Piis0025619618307511 PDF

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

THEMATIC REVIEW SERIES ON GASTROENTEROLOGICAL DISEASES

Mechanisms of Disease: Inflammatory Bowel


Diseases
Guilherme Piovezani Ramos, MD, and Konstantinos A. Papadakis, MD

CME Activity
Target Audience: The target audience for Mayo Clinic Proceedings is primar- Disclosures: As a provider accredited by Joint Accreditation Interprofessional From the Department of
ily internal medicine physicians and other clinicians who wish to advance Continuing Education, Mayo Clinic College of Medicine and Science (Mayo Internal Medicine (G.P.R.)
their current knowledge of clinical medicine and who wish to stay abreast Clinic School of Continuous Professional Development) must ensure balance,
and Division of Gastroen-
of advances in medical research. independence, objectivity, and scientific rigor in its educational activities.
Statement of Need: General internists and primary care physicians must maintain Course Director(s), Planning Committee Members, Faculty, and all others
terology and Hepatology
an extensive knowledge base on a wide variety of topics covering all body systems who are in a position to control the content of this educational activity are (G.P.R., K.A.P.), Mayo
as well as common and uncommon disorders. Mayo Clinic Proceedings aims to required to disclose all relevant financial relationships with any commercial in- Clinic, Rochester, MN.
leverage the expertise of its authors to help physicians understand best practices terest related to the subject matter of the educational activity. Safeguards
in diagnosis and management of conditions encountered in the clinical setting. against commercial bias have been put in place. Faculty also will disclose any
Accreditation Statement: In support of improving patient care, Mayo off label and/or investigational use of pharmaceuticals or instruments discussed
Clinic College of Medicine and Science is jointly accredited by the Accreditation in their presentation. Disclosure of these relevant financial relationships will be
Council for Continuing Medical Education published in activity materials so those participants in the activity may formulate
(ACCME), the Accreditation Council for Phar- their own judgments regarding the presentation. Disclosure of this information
macy Education (ACPE), and the American will be published in course materials so that those participants in the activity
Nurses Credentialing Center (ANCC) to pro- may formulate their own judgments regarding the presentation.
vide continuing education for the healthcare In their editorial and administrative roles, Karl A. Nath, MBChB, Terry L. Jopke,
team. Kimberly D. Sankey, and Jenna M. Pederson, have control of the content of this
Credit Statements: program but have no relevant financial relationship(s) with industry.
AMA: Mayo Clinic College of Medicine and Science designates this journal- Dr Papadakis is a stock shareholder in Pfizer Inc, GlaxoSmithKline plc, and
based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s)._ Amgen Inc. Dr Ramos reports no competing interests.
Physicians should claim only the credit commensurate with the extent of Method of Participation: In order to claim credit, participants must com-
their participation in the activity. plete the following:
MOC Credit Statement: Successful completion of this CME activity, which in- 1. Read the activity.
cludes participation in the evaluation component, enables the participant to 2. Complete the online CME Test and Evaluation. Participants must achieve
earn up to 1 MOC point in the American Board of Internal Medicine’s a score of 80% on the CME Test. One retake is allowed.
(ABIM) Maintenance of Certification (MOC) program. Participants will earn Visit www.mayoclinicproceedings.org, select CME, and then select CME arti-
MOC points equivalent to the amount of CME credits claimed for the activity. cles to locate this article online to access the online process. On successful
It is the CME activity provider’s responsibility to submit participant completion completion of the online test and evaluation, you can instantly download and
information to ACCME for the purpose of granting ABIM MOC credit. print your certificate of credit.
Learning Objectives: On completion of this article, you should be able to (1) Estimated Time: The estimated time to complete each article is approxi-
define key elements of the intestinal barrier responsible for maintaining intestinal mately 1 hour.
homeostasis, (2) understand the major mechanisms involved in the pathogenesis Hardware/Software: PC or MAC with Internet access.
of inflammatory bowel diseases (IBDs), (3) recognize the importance of genetic Date of Release: 1/1/2019
factors in the contribution to IBD development, (4) identify environmental factors, Expiration Date: 12/31/2020 (Credit can no longer be offered after it has
such as diet and medications, and their association with microbial dysbiosis and passed the expiration date.)
mucosal inflammation, and (5) describe different inflammatory pathways involved Privacy Policy: http://www.mayoclinic.org/global/privacy.html
in patients with IBD and how they can be targeted therapeutically. Questions? Contact [email protected].

Abstract

Inflammatory bowel diseases (IBDs), represented by Crohn disease and ulcerative colitis, are associated with
major morbidity in Western countries and with increasing incidence in the developing world. Although analysis
of the genome of patients with IBD, especially through genome-wide association studies, has unraveled multiple
pathways involved in IBD pathogenesis, only part of IBD heritability has been explained by genetic studies. This
finding has revealed that environmental factors also play a major role in promoting intestinal inflammation,
mostly through their effects in the composition of the microbiome. However, in order for microbial dysbiosis to
result in uncontrolled intestinal inflammation, the intestinal barrier formed by intestinal epithelial cells and the
innate immune system should also be compromised. Finally, activation of the immune system depends on the
working balance between effector and regulatory cells present in the intestinal mucosa, which have also been
found to be dysregulated in this patient population. Therefore, IBD pathogenesis is a result of the interplay of
genetic susceptibility and environmental impact on the microbiome that through a weakened intestinal barrier
will lead to inappropriate intestinal immune activation. In this article, we will review the mechanisms proposed
to cause IBD from the genetic, environmental, intestinal barrier, and immunologic perspectives.
ª 2018 Mayo Foundation for Medical Education and Research n Mayo Clin Proc. 2019;94(1):155-165

Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 155


www.mayoclinicproceedings.org n ª 2018 Mayo Foundation for Medical Education and Research
MAYO CLINIC PROCEEDINGS

