1,552
Views
50
CrossRef citations to date
0
Altmetric
Review

Th17 cells: interactions with predisposing factors in the immunopathogenesis of inflammatory bowel disease

, , &
Pages 161-168 | Published online: 10 Jan 2014

Abstract

Inflammatory bowel disease (IBD) is a chronic inflammatory state of the GI tract of unknown etiology. Classically, tissue injury in IBD is thought to be primarily mediated by Th1 cells in Crohn’s disease or Th2 cells in ulcerative colitis. The discoveries of new subsets of T-helper cells, especially Th17 cells, have revolutionized our understanding of the disease immunopathology. Th17 cells seem to affect both innate and adaptive immune responses by the release of regulatory cytokines. Understanding the role of Th17 cells in IBD pathogenesis and targeting their regulatory cytokines may provide potential therapeutic approaches for the treatment of IBD in the future.

Medscape: Continuing Medical Education Online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint sponsorship of Medscape, LLC and Expert Reviews Ltd. Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 70% minimum passing score and complete the evaluation at http://www.medscape.org/journal/expertimmunology; (4) view/print certificate.

Release date: 30 January 2012; Expiration date: 30 January 2013

Learning objectives

Upon completion of this activity, participants should be able to:

  • • Describe the role of Th17 cells in regulating immune responses and in the gut mucosal immune barrier

  • • Describe factors involved in the pathogenesis of IBD

  • • Describe the regulatory cytokines of Th17 cells that could potentially offer future strategies to treat IBD

Th17 cells: new players in inflammatory bowel disease immunopathogenesis

Inflammatory bowel disease (IBD) is an inflammatory condition of GI tract that is chronic, remitting and relapsing and also progressive in its course. IBD includes two major clinical entities, ulcerative colitis (UC) and Crohn’s disease (CD). The specific etiology of IBD is unknown. However, the pathogenesis and clinical course of IBD is affected by many factors, broadly categorized as genetic susceptibility of the patient, intestinal flora, various lifestyle issues and the patient’s immune system Citation[1].

The immune system is the principal mediator in IBD pathogenesis and its clinical picture. Genetic mutations causing aberrant immune responses towards environmental factors (most importantly towards gut bacterial flora) are largely believed to be responsible for IBD pathogenesis. Nevertheless, different environmental factors can cause an increase in the incidence of IBD by altering the immune responses in different ways that are not fully understood Citation[2,3].

The immune system can be divided into innate and adaptive immunity. The main cellular components of innate immunity are macrophages, neutrophils, natural killer cells and epithelial cells, while adaptive immunity primarily consists of B lymphocytes (responsible for humoral immunity) and T lymphocytes (responsible for cell-mediated immunity). T lymphocytes are further subdivided into CD4+ (T helper [Th]) cells and CD8+ (cytotoxic T) cells. Complex immunological interactions occur between innate and adaptive immune systems, which may play a critical role in the pathogenesis of IBD, as shown in animal studies Citation[4,5]. This interaction is tightly regulated. Rapid progress in the field of immunology has led to the discovery of many new Th cell subsets. These newly discovered cell lineages play an important role in immune regulation, mainly by cytokine production. One of these Th subsets, Th17 cells, has gained particular attention due to its proinflammatory role in mucosal immune responses and their involvement in autoimmune disorders. In this review, we will discuss different functions of the newly discovered Th17 cell, its relationship with other Th subsets and its potential role in the pathogenesis of IBD.

