et al., Human NK cells display major phenotypic
and functional changes over the lifespan. Aging Cell 2010. 9: 527–535.
100 Gayoso, I., Sanchez-Correa, B., Campos, C., Alonso, C., Pera, A., Casado,
J. G., Morgado, S. et al., Immunosenescence of human natural killer
cells. J. Inn. Immunity 2011. 3: 337–343.
101 Mariani, E., Meneghetti, A., Neri, S., Ravaglia, G., Forti, P., Cattini, L.
and Facchini, A., Chemokine production by natural killer cells from
nonagenarians. Eur. J Immunol. 2002. 32: 1524–1529.
102 Solana, R., Tarazona, R., Gayoso, I., Lesur, O., Dupuis, G. and Fulop, T.,
Innate immunosenescence: effect of aging on cells and receptors of the
innate immune system in humans. Sem Immunol 2012. 24: 331–341.
103 Sanchez-Correa, B., Gayoso, I., Bergua, J. M., Casado, J. G., Morgado, S.,
Solana, R. and Tarazona, R., Decreased expression of DNAM-1 on NK
cells from acute myeloid leukemia patients. Immunol. Cell Biol. 2012. 90:
109–115.
104 Lutz, C. T., Karapetyan, A., Al-Attar, A., Shelton, B. J., Holt, K. J., Tucker,
J. H. and Presnell, S. R., Human NK cells proliferate and die in vivo more
rapidly than T cells in healthy young and elderly adults. J. Immunol. 2011.
186: 4590–4598.
105 Bayard, C., Lepetitcorps, H., Roux, A., Larsen, M., Fastenackels, S., Salle,
V., Vieillard, V. et al., Coordinated expansion of both memory T cells
and NK cells in response to CMV infection in humans. Eur. J. Immunol.
2016. 46: 1168–1179.
106 White, M. J., Nielsen, C. M., McGregor, R. H., Riley, E. M. and Goodier,
M. R., Differential activation of CD57-defined natural killer cell subsets
during recall responses to vaccine antigens. Immunology 2014. 142: 140–
150.
107 Bjorkstrom, N. K., Riese, P., Heuts, F., Andersson, S., Fauriat, C., Ivarsson, M. A., Bjorklund, A. T. et al., Expression patterns of NKG2A, KIR,
www.eji-journal.eu
HIGHLIGHTS
Eur. J. Immunol. 2016. 46: 2286–2301
and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. Blood 2010. 116: 3853–3864.
108 Zhang, Y., Wallace, D. L., de Lara, C. M., Ghattas, H., Asquith, B., Worth,
A., Griffin, G. E. et al., In vivo kinetics of human natural killer cells: the
effects of ageing and acute and chronic viral infection. Immunology 2007.
121: 258–265.
109 Lopez-Verges, S., Milush, J. M., Schwartz, B. S., Pando, M. J., Jarjoura, J.,
York, V. A., Houchins, J. P. et al., Expansion of a unique CD57(+)NKG2Chi
natural killer cell subset during acute human cytomegalovirus infection.
Proc Natl Acad Sci USA 2011. 108: 14725–14732.
110 Guma, M., Angulo, A., Vilches, C., Gomez-Lozano, N., Malats, N. and
Lopez-Botet, M., Imprint of human cytomegalovirus infection on the
NK cell receptor repertoire. Blood 2004. 104: 3664–3671.
111 Beziat, V., Liu, L. L., Malmberg, J. A., Ivarsson, M. A., Sohlberg, E., Bjorklund, A. T., Retiere, C. et al., NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve
activating KIRs. Blood 2013. 121: 2678–2688.
112 Waldhauer, I. and Steinle, A., NK cells and cancer immunosurveillance.
Oncogene 2008. 27: 5932–5943.
113 Ademokun, A., Wu, Y. C. and Dunn-Walters, D., The ageing B cell population: composition and function. Biogerontology 2010. 11: 125–137.
114 Frasca, D., Diaz, A., Romero, M., Phillips, M., Mendez, N. V., Landin, A.
M. and Blomberg, B. B., Unique biomarkers for B-cell function predict
the serum response to pandemic H1N1 influenza vaccine. Int Immunol
2012. 24: 175–182.
115 Frasca, D., Landin, A. M., Lechner, S. C., Ryan, J. G., Schwartz, R., Riley,
R. L. and Blomberg, B. B., Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and Ig class switch in
human B cells. J Immunol 2008. 180: 5283–5290.
116 Shi, Y., Yamazaki, T., Okubo, Y., Uehara, Y., Sugane, K. and Agematsu,
K., Regulation of aged humoral immune defense against pneumococcal
bacteria by IgM memory B cell. J Immunol 2005. 175: 3262–3267.