endoscopic findings observed in patients

I
nflammatory bowel diseases (IBDs), rep-
resented by Crohn disease (CD) and ul- with IBD (Figures 1 and 2). In this article,
cerative colitis (UC), are chronic and we will review the mechanisms proposed to
remitting disorders responsible for causing cause IBD from the genetic, environmental, in-
inflammation of the gastrointestinal tract. testinal barrier, and immunologic perspectives.
These conditions are associated with major
morbidity and mortality resulting in substan-
tial patient burden and costs to health care GENETIC FACTORS: HOST SUSCEPTIBILITY
systems. Approximately 1.6 million United Family aggregation of IBD has long been
States residents are affected by IBD, 785,000 recognized, but the inheritable component
with CD and 910,000 with UC.1 Prevalence seems to be stronger in CD than in UC, with
is higher in developed Western countries, a concordance rate in monozygotic twins of
with up to 2 million people with these condi- 30% to 58% in CD compared to 10% to 15%
tions in Europe.2 Interestingly, at the turn of in UC.5,7,8 The relative risk for disease devel-
the 21st century, newer epidemiological opment in first-degree relatives of patients
studies have revealed that the incidence of with IBD can be up to 5-fold higher than
IBD has been increasing in developing coun- that in unaffected individuals.9,10 The study
tries in South America, Asia, Africa, and of patients with very early-onset IBD, usually
Eastern Europe.1-3 The change in epidemio- with a family history of IBD and severe mani-
logical patterns in populations in which IBD festations of the disease, have helped identify
was not previously common raises again the rare genetic variants that may interfere with
question of important environmental factors pathways leading to intestinal inflamma-
that are involved in the development of these tion.11,12 For example, a rare mutation
chronic inflammatory disorders. affecting the regulatory function of the X-
The increased prevalence in specific pop- linked inhibitor of apoptosis (XIAP) gene
ulations around the world suggests an was found to result in early-onset IBD refrac-
important genetic component in the devel- tory to treatment in a 15-month old boy.13
opment of IBD, and the new epidemiological XIAP (X-linked inhibitor of apoptosis) is a
trends also reveal that environmental factors positive regulator of nucleotide-binding oligo-
play a critical role in the pathogenesis of CD merization domainecontaining protein 2
and UC. This change may be the result of
industrialization of developing countries.2,3
Most environmental triggers could mediate
GENETICS ENVIRONMENT
IBD pathogenesis through their impact on
the microbiome.3 However, in order for
Susceptibility Smoking
microbiome changes to result in inappro- Inheritance Diet
priate and continuing inflammation, the GWAS Medications
Noncoding variation Microbiota
integrity of the intestinal barrier separating
IBD
the lumen and the mucosa should also be Permeability Innate response
compromised.4,5 Finally, activation of the Goblet cells Cytokines
Paneth cells Effector cells
immune system, which would result in the Autophagy Regulatory cells
phenotypes seen in clinical practice, depends
on the working balance between effector and INTESTINAL IMMUNE
regulatory cells present in the intestinal mu- BARRIER RESPONSE
cosa, which have also been reported to be
dysregulated in this patient population.6 FIGURE 1. Mechanisms involved in the patho-
Therefore, it is the interplay of genetic sus- genesis of inflammatory bowel disease (IBD).
The pathogenesis of IBD is a result of the
ceptibility and environmental impact in the
interplay between genetic, environmental, in-
microbiome that through a weakened intesti-
testinal barrier, and immune response factors.
nal barrier will lead to inappropriate immune GWAS ¼ genome-wide association studies.
activation responsible for the clinical and
n n
156 Mayo Clin Proc. January 2019;94(1):155-165 https://doi.org/10.1016/j.mayocp.2018.09.013
www.mayoclinicproceedings.org
MECHANISMS OF IBD

FIGURE 2. The intestinal mucosa in the normal bowel and in inflammatory bowel disease (IBD). On exposure to environmental
factors, patients with IBD have development of microbial dysbiosis with a decrease of short chain fatty acids (SCFA) producing
bacteria and an increase in Proteobacteria. Mechanisms that maintain the intestinal barrier are also disrupted in the IBD mucosa,
including down-regulation of epithelial cadherin (E-cadherin) in tight junctions; thickness of the mucus layer; abnormal goblet cell
function, including mucin 2 (Muc2) and resistin-like molecule b (RELMb) proteins; and dysfunctional Paneth celleassociated
mechanisms, including secretion of antimicrobial products, nucleotide-binding oligomerization domainecontaining protein 2
(NOD2), and autophagy-related 16-like 1 (ATG16L1) gene-associated functions. From the innate immune system perspective, the IBD
mucosa has been found to exhibit a decrease in colonic macrophages expressing CD14, defective CX3CR1 (C-X3-C motif che-
mokine receptor 1) antigen presentation by dendritic cells; and impaired autophagy. Lastly, although leukocyte migration via integrin
cellular adhesion molecule (CAM) interactions also occurs in the normal mucosa, the balance between effector and regulatory T cells
(T-reg) appears to be disturbed in the IBD mucosa, resulting in uncontrolled activation of different T-cell lineages that migrate to the
inflamed intestine. G ¼ goblet cells; IFNg ¼ interferon g; IL ¼ interleukin; NK-T ¼ natural killer T cell; P ¼ Paneth cells; Th ¼ T helper
cell; TGFb ¼ tumor growth factor b; TNFa ¼ tumor necrosis factor a.

(NOD2) function, which is compromised in childhood-onset IBD represents only 10% to


patients with IBD.14 Furthermore, mutations 25% of all IBD cases, and not all cases have
in the interleukin (IL)10 receptor (IL10RA) been associated with these types of genetic ab-
gene region, which can ultimately manifest normalities, suggesting that IBD is likely a
as early-onset IBD, have also been found to complex polygenic disease.16
be characterized by Medelian-like inheritance Most of the information related to the ge-
with highly penetrant variants.15 Nevertheless, netics of IBD has been obtained through

Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 157


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

genome-wide association studies polymorphisms within the autophagy-


(GWAS).5,8,17 The conceptual basis of related 16-like 1 (ATG16L1) gene, have
GWAS is that complex genetic disorders also revealed autophagy as an important
like IBD are polygenic, being driven by mul- pathogenic process in IBD.24-26 Further-
tiple common genetic polymorphisms.17 more, multiple IBD risk alleles acting at the
Meta-analysis combining GWAS data identi- IL-23 receptor (IL23R) loci suggested a role
fied 201 loci associated with IBD.18,19 of IL-23 signaling in CD, and this pathway
Among those, there are 41 CD-specific and is currently targeted by commercially avail-
30 UC-specific loci that may indicate some able drugs for the treatment of the
of the clinical, endoscopic, and histologic disease.27,28
differences seen in these patients.18,19 How- Up to 80% to 90% of GWAS-identified
ever, approximately 137 of the 201 loci loci, however, are confined to noncoding
(68%) are associated with both CD and variation that exerts its pathogenic effects
UC, indicating that these diseases share com- through modulation of gene expression.17
mon inflammatory pathways.19 Interestingly, Recent studies are focusing on small intranu-
around 70% of IBD loci are shared with clear molecules that can broadly regulate
other complex autoimmune and immunode- gene expression such as epigenetic markers,
ficiency diseases.19 Almost half of IBD- microRNAs, and noncoding RNAs, which
specific loci are associated with other have all been implicated in the pathogenesis
immune-mediated diseases, with strong of IBD through different pathways.
overlaps with ankylosing spondylitis and
psoriasis, which can often present as extrain- ENVIRONMENTAL FACTORS: THE ROLE OF
testinal manifestations of CD and UC.19,20 THE MICROBIOME
Genes implicated in the overlap between Despite the aforementioned advances, only
IBD and primary immunodeficiencies are about 25% of IBD heritability has been
associated with reduced levels of circulating explained by genetic studies.3,17 Further-
T cells or specific T-cell subsets, such as T more, new epidemiological trends of IBD in
helper (Th) 17 cells or regulatory T cells developing countries following industrializa-
(Tregs), again supporting an imbalance be- tion suggest that environmental factors may
tween effector and regulatory cells in the in- play an important role in promoting intesti-
testinal mucosa.6,19,21 nal inflammation in genetically susceptible
The benefit of identifying genes and ge- individuals.2,3
netic loci implicated in IBD is to understand Smoking has been one of the most stud-
disease-relevant biological pathways that are ied factors in the setting of IBD. Clinically,
crucial for the development of intestinal cigarette smoking has been found to be
inflammation.8,17,22 Biological processes harmful in patients with CD but protective
implicated by IBD-specific loci include bar- to those with UC.8 Different pathways have
rier function, epithelial restitution, microbial been hypothesized to explain these findings,
defense, innate immune regulation, reactive including impairments of autophagy, direct
oxygen species generation, autophagy, regu- toxicity to immune- and mucus-producing
lation of adaptive immunity, endoplasmic re- cells, and inducing changes in the micro-
ticulum stress, and metabolic pathways biome.29-31 Moreover, exposure to diets
associated with cellular hemostasis.8,22 For rich in saturated fatty acids and processed
example, NOD2, the first gene to be associ- meats has been reported to increase the
ated with IBD,23 recognizes the peptido- risk of IBD.32-34 Conversely, a high-fiber
glycan product muramyl dipeptide, which diet has been found to reduce the risk of
modulates both innate and adaptive immune CD by 40%.35 The metabolism of dietary fi-
responses.23 Identification of this molecule ber by colonic bacteria into light chain fatty
unraveled the role of the epithelial barrier acids that have anti-inflammatory properties
in maintaining mucosal hemostasis. NOD2 could potentially explain this protective
mutations, as well as others such as effect.36
n n
158 Mayo Clin Proc. January 2019;94(1):155-165 https://doi.org/10.1016/j.mayocp.2018.09.013
www.mayoclinicproceedings.org
MECHANISMS OF IBD