Th17 discovery

Traditionally, Th cells (CD4+ cells) are divided into two subsets; Th1 and Th2 cells based on initial bioassays, protein expression studies and the cytokines production. Apart from immune response regulation, they were linked to different immune-mediated diseases like autoimmunity and allergy; Th1 responses were seen to be mediating autoimmunity Citation[6] and Th2 cells appeared to be involved in allergy and asthma Citation[7]. To understand the sequence of events leading to Th17 discovery, we will briefly review the development of T-cell subsets. Th cell development begins in the thymus while the functional differentiation of Th cells start when the cell comes in contact with activated antigen-presenting cells (APCs), such as dendritic cells (DCs) in the mucosa. APCs, as well as other immune cells, produce either IL-12 and drive the naive CD4+ cells to become IFN-γ-producing Th1 cells, or IL-4 to induce Th2 cells producing IL-4, IL-5 and IL-13. The Th1 and Th2 axis was very useful for the explanation of autoimmunity and allergy; however, the role of Th1 as the sole driver of autoimmunity was questioned when injections of IFN-γ or IL-12, the driving force for Th1 development, prevented the development of autoimmune disease in animal studies Citation[8,9]. This contradicts the link of Th1 with autoimmunity, hence suggesting the possibility of other pathways involved in its pathogenesis.

Almost a decade ago, a new member of the IL-12 family, IL-23, was described. Soon after, its role in the development of autoimmune diseases was established. In 2003, animal studies showed mice lacking IL-23 were resistant to autoimmune disease; however, mice lacking IL-12 but expressing intact IL-23 suffered from a severe form of autoimmune disease Citation[10]. These surprising results supported the idea that IL-23 may be acting independently of IL-12 on a different population of lymphocytes to mediate autoimmunity. It was also seen that IL-23 stimulates the production of IL-17, a proinflammatory cytokine, from a subpopulation of T cells, designated as Th17 cells, based on their characteristics and specific cytokine production Citation[11].

Multiple studies in humans have described the role of Th17 cells, IL-17 and IL-23 in the pathogenesis of several autoimmune diseases. Their results were comparable to the data in animal models Citation[12–15].

Other recently discovered Th cells include follicular T helper cells, natural regulatory T cells, induced regulatory T cells, Th9 and CD4+CD28null T cells Citation[16,17]. Immune regulation was first presented by Mosmann et al.Citation[18]; however, with new Th cells (especially Th17 cells) playing a central role in immunopathogenesis, our understanding of immune regulation has changed significantly.

Th17 development

Upon contact with APCs, naive CD4+ cells have the potential to differentiate into Th1, Th2, induced regulatory T cell and Th17 cells. APCs control this differentiation through their effector cytokines. These cytokines transmit their signals by binding with the Jak–STAT pathway. When a cytokine binds to its receptor, it activates transcription factors belonging to the STAT family (seven members), which determines the fate of the cells by regulating gene expression Citation[19].

STAT-3 has been shown to regulate various genes involved in the development of Th17 cells in both humans and mice. These transcription factors not only regulate the genes producing Th17 cytokines (IL-17, IL-21) and IL-23 receptors, but also three other transcription factors that are necessary for Th17 differentiation, namely RoRγt, IRF4 and Batf Citation[20].

Retinoid receptors are an important group of nuclear transcription factors. Whereas RoRγ is universally expressed, a transcript denoted RoRγt, is solely expressed in lymphoid cells, and is critical for T-cell development. Two members of retinoic acid orphan receptors (RoRγt and RoRα) are preferentially expressed in Th17 cells and are critical for their differentiation and development; their deletion results in a complete absence of Th17 cells Citation[20]. Similarly, Th1 cell development is promoted by STAT-4 and transcription factor T-bet, Th2 cells require STAT-6 and transcription factor Gata-3, and Treg cells need STAT-5 and transcription factor FoxP3 Citation[19].

Characterization of Th17 cells

Th17 cell development is controlled by different cytokines and in turn, after stimulation, they secrete their own cytokines that control the functions of other cells during the immune response. In the following section, we describe the mechanisms of stimulation of Th17 cells and the effects of different cytokines secreted by these cells on different tissues.

Stimulation of Th17 cells

Several cytokines have been shown to stimulate Th17 cell development and function, notably TGF-β, IL-6, IL-23, IL-21 and IL-1β Citation[21].