117 Fecteau, J. F., Cote, G. and Neron, S., A new memory CD27-IgG+ B cell
population in peripheral blood expressing VH genes with low frequency
of somatic mutation. J. Immunol. 2006. 177: 3728–3736.
118 Colonna-Romano, G., Bulati, M., Aquino, A., Pellicano, M., Vitello, S.,
Lio, D., Candore, G. et al., A double-negative (IgD-CD27-) B cell population is increased in the peripheral blood of elderly people. Mech. Ageing
Dev. 2009. 130: 681–690.
119 Pritz, T., Lair, J., Ban, M., Keller, M., Weinberger, B., Krismer, M. and
Grubeck-Loebenstein, B., Plasma cell numbers decrease in bone marrow
of old patients. Eur. J. Immunol. 2015. 45: 738–746.
120 Frasca, D. and Blomberg, B. B., Aging, cytomegalovirus (CMV) and
influenza vaccine responses. Hum. Vaccin. Immunother. 2015. 12: 682–
690.
121 Frasca, D., Diaz, A., Romero, M., Landin, A. M. and Blomberg, B. B.,
High TNF-alpha levels in resting B cells negatively correlate with their
response. Exp. Gerontol. 2014. 54: 116–122.
122 Frasca, D., Diaz, A., Romero, M., Landin, A. M. and Blomberg, B. B.,
125 Frasca, D., Diaz, A., Romero, M., Landin, A. M., Phillips, M., Lechner,
S. C., Ryan, J. G. et al., Intrinsic defects in B cell response to seasonal influenza vaccination in elderly humans. Vaccine 2010. 28: 8077–
8084.
126 Derhovanessian, E., Theeten, H., Hahnel, K., Van Damme, P., Cools,
N. and Pawelec, G., Cytomegalovirus-associated accumulation of latedifferentiated CD4 T-cells correlates with poor humoral response to
influenza vaccination. Vaccine 2013. 31: 685–690.
127 McElhaney, J. E., Zhou, X., Talbot, H. K., Soethout, E., Bleackley, R. C.,
Granville, D. J. and Pawelec, G., The unmet need in the elderly: how
immunosenescence, CMV infection, co-morbidities and frailty are a
challenge for the development of more effective influenza vaccines.
Vaccine 2012. 30: 2060–2067.
128 Tabibian-Keissar, H., Hazanov, L., Schiby, G., Rosenthal, N., Rakovsky,
A., Michaeli, M., Shahaf, G. L. et al., Aging affects B-cell antigen receptor
repertoire diversity in primary and secondary lymphoid tissues. Eur J
Immunol. 2016. 46:480–492.
129 Gibson, K. L., Wu, Y. C., Barnett, Y., Duggan, O., Vaughan, R., Kondeatis, E., Nilsson, B. O. et al., B-cell diversity decreases in old age and
is correlated with poor health status. Aging Cell 2009. 8: 18–25.
130 Ademokun, A., Wu, Y. C., Martin, V., Mitra, R., Sack, U., Baxendale, H.,
Kipling, D. et al., Vaccination-induced changes in human B-cell repertoire and pneumococcal IgM and IgA antibody at different ages. Aging
Cell 2011. 10: 922–930.
131 Jiang, N., He, J., Weinstein, J. A., Penland, L., Sasaki, S., He, X. S.,
Dekker, C. L. et al., Lineage structure of the human antibody repertoire
in response to influenza vaccination. Sci Transl Med 2013. 5: 171ra119.
132 Fülöp, T., Le Page, A., Fortin, C., Witkowski, J. M., Dupuis, G. and Larbi,
A., Cellular signaling in the aging immune system. Curr. Opin. Immunol.
2014. 29: 105–111.
133 Montgomery, R. R. and Shaw, A. C., Paradoxical changes in innate
immunity in aging: recent progress and new directions. J. Leukoc. Biol.
2015; 98: 937–943.
134 Fortin, C. F., Larbi, A., Lesur, O., Douziech, N. and Fulop, T. Jr., Impairment of SHP-1 down-regulation in the lipid rafts of human neutrophils
under GM-CSF stimulation contributes to their age-related, altered functions. J. Leukoc. Biol. 2006. 79: 1061–1072.
135 Aspinall, R., Lapenna, A., Lynch, C. and Lang, P. O., Cellular signalling
pathways in immune aging and regeneration. Biochem Soc Trans. 2014.
42: 651–656.