The use of medications, most notably an- Furthermore, the number of mucolytic bacte-
tibiotics, has also been associated with ria seems to be increased in IBD, which
increased risk of IBD.37,38 This association consequently leads to a higher presence of
usually results from changes in the intestinal mucosa-associated bacteria in these pa-
microbiome after use of antibiotics during tients.55,56 Lastly, the number of sulfate-
early stages of life, when the microbiota reducing bacteria, such as Desulfovibrio, is
plays a critical role in shaping immune cell also increased in IBD, resulting in the produc-
39
development. Nonsteroidal anti- tion of hydrogen sulfate that damages the in-
inflammatory drugs, anticontraceptives, and testinal barrier and allows activation of
statins are other examples of medications mucosal inflammation.57
that have been associated with up to 2-
fold-increased risk for CD and UC.40-42 Early THE INTESTINAL BARRIER: EPITHELIUM
life events such as mode of delivery, breast- AND INNATE IMMUNITY
feeding, exposure to pets, and infections The intestinal mucosa exists in a functional
(“hygiene hypothesis”) are also factors asso- equilibrium with the luminal contents, and
ciated with considerable risk for develop- disturbance of this equilibrium can lead to
ment of IBD, mostly given influences in the diseases such as IBD. The intestinal barrier,
composition of the intestinal microbiota.43-46 consisting of intestinal epithelial cells
The intestinal microbiome has long been (IECs) and innate immune cells, maintains
recognized to establish the connection be- this balance between luminal contents and
tween the outside environment and the intes- the mucosa. The importance of the epithelial
tinal mucosa. Microbial dysbiosis, as a result barrier in IBD predisposition is supported by
of decreased diversity of the microbiome, the finding of abnormal intestinal perme-
has been described in patients with IBD.47,48 ability in patients with CD and some of their
Whether this is the cause or consequence of first-degree relatives.58-61 Furthermore, anal-
the observed intestinal inflammation, or ysis of intestinal biopsies from patients with
both, is yet to be determined.47 In patients CD has revealed down-regulation of the
with IBD, there is a decrease of bacteria junctional protein epithelial cadherin, which
with anti-inflammatory capacities and an in- comprises the tight junctions of this physical
crease of bacteria with inflammatory capac- barrier.62,63 Other studies have also associ-
ities compared with healthy individuals.48,49 ated IBD with several transcription factors
The most frequently observed changes involved in epithelial regeneration, such as
include a decrease of Firmicutes and an in- HNF4A (hepatocyte factor 4a) and NKX2-
crease in Proteobacteria and Bacteroidetes.4 3 (NK2 homeobox 3).64,65
The number of short chain fatty acids pro- Beyond just a promoting physical barrier
ducing bacteria (eg, Faecalibacterium praus- between lumen and mucosa, IECs of the in-
nitzii) has been reported to be decreased in testinal barrier consist of different cell types
patients with IBD, consequently affecting dif- that maintain the equilibrium of the lumen-
ferentiation and expansion of Tregs as well as mucosa through different mechanisms; these
growth of epithelial cells.50 Interestingly, the cells include enterocytes, goblet cells, neuro-
lower number of F praunitzii colonization endocrine cells, Paneth cells, and M cells
has been found to correlate with the risk of (Figure 2).66 Goblet cells produce the mucus
ileal CD after surgery and maintenance of matrix covering the epithelium, essential to
clinical remission in UC.51-53 In contrast, both mucosal defense and repair.67 Genetic
the increase in Proteobacteria, most notably deletion of Muc2 (mucin 2), a major goblet
Escherichia coli, with the ability to adhere to cellederived secretory mucin, results in
the intestinal epithelium affects the perme- spontaneous colitis in murine models.68
ability of the intestine, alters the diversity Resistin-like molecule b (RELMb), another
and composition of the microbiota, and in- goblet cellespecific protein, creates a bridge
duces inflammatory responses by regulating with the immune system by directing Th2
the expression of inflammatory genes.54 cell immunity and delivering luminal
Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 159
www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