The initial phase of Th17 cell development is mainly regulated by IL-6 and TGF-β. IL-6 is produced by many APCs and T cells. It activates the STAT-3 pathway as described above and leads to increased expression of Th17 cytokines, IL-23 receptors and RoRγt. Additionally, TGF-β upregulates the transcription factors FoxP3 and RoRγt, leading to the development of Treg or Th17 cells from CD4+ cells. It is believed that IL-6 signaling is important in blocking FoxP3-mediated suppression of RORγt, which ultimately results in the formation of Th17 cells instead of Treg cells Citation[21]. IL-1β, produced by various cells, is also an important stimulant of Th17 cells in the presence of IL-6 and TGF-β Citation[22]. IL-21, a member of the IL-2 cytokine family produced by Th17 cells, stimulates further development of Th17 cells in the presence of TGF-β. It engages with its receptor and increases the transcription factor IRF4, which promotes cell development. This pathway seems to work independently of the IL-6/TGF-β pathway Citation[23]. In humans IL-1 as well as IL-23, mainly produced by DCs, help in the development and expansion of Th17 cells by engaging with the STAT-3 pathway Citation[22,24].

Cytokines secreted by Th17 cells

Th17 cells are characterized by the secretion of important effector cytokines such as IL-17A (also known as IL-17), IL-17F, IL-22 IL-21, and IFN-γ.

IL-17A and IL-17F are structurally very similar, being proinflammatory in nature; these cytokines play important roles in combating infections and mediating autoimmunity Citation[25,26]. Both cytokines activate the cellular components of innate immunity (epithelial cells, fibroblasts, keratinocytes and endothelial cells) to carry out their functions. Suboptimal production of these cytokines, due to defects in Th17 cells, can lead to increased bacterial infections in humans. Also, blocking IL-17A improved many autoimmune diseases such as rheumatoid arthritis, psoriasis and uveitis, suggesting its role in autoimmunity Citation[25]. IL-22, a member of the IL-10 family, is mainly produced by Th subsets including Th17 and Th1 cells Citation[27]. Unlike other Th17 cytokines, IL-22 does not mediate immune regulation by interacting with other B and T cells; instead it mainly affects the cells in barrier surfaces of the body such as epithelia of the respiratory and gastrointestinal system, and skin. This cytokine appears to enhance the protective ability of the barrier at these sites by enhancing innate immune response Citation[28]. IL-21, a member of the IL-2 family, is mainly produced by Th and NK cells Citation[29], and acts on various other cellular components of innate and adaptive immunity Citation[30–32]. It not only promotes the development of B cells into plasma cells Citation[33] but also expands cytotoxic T cells Citation[34], NK cells and helps in Th17 development Citation[35]. It also has inhibitory effects on some APCs such as DC Citation[36].

Briefly, IL-21 has multiple functions and its target cells include members from both innate and adaptive arms.

Role of Th17 cells in regulation of the immune responses

Two intriguing features of Th17 cells further elaborate their importance in immune regulation, which are their relationship with Treg cells and their plasticity. Treg cells are known for their immunosuppressive function, preventing excessive damage during infection and harmful reactions against self-antigens. Th17 and Treg cells seem to negatively impact each other’s development. Th17 cells can inhibit Treg expansion by producing IL-21; similarly IL-10 produced by Treg cells suppresses Th17 cell development Citation[1]. A fine balance between these immunosuppressive Treg cells and pro-inflammatory Th17 cells is essential for appropriate immune responses that do not cause self-damage and are also able to prevent damage from harmful pathogens.

Recent reports showed that Th17 cells are not one of the most stable cells and under various influences can undergo transformation into other Th subsets Citation[37,38]. This ability to convert and then produce different Th cytokines is called plasticity. Plasticity is an important feature of Th17 cells that seems to help maintain homeostasis and balance in immune responses Citation[16].

The interaction between Th17 and Th1 pathways in IBD is still controversial Citation[39]. In a recent data report from our laboratory [Ali Raza A, Shata MT, Ahmed K et al. The numbers of IFN-γ secreting T cells in peripheral blood stimulated by anti-CD3 or lipopolysaccharide in inflammatory bowel disease patients significantly correlated with disease severity in Crohn’s disease but not in ulcerative colitis (2011), Submitted] as well as other laboratories Citation[40], the increase in IL-17 production was only significantly enhanced in UC, while IFN-γ production was enhanced in CD. This might indicate that IFN-γ may mediate Th1 inflammation in CD and Th17-mediated disease in UC, or that the capacity for producing IL-17 by Th17 may be disturbed by the enhanced Th1 cytokines in the CD intestinal mucosa Citation[40].