136 Goronzy, J. J., Li, G., Yu, M. and Weyand, C. M., Signaling pathways in
aged T cells - A reflection of T cell differentiation, cell senescence and
host environment. Semin. Immunol. 2012. 24: 365–372.
137 Ponnappan, U., Trebilcock, G. U. and Zheng, M. Z., Studies into the effect
of tyrosine phosphatase inhibitor phenylarsine oxide on NFkappaB activation in T lymphocytes during aging: evidence for altered IkappaBalpha phosphorylation and degradation. Exp Gerontol 1999, 34:95–107.
138 LePage, A., Fortin, C., Garneau, H., Allard, N., Tsvetkova, K., Larbi, A.,
Dupuis, G. et al., Modulation of signaling in lymphocytes from elderly
donors. Inhibition of SRC homology 2 domain-containing phosphatase-
Cytomegalovirus (CMV) seropositivity decreases B cell responses to the
1 (SHP-1) leads to recovery of T cell. Cell Commun. Signal. 2014; 12:2 (doi:
influenza vaccine. Vaccine 2015. 33: 1433–1439.
10.1186/1478-811X-12-2).
123 Frasca, D., Diaz, A., Romero, M., Mendez, N. V., Landin, A. M. and
139 Larbi, A., Dupuis, G., Khalil, A., Douziech, N., Fortin, C. and Fulop, T.
Blomberg, B. B., Effects of age on H1N1-specific serum IgG1 and IgG3
Jr., Differential role of lipid rafts in the functions of CD4+ and CD8+
levels evaluated during the 2011–2012 influenza vaccine season. Immun.
Ageing 2013. 10: 14.
124 Khurana, S., Frasca, D., Blomberg, B. and Golding, H., AID activity in B
cells strongly correlates with polyclonal antibody affinity maturation invivo following pandemic 2009-H1N1 vaccination in humans. PLoS Path.
2012. 8: e1002920.
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
human T lymphocytes with aging. Cell Signal. 2006. 18: 1017–1030.
140 Larbi, A., Kempf, J. and Pawelec G., Oxidative stress modulation and T
cell activation. Exp. Gerontol. 2007. 42:852–858.
141 Lenton, K. J., Therriault, H., Cantin, A. M., Fülöp, T., Payette, H. and
Wagner, J. R., Direct correlation of glutathione and ascorbate and their
www.eji-journal.eu
2299
2300
Marcello Pinti et al.
dependence on age and season in human lymphocytes. Am. J. Clin. Nutr.
2000. 71: 1194–2000.
142 Griffiths, H. R., Dunston, C. R., Bennett, S. J., Grant, M. M., Phillips, D.
Eur. J. Immunol. 2016. 46: 2286–2301
159 Zhou, X. and McElhaney, J. E., Age-related changes in memory and
effector T cells responding to influenza A/H3N2 and pandemic A/H1N1
strains in humans. Vaccine 2011. 29: 2169–2177.
C. and Kitas, G. D., Free radicals and redox signalling in T-cells during
160 Holland, D., Booy, R., De Looze, F., Eizenberg, P., McDonald, J., Karrasch,
chronic inflammation and ageing. Biochem. Soc. Trans. 2011. 39:1273–
J., McKeirnan, M. et al., Intradermal influenza vaccine administered
1278.
using a new microinjection system produces superior immunogenicity
143 Larbi, A. and Fülöp, T., From "truly naı̈ve" to "exhausted senescent" T
cells: when markers predict functionality. Cytometry A. 2014. 85:25–35.
144 Di Mitri, D., Azevedo, R. I., Henson, S. M., Libri, V., Riddell, N. E.,
Macaulay, R., Kipling, D. et al., Reversible senescence in human
CD4+CD45RA+CD27- memory T cells. J. Immunol. 2011. 187:2093–2100.
145 Mannick, J. B., Del Giudice, G., Lattanzi, M., Valiante, N. M., Praestgaard, J., Huang, B., Lonetto, M. A. et al., mTOR inhibition improves
immune function in the elderly. Sci. Transl. Med. 2014. 6: 268ra179 (doi:
10.1126/scitranslmed.3009892).
146 Chisolm, D. A. and Weinmann, A. S., TCR-signaling events in cellular
metabolism and specialization. Front. Immunol. 2015. 6: 292.
147 Gavazzi, G. and Krause, K. H., Ageing and infection. Lancet Infect. Dis.
2002. 2: 659–666.
148 van Duin, D., Allore, H. G., Mohanty, S., Ginter, S., Newman, F. K.,
Belshe, R. B., Medzhitov, R. et al., Prevaccine determination of the
in elderly adults: a randomized controlled trial. J. Infect. Dis. 2008. 198:
650–658.