antigens to tolerogenic sets of dendritic have the capacity of producing anti-


cells.69,70 Disturbance of the RELMb gene re- inflammatory cytokines that promote regula-
duces the severity of colitis in murine tory T-cell differentiation and restrain Th1
models.71 cell and Th17 cell responses.80 Patients
Although Goblet cells seem to play a part with CD have defective innate immune re-
in protecting individuals from colitis devel- sponses, including attenuated macrophage
opment, defects in Paneth cell biology are activity in vitro and impaired neutrophil
associated with increased risk for CD.72,73 recruitment, facilitating microbe passage
Paneth cells reside at the base of small intes- through the mucosa.81 These patients also
tinal crypts and are responsible for crypt ho- present with another inflammatory macro-
meostasis, maintenance of intestinal stem phage population that expresses dendritic
cell niche, and secretion of antimicrobial cell markers, including CD14, and produce
effector cells that control the equilibrium be- large amounts of proinflammatory cytokines,
tween microbiota and mucosa.73 The path- such as tumor necrosis factor a and IL-6.82
ways of several key genetic risk factors for In contrast to phagocytic macrophages, den-
IBD impair Paneth cell function and lead to dritic cells constitute an interface for moni-
colitis, most notably NOD2 and autophagy. toring the environment and relaying signals
NOD2 is expressed by Paneth cells, dendritic to initiate appropriate adaptive responses.83
cells, macrophages, and absorptive IECs. Sampling of bacteria by resident dendritic
NOD2 risk variants are associated with lower cells is mediated in part by a CX3CR1 (C-
levels of a-defensins in Paneth cells leading X3-C motif chemokine receptor 1)edepen-
to impaired antimicrobial function.74 Not dent mechanism that permits direct den-
only through NOD2 but also via CD risk lo- dritic cell to microbe contact.84 Genetic
cus ATG16L1, Paneth cells’ function in auto- deletion of CX3CR1 results in decreased
phagy is compromised in patients with numbers of lamina propria macrophages
CD.25,75 Autophagy is an intracellular “self- and increased translocation of bacteria to
cannibalism” that involves degradation and mesenteric lymph nodes.85 Dendritic cells
recycling of cytosolic contents and organ- have been found to accumulate in the mu-
elles, as well as resistance against infection cosa of patients with IBD. Blockage of
and removal of intracellular microbes.76 Pa- CD40/CD40L interactions between dentritic
tients with ATG16L1 and NOD2 mutations, cell and effector T-cell populations, prevents
as well equivalent murine models, demon- experimental T cellemediated colitis.86,87
strate aberrancies in the secretory apparatus
of Paneth cells resulting in defects in anti- ADAPTIVE IMMUNE RESPONSE: EFFECTOR
bacterial autophagy.77 In dendritic cells, de- AND REGULATORY CELLS
fects in these genes also result in an Upon encounter with antigen and microbial
impaired ability to present exogenous anti- products that gain access through the intes-
gens to T cells.26 tinal barrier, dendritic cells and other
Dendritic cells, macrophages, innate antigen-presenting cells (APCs) initiate a
lymphoid cells, and neutrophils complement cascade of proinflammatory and anti-
the physical and functional barrier of IECs as inflammatory signals to activate different
the first line of defense of a well-developed subsets of local and circulating lymphocytes
mucosal innate immune system. In the to migrate to effector sites where inflamma-
healthy human gut, intestinal macrophages, tion takes place. Migration of leukocytes to
which are characterized by lack of CD14 the inflamed intestine occurs via binding of
expression, exist in a state of hyporespon- integrin molecules located on leukocyte sur-
siveness, showing attenuated proliferation face to cellular adhesion molecules
and chemotactic activity in response to expressed on the surface of endothelial
either microbial ligands or host cytokines cells.88 These endothelial cells, as a reflex
while retaining the phagocytic and bacteri- of inflammatory signals in the mucosa, can
cidal function.78,79 Furthermore, they also also produce chemokines to attract
n n
160 Mayo Clin Proc. January 2019;94(1):155-165 https://doi.org/10.1016/j.mayocp.2018.09.013
www.mayoclinicproceedings.org
MECHANISMS OF IBD

leukocytes to sites where inflammation is production of IL-12, IL-18, IL-23, and trans-
taking place.89 The integrins aLb2, a4b1, forming growth factor b by APCs and macro-
a4b7, and aEb7 are heterodimeric receptors phages. In turn, Th1 and Th17 cells secrete
expressed on the surface of circulating leu- the proinflammatory cytokines IL-17, inter-
kocytes capable of interacting with different feron g, and tumor necrosis factor a that
adhesion molecules for specific leukocyte feed into a self-sustaining amplification cycle
trafficking to the intestine.88 Currently, mul- whereby they stimulate APCs, macrophages,
tiple commercially available drugs are suc- fibroblasts, and endothelial cells to produce
cessfully used in clinical practice to target tumor necrosis factor a, IL-1, IL-6, IL-8,
these molecules in order to prevent leuko- IL-12, and IL-18.5,22,98 Studies have found
cyte migration to the intestine and control that mice deficient in IL-12 p40 subunit
inflammation in patients with IBD.90,91 and IL-23 are resistant to experimentally
New therapies that control inflammation in induced colitis, suggesting that these cyto-
patients with CD and UC target endothelial kines are in the center of the CD inflamma-
cell cellular adhesion molecules or preven- tory pathway.99,100 Therapeutic agents
tion of lymphocyte egression from lymph targeting the shared p40 subunit between
nodes are currently under evaluation in clin- IL-12 and IL-23 are currently used in clinical
ical trials.89,92,93 practice to control intestinal inflammation,
The disturbed balance between anti- and new therapies targeting the IL-
inflammatory and proinflammatory signals 23especific p19 subunit are currently under
with consequent migration of leukocytes to clinical evaluation.101,102 Conversely, drugs
the intestinal mucosa results in and is perpet- directed to the Th17 cellespecific cytokine
uated by an exaggerated T-cell immune IL-17A may worsen the inflammation in pa-
response, which is seen in both CD and UC tients with CD.103 In addition to secretion of
(Figure 2). The T cells involved in immune effector cytokines, Th17 cells also fulfill
responses in these diseases, however, seem important homeostatic functions in the in-
to be different, which may explain phenotypic testine, which may explain these conflicting
differences seen in clinical practice as well as results. In this scenario, IL-17A may also
response to new targeted therapies. Whereas play a role in protecting barrier integrity
the intercalated transmural inflammation of and Treg function, which are crucial to con-
CD results from an excessive Th1 cell and trolling immune activation and limiting in-
Th17 cell response, the uniform mucosal flammatory responses.104
inflammation seen in UC is secondary to a Tregs and Th17 cells have opposing ac-
Th2 celletype-like cytokine profile.22 In UC, tivities but arise from a common precursor
increased secretion of the Th2 cellespecific on transforming growth factor b (TGF-b)
cytokine IL-5 is associated with more efficient stimulation. The abundance of TGF-b in in-
activation of B cells and the induction of im- testinal tissues contributes to normal ho-
mune responses compared with the Th1 cell meostasis by promoting Treg differentiation
response seen in CD.22,94 Interestingly, UC- in naive lamina propria CD4þ T cells.105 In
specific Th cells show only low levels of IL- inflammatory states such as IBD, Th17-cell
4 production, which suggests they do not differentiation is promoted instead as a com-
display all the features of classic Th2 cells.94,95 bination of mucosal TGF-b with other sig-
Instead, UC is associated with the presence of nals, including cytokine, metabolite, and
CD1d-restricted nonclassic natural killer T microbial signals.106 Therefore, the gut may
cells that produce IL-13, which is increased use TGF-b pathways to promote T cells to
in the lamina of patients with UC but not in carry out both proinflammatory and anti-
those with CD.22,95 AntieIL-13 inhibitors inflammatory programs depending on the
have had conflicting results in treating pa- local presence of cytokines and microbial
tients with UC in clinical trials.96,97 products that may consequently affect initia-
In CD, the differentiation of Th cell types tion, persistence, and relapses in human IBD.
1 and 17 occurs in response to the Many of the genes required for Treg and
Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 161
www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