Role of Th17 cells in the gut mucosal immune barrier

Th17 cells play an important role in the gut mucosal barrier by affecting the innate and adaptive responses Citation[41]. Mucosal Th17 cells regulate the integrity of the physical barrier by epithelial cells, through chemotaxis of neutrophils and macrophages to prevent against invading pathogens. Its effector cytokines further stimulate the tight junction formation in the mucosal epithelial cells, thus providing resistance to bacterial translocation across gastrointestinal mucosa into the bloodstream Citation[41,42]. Evidence of this important barrier function can be observed in HIV-positive patients who lack Th17 cells and have increased risk of bacteremia from intestinal translocation Citation[41]. Th17 cells also affect the cellular components of the immune system by recruiting neutrophils to the site of inflammation, stimulating antimicrobial peptide production from epithelial cells, increasing matrix metalloproteinase production from fibroblasts, enhancing immunoglobulin production from B cells and regulating different T cells. Thus, Th17 cells contribute to the mucosal barrier by several mechanisms that help the host against invading pathogens.

Pathogenesis of IBD

IBD may affect the entire GI tract as in CD or colonic mucosa as in UC. Various genetic factors, gut flora and lifestyle factors play a dynamic role with the host immune system in the pathogenesis of IBD. Briefly, the development and course of IBD can be divided into four categories:

  • • Immune system

  • • Intestinal flora

  • • Genetic susceptibility

  • • Dietary and lifestyle factors

Immune system

Traditionally, CD has been linked with Th1 cells Citation[43], and UC has been linked with Th2 cells Citation[44], based on the cytokine environment and transcription factors found in their respective mucosa. However, several studies have recently shown that the inflamed gastrointestinal mucosa of patients with IBD has a massive infiltration of Th17 cells Citation[45,46].

Th17 cells are mainly found in the mucosa of the GI tract, especially in the small intestine. This homing of Th17 cells could be affected indirectly by intestinal flora in the large intestine by priming the immune system, and stimulates the development and migration of Th17 to the small intestine Citation[47,48]. Their location together with their plasticity and regulatory effects on other immune cells suggest its importance in mediating GI inflammation. A recent report showed that levels of IL-17 and Th17 were higher in patients with UC and CD compared with normal subjects and patients with ischemic colitis, and these Th17 cells were found abundantly in the mucosa and submucosa of UC and CD, respectively Citation[45].

Recent data from our laboratory suggested a correlation between disease severity and levels of IL-17 secreted by PBMCs from UC patients but not from CD patients, suggesting a differential role of Th17 in these two categories of IBD diseases Citation[49]. Various studies have shown increased production of Th17 cytokines (IL-21 and IL-22), which exacerbates inflammation in IBD by promoting Th1 responses Citation[50,51]. IL-23, also increased in IBD, promotes Th17 and Th1 responses, resulting in inflammation of the GI tract Citation[51,52]. Based on the above observations, it is obvious that Th17 cells play an important role in IBD pathogenesis, which was solely attributed to Th1 and Th2 cells in the past.

Treg cells represent another type of Th cells that have an immunosuppressive function. Treg cells produce important anti-inflammatory cytokines such as IL-10 and TGF-β. Blocking the Treg cytokine, IL-10, led to the development of spontaneous GI tract inflammation in mice Citation[53], possibly due to the inhibition of its suppressive effects on Th17 cells. Therefore, a fine balance needs to be maintained between Th17 and Treg cells in order to avoid excessive inflammation.