161 Falsey, A. R., Treanor, J. J., Tornieporth, N., Capellan, J. and Gorse, G.
J., Randomized, double-blind controlled phase 3 trial comparing the
immunogenicity of high-dose and standard-dose influenza vaccine in
adults 65 years of age and older. J. Infect. Dis. 2009. 200: 172–180.
162 Ott, G., Barchfeld, G. L. and van Nest, G., Enhancement of humoral
response against human influenza vaccine with the simple submicron oil/water emulsion adjuvant MF59. Vaccine 1995. 13: 1557–
1562.
163 Ansaldi, F., Zancolli, M., Durando, P., Montomoli, E., Sticchi, L.,
Del Giudice, G. and Icardi, G., Antibody response against heterogeneous circulating influenza virus strains elicited by MF59- and nonadjuvanted vaccines during seasons with good or partial matching
between vaccine strain and clinical isolates. Vaccine 2010. 28: 4123–
4129.
expression of costimulatory B7 molecules in activated monocytes pre-
164 Osterholm, M. T., Kelley, N. S., Sommer, A. and Belongia, E. A., Efficacy
dicts influenza vaccine responses in young and older adults. J Infect Dis.
and effectiveness of influenza vaccines: a systematic review and meta-
2007. 195: 1590–1597.
149 Haynes, L. and Eaton, S. M., The effect of age on the cognate function
of CD4+ T cells. Immunol Rev. 2005. 205: 220–228.
150 Goodwin, K., Viboud, C. and Simonsen, L., Antibody response to
influenza vaccination in the elderly: a quantitative review. Vaccine 2006.
24: 1159–1169.
151 Rockwood, K., Song, X., MacKnight, C., Bergman, H., Hogan, D. B.,
McDowell, I. and Mitnitski, A., A global clinical measure of fitness and
frailty in elderly people. CMAJ. 2005. 173: 489–495.
152 Strandberg, T. E. and Pitkälä, K. H., Frailty in elderly people. Lancet. 2007.
369: 1328–1329
153 Leng, S. X., Cappola, A. R., Andersen, R. E., Blackman, M. R., Koenig,
K., Blair, M. and Walston, J. D., Serum levels of insulin-like growth
factor-I (IGF-I) and dehydroepiandrosterone sulfate (DHEA-S), and their
relationships with serum interleukin-6, in the geriatric syndrome of
frailty. Aging Clin Exp Res. 2004. 16: 153–157.
154 Yao, X., Hamilton, R. G., Weng, N. P., Xue, Q. L., Bream, J. H., Li, H.,
Tian, J. et al., Frailty is associated with impairment of vaccine-induced
antibody response and increase in post-vaccination influenza infection
in community-dwelling older adults. Vaccine 2011. 29: 5015–5021.
155 Nakaya, H. I., Hagan, T., Duraisingham, S. S., Lee, E. K., Kwissa,
M., Rouphael, N., Frasca, D. et al., Systems analysis of immunity to
analysis. Lancet Infect. Dis. 2012. 12: 36–44.
165 Beyer, W. E., McElhaney, J., Smith, D. J., Monto, A. S., Nguyen-Van-Tam,
J. S. and Osterhaus, A. D., Cochrane re-arranged: support for policies to
vaccinate elderly people against influenza. Vaccine 2013. 31: 6030–6033.
166 Wiersma, L. C., Rimmelzwaan, G. F. and de Vries, R. D., Developing universal influenza vaccines: hitting the nail, not just on the head. Vaccines
(Basel) 2015. 3: 239–262.
167 Melegaro, A. and Edmunds, W. J., The 23-valent pneumococcal polysaccharide vaccine. Part I. Efficacy of PPV in the elderly: a comparison of
meta-analyses. Eur. J. Epidemiol. 2004. 19: 353–363.
168 Bonten, M. J., Huijts, S. M., Bolkenbaas, M., Webber, C., Patterson, S.,
Gault, S., van Werkhoven C. H. et al., Polysaccharide conjugate vaccine
against pneumococcal pneumonia in adults. N. Engl. J. Med. 2015. 372:
1114–1125.
169 van Werkhoven, C. H., Huijts, S. M., Bolkenbaas, M., Grobbee, D. E.
and Bonten, M. J., The impact of age on the efficacy of 13-valent pneumococcal conjugate vaccine in elderly. Clin. Infect. Dis. 2015. 61: 1835–
1838.