Th17-cell differentiation have been impli- in controlling inflammation in refractory


cated in IBD pathogenesis.8 In experimental UC.113
models, inflammation driven by Th17 cells
leads to colitis and can be suppressed by
Tregs. In this setting, Tregs have an impor- FUTURE PERSPECTIVES: TARGETED
tant function not only in restraining effector EVALUATION AND THERAPY
T-cell populations but also in controlling Understanding the mechanisms that lead to
innate inflammatory mechanisms.107 Loss- IBD pathogenesis is crucial in order to find
of-function mutations in the key Treg tran- new therapeutic strategies to improve patient
scription factor FOXP3 (forkhead box P3) therapies. Beyond identifying new pathways
results in IPEX (immune dysregulation, pol- amendable for therapeutic intervention, new
yendocrinopathy, enteropathy, X-linked syn- genomic studies should also aim to assist
drome), which can be accompanied by with risk stratification and possibly under-
intestinal inflammation.108 Tregs are standing different responses to available ther-
expanded in both inflamed and noninflamed apies and the development of more
UC and could have a critical role in modu- personalized medicine. Aiming for this tar-
lating the clinical spectrum of the disease.109 geted evaluation from an environmental
Therefore, the use of Tregs has been pro- perspective, studies should attempt to charac-
posed as an alternative strategy to control terize the composition of the microbiome,
excessive inflammation in IBD.110 both quantitatively and functionally (metabo-
lome), in order to assess how it interferes
New immunotherapies currently in
with outcomes of intestinal inflammation
development and clinical trials aim to reduce
and response to therapy. Modulation of the
the inflammatory response of IBD by block-
microbiome has already had promising re-
ing downstream signaling pathways associ-
sults in small trials of fecal microbiota trans-
ated with different cytokines. Janus kinases
plant, mostly in patients with UC.114
(JAKs) are a family of tyrosine kinases
Furthermore, the use of cell-based therapies,
(JAK1, JAK2, JAK3, and TYK2 [tyrosine ki-
most notably stem cells, is in the horizon of
nase 2]) that working in pairs mediate intra-
individualized IBD therapy with the purpose
cellular communication between cytokine
of assisting with mucosal healing, recovery
receptors and nuclear signals.111 These mol-
of the intestinal barrier, and control of aber-
ecules act as a bridge between cytokine re-
rant immune responses.115 Lastly, with a va-
ceptors and intranuclear proteins, most
riety of immunotherapies approaching
notably signal transducer and activator of
clinical practice, future studies should
transcription factors (STATs). The 7
consider an individualized treatment
different members of the STAT family
approach based on specific features of each
(STAT1, STAT2, STAT3, STAT4, STAT5A,
individual’s inflammatory response, because
STAT5B, and STAT6) combine in pairs
inflammatory responses vary from patient to
with different JAKs in order to mediate intra-
patient as well as over time in a single patient.
cellular effects of specific cytokine pathways
and are, therefore, potential therapeutic tar-
Abbreviations and Acronyms: APC = antigen-presenting
gets in the modulation of inflammation.112
cell; CD = Crohn disease; GWAS = genome-wide associa-
In the case of IL-12 and IL-23, which are tion studies; IBD = inflammatory bowel disease; IEC = in-
important cytokines in the pathogenesis of testinal epithelial cell; IL = interleukin; JAK = Janus kinase;
CD, JAK2 and TYK are activated by the STAT = signal transducer and activator of transcription; Th
= T helper; Treg = regulatory T cell; UC = ulcerative colitis
membrane receptors and will consequently
recruit STAT3 and STAT4 to deliver intranu- Grant Support: Dr Papadakis is supported by National Insti-
clear signals associated with controlling Th1/ tutes of Health grant 1R03AI131011-01A1.
Th17 cell responses seen in these pa-
Potential Competing Interests: Dr Papadakis is a stock
tients.100,111 Recent studies have also re- shareholder in Pfizer Inc, GlaxoSmithKline plc, and Amgen
ported promising results of JAK inhibitors Inc. Dr Ramos reports no competing interests.

n n
162 Mayo Clin Proc. January 2019;94(1):155-165 https://doi.org/10.1016/j.mayocp.2018.09.013
www.mayoclinicproceedings.org
MECHANISMS OF IBD