Intestinal flora

Humans are colonized by both beneficial and potentially pathogenic microorganisms. The human GI tract harbors more than 1014 microbes belonging to more than 1000 species. Most of these bacteria are found in the colon. Imbalances in the composition of the bacterial microbiota may be one of the factors in human disorders such as IBD. Bacteroidetes (Gram negative) and Firmicutes (Gram positive) make up the majority of the gut flora. This gut flora affects the development, composition and function of innate and adaptive immune responses Citation[1]. In an animal model, it has been shown that Bacteroides fragilis protects from experimental colitis induced by a commensal bacterium with pathogenic potential. There is strong evidence that molecules of the bacterial microbiota can mediate the critical balance between health and disease Citation[54].

The concept of intestinal bacteria as the initiator of immune response comes from observations in animal studies showing that mice in a germ-free state did not develop GI inflammation Citation[55], and the T cells that were reactive against the intestinal flora caused colitis Citation[56]. These microorganisms can also control the responses of Th17 and Treg cells, as shown by various studies Citation[48,54,57,58] directly affecting the immune response Citation[59,60].

Genetic susceptibility

Multiple genes have been linked to IBD pathogenesis. Recent advancements in genetic studies have led to the discovery of more genetic loci for IBD. Thus, approximately 60 genetic loci have been associated with IBD, roughly a third have been associated with UC, another third with CD, and the rest are common with both UC and CD Citation[61]. These genetic studies also elaborate the recent discovery of the IL-23 and Th17 cell pathway. Among many genetic loci, important genes linking Th17 cells to IBD are the IL-23 receptor gene (associated with both UC and CD), IL-10 gene (associated with UC) and IL-17 receptor E-like (IL-17REL) gene (associated with UC) Citation[24]. Other important genetic loci, not directly related to Th17, include NOD2 and TLR4 genes, which are associated with CD and influenced by IL-23 expression Citation[1].

Dietary & lifestyle factors

The incidence of IBD has increased in recent years, in parallel with the changes in the surrounding environment. Major changes in diet, physical activity, obesity and increased smoking habits have been linked to increased incidence of IBD Citation[62,63]. In a recent review, high dietary intakes of specific groups of food, including polyunsaturated fatty acids, ω-6 fatty acids and meat were found to increase the incidence of IBD. Conversely, people with a high fiber and fruit diet were associated with decreased risk of IBD Citation[64]. Obesity seems to adversely affect the clinical course of UC Citation[65], while its effect on CD has variable results Citation[63]. Interestingly, a recent animal study showed that obesity favors the development of Th17 cells, subsequently leading to severe colitis Citation[62].

Expert commentary & five-year view

IBD is a chronic, debilitating illness with significant morbidity. Various environmental and genetic factors affect the initiation and progression of the disease. All these factors affect the immune system in one way or another, causing dysregulated immune response and tissue injury. The immune system being the main mediator of the extent and the type of tissue injury provides the basis for the future therapeutic interventions. Recent advances in the field of immunology have revolutionized our understanding of immune damage in IBD. New cellular components of the immune system have been discovered and described in the past decade. Through animal models, we gathered important information about these newly discovered cellular components of the immune system. Given some minor differences between the immunopathology of humans and animal models, more research aimed towards better characterization of these cellular components in human tissue is required. IBD has a complex pathophysiology and may represent more than one disease with different immunopathological mechanisms. With rapid progress in the immunology and genetic fields, our understanding of the mechanisms underlying the development of IBD has dramatically changed during the last decade. Gut microbiota, genetic associations and immune dysregulation are the focus of interest in IBD pathogenesis. Genome-wide association studies have helped us identify various high-risk groups in the population. Similarly, many ongoing studies are looking at the association between the gut microbiota and IBD pathogenesis. Finally, how to shift the immune balance towards tolerance from autoimmunity will be the cornerstone for IBD therapeutic interventions in the future. Th17 and Tregs, among other newly discovered T cells, hold much promise to understand the intricate balance between self-tolerance and autoimmunity. Th17 cells also hold a key position in IBD immunopathogenesis due to the location of the disease in the gastrointestinal mucosa and plasticity. In the next 5 years, targeted research on the development, characterization and behavior of the mucosal Th17 cells and their relationship with immunosuppressant cells will provide key information about the pathways responsible for IBD pathogenesis, which can serve as potential targets of future therapeutics. Blocking the secretion of Th17 cytokines have already shown promising results in alleviating various autoimmune diseases in humans. The same concept will be tested in the near future for IBD management, which could result in the development of very effective biological agents, which can work in resistant cases of IBD. Other immunomodulatory agents have been tested in IBD patients, including anti-IL-23R antibodies, as well anti-TNF-α antibodies that have shown promising results. It is expected that revolutionized lines of treatment in IBD patients will be developed within the next few years, building on our recent understanding of the immunopathology of IBD patients and the characteristic features of UC and CD.