170 Oxman, M. N., Levin, M. J., Johnson, G. R., Schmader, K. E., Straus, S.
E., Gelb, L. D., Arbeit, R. D. et al., A vaccine to prevent herpes zoster
and postherpetic neuralgia in older adults. N. Engl. J. Med. 2005. 352:
2271–2284.
influenza vaccination across multiple years and in diverse popula-
171 Lal, H., Cunningham, A. L., Godeaux, O., Chlibek, R., Diez-Domingo,
tions reveals shared molecular signatures. Immunity 2015. 43: 1186–
J., Hwang, S. J., Levin M. J. et al., Efficacy of an adjuvanted herpes
1198
156 Park, H. L., Shim, S. H., Leem E. Y., Cho, W., Park, S., Jeon, H. J., Ahn
zoster subunit vaccine in older adults. N. Engl. J. Med. 2015. 372: 2087–
2096.
et al., Obesity-induced chronic inflammation is associated with the
172 Chlibek, R., Bayas, J. M., Collins, H., de la Pinta, M. L., Ledent, E., Mols,
reduced efficacy of influenza vaccine. Hum Vaccin Immunother. 2014. 10:
J. F. and Heineman, T. C., Safety and immunogenicity of an AS01-
1181–1186.
adjuvanted varicella-zoster virus subunit candidate vaccine against
157 Frasca, D., Ferracci, F., Diaz, A., Romero, M., Lechner, S. and Blomberg,
B. B., Obesity decreases B cell responses in young and elderly individuals. Obesity (Silver Spring). 2016 . 24: 615–625.
158 Frasca D. and Blomberg, B. B., Inflammaging decreases adaptive and
innate immune responses in mice and humans. Biogerontology 2016. 17:
7–19.
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
herpes zoster in adults >=50 years of age. J Infect Dis. 2013. 208: 1953–
1961.
173 Leroux-Roels, I., Leroux-Roels, G., Clement, F., Vandepapelière, P., Vassilev, V., Ledent, E. and Heineman, T. C., A phase 1/2 clinical trial evaluating safety and immunogenicity of a varicella zoster glycoprotein e
subunit vaccine candidate in young and older adults. J Infect Dis. 2012.
206: 1280–1290
www.eji-journal.eu
HIGHLIGHTS
Eur. J. Immunol. 2016. 46: 2286–2301
174 Didierlaurent, A. M., Collignon, C., Bourguignon, P., Wouters,
S., Fierens, K., Fochesato, M., Dendouga, N. et al., Enhancement of adaptive immunity by the human vaccine adjuvant AS01
depends on activated dendritic cells. J Immunol. 2014. 193: 1920–
1930
175 Weinberger, B., Schirmer, M., Matteucci, G. R., Siebert, U., Fuchs, D. and
Grubeck-Loebenstein, B., Recall responses to tetanus and diphtheria
vaccination are frequently insufficient in elderly persons. PLoS One 2013.
United Kingdom. BMC Infect. Dis. 2015. 15: 443 (doi: 10.1186/s12879-0151218-z.).
179 Shaw, C. A., Ciarlet, M., Cooper, B. W., Dionigi, L., Keith, P., O’Brien, K.
B., Rafie-Kolpin, M. et al., The path to an RSV vaccine. Curr. Opin. Virol.
2013. 3: 332–342.
180 Cross, A. S, Chen, W. H. and Levine, M. M., A case for immunization
against nosocomial infections. J. Leukoc. Biol. 2008. 83: 483–488.
8: e82967.
176 Ridda, I., Yin, J. K., King, C., Raina, M. C. and McIntyre, P., The importance of pertussis in older adults: a growing case for reviewing vaccination strategy in the elderly. Vaccine 2012. 30: 6745–6752.
177 Kaml, M., Weiskirchner, I., Keller, M., Luft, T., Hoster, E., Hasford, J.,
Young, L. et al., Booster vaccination in the elderly: their success depends
Full correspondence: Prof. Andrea Cossarizza, Department of Surgery,
Medicine, Dentistry and Morphological Sciences, University of Modena
and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena,
Italy.
Fax: +39-059-2055-426
e-mail:
[email protected]
on the vaccine type applied earlier in life as well as on pre-vaccination
antibody titers. Vaccine 2006. 24: 6808–6811.
178 Fleming, D. M., Taylor, R. J., Lustig, R. L., Schuck-Paim, C., Haguinet,
F., Webb, D. J., Logie, J. et al., Modelling estimates of the burden of
respiratory syncytial virus infection in adults and the elderly in the
C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Received: 3/3/2016
Revised: 20/8/2016
Accepted: 30/8/2016
Accepted article online: 5/9/2016
www.eji-journal.eu
2301