The Thematic Review Series on Gastroenterological and highlight shared genetic risk across populations. Nat
Diseases will continue in an upcoming issue. Genet. 2015;47(9):979-986.
19. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interac-
Correspondence: Address to Konstantinos A. Papadakis, tions have shaped the genetic architecture of inflammatory
MD, Division of Gastroenterology and Hepatology, Mayo bowel disease. Nature. 2012;491(7422):119-124.
Clinic, 200 First St SW, Rochester, MN 55905 (Papadakis. 20. Hindorff LA, Sethupathy P, Junkins HA, et al. Potential etio-
logic and functional implications of genome-wide association
[email protected]).
loci for human diseases and traits. Proc Natl Acad Sci U S A.
2009;106(23):9362-9367.
Individual reprints of this article and a bound reprint of the 21. International Union of Immunological Societies Expert Commit-
entire Thematic Review on Gastroenterological Diseases tee on Primary Immunodeficiencies, Notarangelo LD, Fischer A,
will be available for purchase from our website www. Geha RS, et al. Primary immunodeficiencies: 2009 update [pub-
mayoclinicproceedings.org. lished correction appears in J Allergy Clin Immunol. 2010;125(3):
771-773]. J Allergy Clin Immunol. 2009;124(6):1161-1178.
22. Bouma G, Strober W. The immunological and genetic basis
REFERENCES of inflammatory bowel disease. Nat Rev Immunol. 2003;3(7):
1. Aniwan S, Park SH, Loftus EV Jr. Epidemiology, natural history, 521-533.
and risk stratification of Crohn’s disease. Gastroenterol Clin 23. Shaw MH, Kamada N, Warner N, Kim YG, Nuñez G. The
North Am. 2017;46(3):463-480. ever-expanding function of NOD2: autophagy, viral recogni-
2. Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and tion, and T cell activation. Trends Immunol. 2011;32(2):73-79.
prevalence of inflammatory bowel disease in the 21st century: 24. Kuballa P, Huett A, Rioux JD, Daly MJ, Xavier RJ. Impaired auto-
a systematic review of population-based studies. Lancet. 2018; phagy of an intracellular pathogen induced by a Crohn’s disease
390(10114):2769-2778. associated ATG16L1 variant. PLoS One. 2008;3(10):e3391.
3. Shouval DS, Rufo PA. The role of environmental factors in the 25. Travassos LH, Carneiro LA, Ramjeet M, et al. Nod1 and Nod2
pathogenesis of inflammatory bowel diseases: a review. JAMA direct autophagy by recruiting ATG16L1 to the plasma mem-
Pediatr. 2017;171(10):999-1005. brane at the site of bacterial entry. Nat Immunol. 2010;11(1):
4. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. 55-62.
Gut microbiota in the pathogenesis of inflammatory bowel 26. Cooney R, Baker J, Brain O, et al. NOD2 stimulation induces
disease. Clin J Gastroenterol. 2018;11(1):1-10. autophagy in dendritic cells influencing bacterial handling and
5. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of in- antigen presentation. Nat Med. 2010;16(1):90-97.
flammatory bowel disease. Nature. 2007;448(7152):427-434. 27. Duerr RH, Taylor KD, Brant SR, et al. A genome-wide associ-
6. Cader MZ, Kaser A. Recent advances in inflammatory bowel ation study identifies IL23R as an inflammatory bowel disease
disease: mucosal immune cells in intestinal inflammation. Gut. gene. Science. 2006;314(5804):1461-1463.
2013;62(11):1653-1664. 28. Sandborn WJ, Feagan BG, Fedorak RN, et al; Ustekinumab
7. Spehlmann ME, Begun AZ, Burghardt J, Lepage P, Raedler A, Crohn’s Disease Study Group. A randomized trial of Usteki-
Schreiber S. Epidemiology of inflammatory bowel disease in a numab, a human interleukin-12/23 monoclonal antibody, in
German twin cohort: results of a nationwide study. Inflamm patients with moderate-to-severe Crohn’s disease. Gastroen-
Bowel Dis. 2008;14(7):968-976. terology. 2008;135(4):1130-1141.
8. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of in- 29. Allais L, Kerckhof FM, Verschuere S, et al. Chronic cigarette
flammatory bowel disease. Nature. 2011;474(7351):307-317. smoke exposure induces microbial and inflammatory shifts
9. Orholm M, Munkholm P, Langholz E, Nielsen OH, and mucin changes in the murine gut. Environ Microbiol.
Sørensen TI, Binder V. Familial occurrence of inflammatory 2016;18(5):1352-1363.
bowel disease. N Engl J Med. 1991;324(2):84-88. 30. Monick MM, Powers LS, Walters K, et al. Identification of an
10. Tysk C, Lindberg E, Järnerot G, Flodérus-Myrhed B. Ulcerative autophagy defect in smokers’ alveolar macrophages.
colitis and Crohn’s disease in an unselected population of J Immunol. 2010;185(9):5425-5435.
monozygotic and dizygotic twins: a study of heritability and 31. Sher ME, Bank S, Greenberg R, et al. The influence of cigarette
the influence of smoking. Gut. 1988;29(7):990-996. smoking on cytokine levels in patients with inflammatory
11. Ruemmele FM, El Khoury MG, Talbotec C, et al. Characteris- bowel disease. Inflamm Bowel Dis. 1999;5(2):73-78.
tics of inflammatory bowel disease with onset during the first 32. Hou JK, Abraham B, El-Serag H. Dietary intake and risk of
year of life. J Pediatr Gastroenterol Nutr. 2006;43(5):603-609. developing inflammatory bowel disease: a systematic review
12. Bianco AM, Zanin V, Girardelli M, et al. A common genetic of the literature. Am J Gastroenterol. 2011;106(4):563-573.
background could explain early-onset Crohn’s disease. Med 33. Jowett SL, Seal CJ, Pearce MS, et al. Influence of dietary factors
Hypotheses. 2012;78(4):520-522. on the clinical course of ulcerative colitis: a prospective cohort
13. Worthey EA, Mayer AN, Syverson GD, et al. Making a defin- study. Gut. 2004;53(10):1479-1484.
itive diagnosis: successful clinical application of whole exome 34. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. Long-term
sequencing in a child with intractable inflammatory bowel dis- intake of dietary fat and risk of ulcerative colitis and Crohn’s
ease. Genet Med. 2011;13(3):255-262. disease. Gut. 2014;63(5):776-784.
14. Krieg A, Correa RG, Garrison JB, et al. XIAP mediates NOD 35. Ananthakrishnan AN, Khalili H, Konijeti GG, et al.
signaling via interaction with RIP2. Proc Natl Acad Sci U S A. A prospective study of long-term intake of dietary fiber and
2009;106(34):14524-14529. risk of Crohn’s disease and ulcerative colitis. Gastroenterology.
15. Glocker EO, Kotlarz D, Boztug K, et al. Inflammatory bowel 2013;145(5):970-977.
disease and mutations affecting the interleukin-10 receptor. 36. Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of
N Engl J Med. 2009;361(21):2033-2045. inflammation by short chain fatty acids. Nutrients. 2011;3(10):
16. Uhlig HH. Monogenic diseases associated with intestinal 858-876.
inflammation: implications for the understanding of inflamma- 37. Shaw SY, Blanchard JF, Bernstein CN. Association between
tory bowel disease. Gut. 2013;62(12):1795-1805. the use of antibiotics in the first year of life and pediatric in-
17. McGovern DP, Kugathasan S, Cho JH. Genetics of inflammatory flammatory bowel disease. Am J Gastroenterol. 2010;105(12):
bowel diseases. Gastroenterology. 2015;149(5):1163-1176.e2. 2687-2692.
18. Liu JZ, van Sommeren S, Huang H, et al. Association analyses 38. Selby W, Pavli P, Crotty B, et al; Antibiotics in Crohn’s Disease
identify 38 susceptibility loci for inflammatory bowel disease Study Group. Two-year combination antibiotic therapy with

Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 163


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

clarithromycin, rifabutin, and clofazimine for Crohn’s disease. utilization of mucin by other bacteria. Am J Gastroenterol.
Gastroenterology. 2007;132(7):2313-2319. 2010;105(11):2420-2428.
39. Olszak T, An D, Zeissig S, et al. Microbial exposure during 58. Soderholm JD, Olaison G, Peterson KH, et al. Augmented in-
early life has persistent effects on natural killer T cell function. crease in tight junction permeability by luminal stimuli in the
Science. 2012;336(6080):489-493. non-inflamed ileum of Crohn’s disease. Gut. 2002;50(3):307-313.
40. Khalili H, Higuchi LM, Ananthakrishnan AN, et al. Oral contra- 59. Buhner S, Buning C, Genschel J, et al. Genetic basis for increased
ceptives, reproductive factors and risk of inflammatory bowel intestinal permeability in families with Crohn’s disease: role of
disease. Gut. 2013;62(8):1153-1159. CARD15 3020insC mutation? Gut. 2006;55(3):342-347.
41. Ananthakrishnan AN, Higuchi LM, Huang ES, et al. Aspirin, 60. Irvine EJ, Marshall JK. Increased intestinal permeability precedes
nonsteroidal anti-inflammatory drug use, and risk for Crohn the onset of Crohn’s disease in a subject with familial risk.
disease and ulcerative colitis: a cohort study. Ann Intern Med. Gastroenterology. 2000;119(6):1740-1744.
2012;156(5):350-359. 61. May GR, Sutherland LR, Meddings JB. Is small intestinal perme-
42. Kvasnovsky CL, Aujla U, Bjarnason I. Nonsteroidal anti- ability really increased in relatives of patients with Crohn’s dis-
inflammatory drugs and exacerbations of inflammatory bowel ease? Gastroenterology. 1993;104(6):1627-1632.
disease. Scand J Gastroenterol. 2015;50(3):255-263. 62. Muise AM, Walters TD, Glowacka WK, et al. Polymorphisms
43. Bager P, Simonsen J, Nielsen NM, Frisch M. Cesarean section in E-cadherin (CDH1) result in a mis-localised cytoplasmic
and offspring’s risk of inflammatory bowel disease: a national protein that is associated with Crohn’s disease. Gut. 2009;
cohort study. Inflamm Bowel Dis. 2012;18(5):857-862. 58(8):1121-1127.
44. Ng SC, Tang W, Leong RW, et al; Asia-Pacific Crohn’s and 63. Gassler N, Rohr C, Schneider A, et al. Inflammatory bowel
Colitis Epidemiology Study ACCESS Group. Environmental disease is associated with changes of enterocytic junctions.
risk factors in inflammatory bowel disease: a population- Am J Physiol Gastrointest Liver Physiol. 2001;281(1):G216-G228.
based case-control study in Asia-Pacific. Gut. 2015;64(7): 64. Darsigny M, Babeu JP, Dupuis AA, et al. Loss of hepatocyte-
1063-1071. nuclear-factor-4a affects colonic ion transport and causes
45. Chu KM, Watermeyer G, Shelly L, et al. Childhood helminth chronic inflammation resembling inflammatory bowel disease
exposure is protective against inflammatory bowel disease: a in mice. PLoS One. 2009;4(10):e7609.
case control study in South Africa. Inflamm Bowel Dis. 2013; 65. Pabst O, Zweigerdt R, Arnold HH. Targeted disruption of the
19(3):614-620. homeobox transcription factor Nkx2-3 in mice results in post-
46. Cholapranee A, Ananthakrishnan AN. Environmental hygiene natal lethality and abnormal development of small intestine
and risk of inflammatory bowel diseases: a systematic review and spleen. Development. 1999;126(10):2215-2225.
and meta-analysis. Inflamm Bowel Dis. 2016;22(9):2191-2199. 66. van der Flier LG, Clevers H. Stem cells, self-renewal, and dif-
47. Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and ferentiation in the intestinal epithelium. Annu Rev Physiol. 2009;
fungi in pathogenesis of inflammatory bowel diseases and 71:241-260.
therapeutic approaches. Gastroenterology. 2017;152(2):327- 67. Johansson ME, Ambort D, Pelaseyed T, et al. Composition
339.e4. and functional role of the mucus layers in the intestine. Cell
48. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Mol Life Sci. 2011;68(22):3635-3641.
Harpaz N, Pace NR. Molecular-phylogenetic characterization 68. Van der Sluis M, De Koning BA, De Bruijn AC, et al. Muc2-
of microbial community imbalances in human inflammatory deficient mice spontaneously develop colitis, indicating that
bowel diseases. Proc Natl Acad Sci U S A. 2007;104(34): Muc2 is critical for colonic protection. Gastroenterology.
13780-13785. 2006;131(1):117-129.
49. Peterson DA, Frank DN, Pace NR, Gordon JI. Metagenomic 69. Herbert DR, Yang JQ, Hogan SP, et al. Intestinal epithelial cell
approaches for defining the pathogenesis of inflammatory secretion of RELM-b protects against gastrointestinal worm
bowel diseases. Cell Host Microbe. 2008;3(6):417-427. infection. J Exp Med. 2009;206(13):2947-2957.
50. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a 70. McDole JR, Wheeler LW, McDonald KG, et al. Goblet cells
rationally selected mixture of Clostridia strains from the hu- deliver luminal antigen to CD103þ dendritic cells in the small
man microbiota. Nature. 2013;500(7461):232-236. intestine. Nature. 2012;483(7389):345-349.
51. Varela E, Manichanh C, Gallart M, et al. Colonisation by Fae- 71. McVay LD, Keilbaugh SA, Wong TM, et al. Absence of bacte-
calibacterium prausnitzii and maintenance of clinical remission rially induced RELMb reduces injury in the dextran sodium
in patients with ulcerative colitis. Aliment Pharmacol Ther. sulfate model of colitis. J Clin Invest. 2006;116(11):2914-2923.
2013;38(2):151-161. 72. Itoh H, Beck PL, Inoue N, Xavier R, Podolsky DK.
52. Fujimoto T, Imaeda H, Takahashi K, et al. Decreased abun- A paradoxical reduction in susceptibility to colonic injury
dance of Faecalibacterium prausnitzii in the gut microbiota of upon targeted transgenic ablation of goblet cells. J Clin Invest.
Crohn’s disease. J Gastroenterol Hepatol. 2013;28(4):613-619. 1999;104(11):1539-1547.
53. Takahashi K, Nishida A, Fujimoto T, et al. Reduced abundance 73. Salzman NH, Underwood MA, Bevins CL. Paneth cells, defen-
of butyrate-producing bacteria species in the fecal microbial sins, and the commensal microbiota: a hypothesis on intimate
community in Crohn’s disease [published correction appears interplay at the intestinal mucosa. Semin Immunol. 2007;19(2):
in Digestion. 2016;93(2):174]. Digestion. 2016;93(1):59-65. 70-83.
54. Ahmed I, Roy BC, Khan SA, Septer S, Umar S. Microbiome, 74. Fritz T, Niederreiter L, Adolph T, Blumberg RS, Kaser A.
metabolome and inflammatory bowel disease. Microorganisms. Crohn’s disease: NOD2, autophagy and ER stress converge.
2016;4(2):20. Gut. 2011;60(11):1580-1588.
55. van der Waaij LA, Harmsen HJ, Madjipour M, et al. Bacterial 75. Hampe J, Franke A, Rosenstiel P, et al. A genome-wide asso-
population analysis of human colon and terminal ileum bi- ciation scan of nonsynonymous SNPs identifies a susceptibility
opsies with 16S rRNA-based fluorescent probes: commensal variant for Crohn disease in ATG16L1. Nat Genet. 2007;39(2):
bacteria live in suspension and have no direct contact with 207-211.
epithelial cells. Inflamm Bowel Dis. 2005;11(10):865-871. 76. Levine B, Kroemer G. Autophagy in the pathogenesis of dis-
56. Schultsz C, Van Den Berg FM, Ten Kate FW, Tytgat GN, ease. Cell. 2008;132(1):27-42.
Dankert J. The intestinal mucus layer from patients with inflamma- 77. Cadwell K, Liu JY, Brown SL, et al. A key role for autophagy
tory bowel disease harbors high numbers of bacteria compared and the autophagy gene Atg16l1 in mouse and human intes-
with controls. Gastroenterology. 1999;117(5):1089-1097. tinal Paneth cells. Nature. 2008;456(7219):259-263.
57. Png CW, Lindén SK, Gilshenan KS, et al. Mucolytic bacteria 78. Smythies LE, Sellers M, Clements RH, et al. Human intestinal
with increased prevalence in IBD mucosa augment in vitro macrophages display profound inflammatory anergy despite

n n
164 Mayo Clin Proc. January 2019;94(1):155-165 https://doi.org/10.1016/j.mayocp.2018.09.013
www.mayoclinicproceedings.org
MECHANISMS OF IBD