Key issues

  • • Inflammatory bowel disease (IBD) is a result of increased immune activation, which may lead to mucosal damage.

  • • Genome-wide association studies have revealed various mutations in the immune system that increase the risk of IBD development.

  • • IBD was thought to be a result of impaired functions of T-helper 1 (Th1) and T-helper 2 (Th2) cells, but recent advances have revealed important roles for T-helper 17 (Th17) and Treg cells.

  • • Various environmental factors may increase the activity of Th17 cells and may lead to mucosal damage and IBD development.

  • • Focused translational studies are required to define specific roles of newly discovered T-helper cells in IBD pathogenesis.

Author contributions

A Raza contributed towards writing the manuscript, gathering information and critical review of the literature; W Yousaf contributed towards writing the manuscript and gathering information from the literature; R Giannella contributed by revising the manuscript for important intellectual content and drafting of the manuscript; MT Shata contributed towards writing the manuscript, conception of writing the review and critical revision of the manuscript.

Financial & competing interests disclosure

EDITOR

Elisa Manzotti,Editorial Director, Future Science Group, London, UK

Disclosure: Elisa Manzotti has disclosed no relevant financial relationships.

CME AUTHOR

Laurie Barclay, MD,Freelance writer and reviewer, Medscape, LLC

Disclosure: Laurie Barclay, MD, has disclosed no relevant financial relationships.

AUTHORS

Ali Raza, MD,Department of Internal Medicine, Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH, USA

Disclosure: Janssen Research and Development partially funded the on-going research in the authors’ laboratory, as mentioned in the manuscript.

Wajeeha Yousaf, MD,Department of Internal Medicine, Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH, USA

Disclosure: Janssen Research and Development partially funded the on-going research in the authors’ laboratory, as mentioned in the manuscript.

Ralph Giannella, MD,Department of Internal Medicine, Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH, USA

Disclosure: Janssen Research and Development partially funded the on-going research in the authors’ laboratory, as mentioned in the manuscript.

Mohamed Tarek Shata, MD, PhD,Department of Internal Medicine, Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH, USA

Disclosure: Janssen Research and Development partially funded the on-going research in the authors’ laboratory, as mentioned in the manuscript.