avid phagocytic and bacteriocidal activity. J Clin Invest. 2005; 98. Nanau RM, Neuman MG. Metabolome and inflamma-
115(1):66-75. some in inflammatory bowel disease. Transl Res. 2012;
79. Smith PD, Ochsenbauer-Jambor C, Smythies LE. Intestinal 160(1):1-28.
macrophages: unique effector cells of the innate immune sys- 99. Oppmann B, Lesley R, Blom B, et al. Novel p19 protein en-
tem. Immunol Rev. 2005;206:149-159. gages IL-12p40 to form a cytokine, IL-23, with biological activ-
80. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. ities similar as well as distinct from IL-12. Immunity. 2000;13(5):
Lamina propria macrophages and dendritic cells differentially 715-725.
induce regulatory and interleukin 17-producing T cell re- 100. Teng MW, Bowman EP, McElwee JJ, et al. IL-12 and IL-23 cy-
sponses. Nat Immunol. 2007;8(10):1086-1094. tokines: from discovery to targeted therapies for immune-
81. Smith AM, Rahman FZ, Hayee B, et al. Disordered macro- mediated inflammatory diseases. Nat Med. 2015;21(7):719-
phage cytokine secretion underlies impaired acute inflamma- 729.
tion and bacterial clearance in Crohn’s disease [published 101. Feagan BG, Sandborn WJ, Gasink C, et al; UNITIeIM-UNITI
correction appears in J Exp Med. 2009;206(10):2301]. J Exp Study Group. Ustekinumab as induction and maintenance
Med. 2009;206(9):1883-1897. therapy for Crohn’s disease. N Engl J Med. 2016;375(20):
82. Kamada N, Hisamatsu T, Okamoto S, et al. Unique CD14þ 1946-1960.
intestinal macrophages contribute to the pathogenesis of 102. Panaccione R, Sandborn WJ, Gordon GL, et al. Briakinumab
Crohn disease via IL-23/IFN-g axis. J Clin Invest. 2008;118(6): for treatment of Crohn’s disease: results of a randomized trial.
2269-2280. Inflamm Bowel Dis. 2015;21(6):1329-1340.
83. Rescigno M. Intestinal dendritic cells. Adv Immunol. 2010;107: 103. Hueber W, Sands BE, Lewitzky S, et al; Secukinumab in
109-138. Crohn’s Disease Study Group. Secukinumab, a human anti-
84. Niess JH, Brand S, Gu X, et al. CX3CR1-mediated dendritic IL-17A monoclonal antibody, for moderate to severe Crohn’s
cell access to the intestinal lumen and bacterial clearance. Sci- disease: unexpected results of a randomised, double-blind pla-
ence. 2005;307(5707):254-258. cebo-controlled trial. Gut. 2012;61(12):1693-1700.
85. Medina-Contreras O, Geem D, Laur O, et al. CX3CR1 regu- 104. Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17
lates intestinal macrophage homeostasis, bacterial transloca- pathway and inflammatory diseases of the intestines, lungs,
tion, and colitogenic Th17 responses in mice. J Clin Invest. and skin. Annu Rev Pathol. 2013;8:477-512.
2011;121(12):4787-4795. 105. Zhou L, Lopes JE, Chong MM, et al. TGF-b-induced Foxp3 in-
86. Hart AL, Al-Hassi HO, Rigby RJ, et al. Characteristics of intes- hibits TH17 cell differentiation by antagonizing RORgt func-
tinal dendritic cells in inflammatory bowel diseases. Gastroen- tion. Nature. 2008;453(7192):236-240.
terology. 2005;129(1):50-65. 106. Ghoreschi K, Laurence A, Yang XP, et al. Generation of path-
87. Uhlig HH, McKenzie BS, Hue S, et al. Differential activity of IL- ogenic TH17 cells in the absence of TGF-b signalling. Nature.
12 and IL-23 in mucosal and systemic innate immune pathol- 2010;467(7318):967-971.
ogy. Immunity. 2006;25(2):309-318. 107. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown
88. Arseneau KO, Cominelli F. Targeting leukocyte trafficking for in inflammatory bowel disease. Nature. 2011;474(7351):298-
the treatment of inflammatory bowel disease. Clin Pharmacol 306.
Ther. 2015;97(1):22-28. 108. Wildin RS, Ramsdell F, Peake J, et al. X-linked neonatal dia-
89. Danese S, Panés J. Development of drugs to target interac- betes mellitus, enteropathy and endocrinopathy syndrome is
tions between leukocytes and endothelial cells and treatment the human equivalent of mouse scurfy. Nat Genet. 2001;
algorithms for inflammatory bowel diseases. Gastroenterology. 27(1):18-20.
2014;147(5):981-989. 109. Yu QT, Saruta M, Avanesyan A, Fleshner PR, Banham AH,
90. Jovani M, Danese S. Vedolizumab for the treatment of IBD: a Papadakis KA. Expression and functional characterization of
selective therapeutic approach targeting pathogenic a4b7 cells. FOXP3þ CD4þ regulatory T cells in ulcerative colitis.
Curr Drug Targets. 2013;14(12):1433-1443. Inflamm Bowel Dis. 2007;13(2):191-199.
91. Vermeire S, O’Byrne S, Keir M, et al. Etrolizumab as induction 110. Himmel ME, Yao Y, Orban PC, Steiner TS, Levings MK.
therapy for ulcerative colitis: a randomised, controlled, phase 2 Regulatory T-cell therapy for inflammatory bowel disease:
trial. Lancet. 2014;384(9940):309-318. more questions than answers. Immunology. 2012;136(2):
92. Vermeire S, Ghosh S, Panes J, et al. The mucosal addressin cell 115-122.
adhesion molecule antibody PF-00547,659 in ulcerative colitis: 111. Galien R. Janus kinases in inflammatory bowel disease: four ki-
a randomised study. Gut. 2011;60(8):1068-1075. nases for multiple purposes. Pharmacol Rep. 2016;68(4):789-
93. Sandborn WJ, Feagan BG, Wolf DC, et al; TOUCHSTONE 796.
Study Group. Ozanimod induction and maintenance treatment 112. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB,
for ulcerative colitis. N Engl J Med. 2016;374(18):1754-1762. Laurence A. The JAK-STAT pathway: impact on human dis-
94. Fuss IJ, Neurath M, Boirivant M, et al. Disparate CD4þ lamina ease and therapeutic intervention. Annu Rev Med. 2015;66:
propria (LP) lymphokine secretion profiles in inflammatory bowel 311-328.
disease: Crohn’s disease LP cells manifest increased secretion of 113. Sandborn WJ, Su C, Sands BE, et al; OCTAVE Induction 1,
IFN-gamma, whereas ulcerative colitis LP cells manifest increased OCTAVE Induction 2, and OCTAVE Sustain Investigators.
secretion of IL-5. J Immunol. 1996;157(3):1261-1270. Tofacitinib as induction and maintenance therapy for ulcera-
95. Neurath MF. Cytokines in inflammatory bowel disease. Nat tive colitis. N Engl J Med. 2017;376(18):1723-1736.
Rev Immunol. 2014;14(5):329-342. 114. Paramsothy S, Paramsothy R, Rubin DT, et al. Faecal micro-
96. Danese S, Rudzinski J, Brandt W, et al. Tralokinumab for biota transplantation for inflammatory bowel disease: a sys-
moderate-to-severe UC: a randomised, double-blind, pla- tematic review and meta-analysis. J Crohns Colitis. 2017;
cebo-controlled, phase IIa study. Gut. 2015;64(2):243-249. 11(10):1180-1199.
97. Reinisch W, Panés J, Khurana S, et al. Anrukinzumab, an anti- 115. Dave M, Mehta K, Luther J, Baruah A, Dietz AB,
interleukin 13 monoclonal antibody, in active UC: efficacy and Faubion WA Jr. Mesenchymal stem cell therapy for inflamma-
safety from a phase IIa randomised multicentre study. Gut. tory bowel disease: a systematic review and meta-analysis.
2015;64(6):894-900. Inflamm Bowel Dis. 2015;21(11):2696-2707.

Mayo Clin Proc. n January 2019;94(1):155-165 n https://doi.org/10.1016/j.mayocp.2018.09.013 165


www.mayoclinicproceedings.org

You might also like