References

  • Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu. Rev. Immunol.28, 573–621 (2010).
  • Tsironi E, Feakins RM, Probert CS, Rampton DS. Incidence of inflammatory bowel disease is rising and abdominal tuberculosis is falling in Bangladeshis in East London, United Kingdom. Am. J. Gastroenterol.99(9), 1749–1755 (2004).
  • Thia KT, Loftus EV, Sandborn WJ, Yang SK. An update on the epidemiology of inflammatory bowel disease in Asia. Am. J. Gastroenterol.103(12), 3167–3182 (2008).
  • Garrett WS, Lord GM, Punit S et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell131(1), 33–45 (2007).
  • Lodes MJ, Cong Y, Elson CO et al. Bacterial flagellin is a dominant antigen in Crohn disease. J. Clin. Invest.113(9), 1296–1306 (2004).
  • Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat. Rev. Immunol.3(7), 521–533 (2003).
  • Reiner SL. Development in motion: helper T cells at work. Cell129(1), 33–36 (2007).
  • Gran B, Chu N, Zhang GX et al. Early administration of IL-12 suppresses EAE through induction of interferon-γ. J. Neuroimmunol.156(1–2), 123–131 (2004).
  • Voorthuis JA, Uitdehaag BM, De Groot CJ, Goede PH, Van Der Meide PH, Dijkstra CD. Suppression of experimental allergic encephalomyelitis by intraventricular administration of interferon-γ in Lewis rats. Clin. Exp. Immunol.81(2), 183–188 (1990).
  • Cua DJ, Sherlock J, Chen Y et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature421(6924), 744–748 (2003).
  • Langrish CL, Chen Y, Blumenschein WM et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med.201(2), 233–240 (2005).
  • Mcgeachy MJ, Cua DJ. Th17 cell differentiation: the long and winding road. Immunity28(4), 445–453 (2008).
  • Lee E, Trepicchio WL, Oestreicher JL et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J. Exp. Med.199(1), 125–130 (2004).
  • Wilson NJ, Boniface K, Chan JR et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat. Immunol.8(9), 950–957 (2007).
  • Burton PR, Clayton DG, Cardon LR et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat. Genet.39(11), 1329–1337 (2007).
  • Bi Y, Liu G, Yang R. Reciprocal modulation between Th17 and other helper T cell lineages. J. Cell. Physiol.226(1), 8–13 (2011).
  • Mai J, Wang H, Yang XF. Th17 cells interplay with FoxP3+ Tregs in regulation of inflammation and autoimmunity. Front Biosci.15, 986–1006 (2010).
  • Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J. Immunol.136(7), 2348–2357 (1986).
  • O’Shea JJ, Murray PJ. Cytokine signaling modules in inflammatory responses. Immunity28(4), 477–487 (2008).
  • Hirahara K, Ghoreschi K, Laurence A, Yang XP, Kanno Y, O’Shea JJ. Signal transduction pathways and transcriptional regulation in Th17 cell differentiation. Cytokine Growth Factor Rev.21(6), 425–434 (2010).
  • Chen Y, Haines CJ, Gutcher I et al. FoxP3(+) regulatory T cells promote T helper 17 cell development in vivo through regulation of interleukin-2. Immunity34(3), 409–421 (2011).
  • Jager A, Kuchroo VK. Effector and regulatory T-cell subsets in autoimmunity and tissue inflammation. Scand. J. Immunol.72(3), 173–184 (2010).
  • Korn T, Bettelli E, Gao W et al. IL-21 initiates an alternative pathway to induce proinflammatory T(h)17 cells. Nature448(7152), 484–487 (2007).
  • Becker C, Wirtz S, Blessing M et al. Constitutive p40 promoter activation and IL-23 production in the terminal ileum mediated by dendritic cells. J. Clin. Invest.112(5), 693–706 (2003).
  • Hot A, Miossec P. Effects of interleukin (IL)-17A and IL-17F in human rheumatoid arthritis synoviocytes. Ann. Rheum. Dis.70(5), 727–732 (2011).
  • Hot A, Zrioual S, Toh ML, Lenief V, Miossec P. IL-17A- versus IL-17F-induced intracellular signal transduction pathways and modulation by IL-17RA and IL-17RC RNA interference in rheumatoid synoviocytes. Ann. Rheum. Dis.70(2), 341–348 (2011).
  • Volpe E, Touzot M, Servant N et al. Multiparametric analysis of cytokine-driven human Th17 differentiation reveals a differential regulation of IL-17 and IL-22 production. Blood114(17), 3610–3614 (2009).
  • Witte E, Witte K, Warszawska K, Sabat R, Wolk K. Interleukin-22: a cytokine produced by T, NK and NKT cell subsets, with importance in the innate immune defense and tissue protection. Cytokine Growth Factor Rev.21(5), 365–379 (2010).
  • Strengell M, Matikainen S, Siren J et al. IL-21 in synergy with IL-15 or IL-18 enhances IFN-γ production in human NK and T cells. J. Immunol.170(11), 5464–5469 (2003).
  • Perez SA, Mahaira LG, Sotiropoulou PA et al. Effect of IL-21 on NK cells derived from different umbilical cord blood populations. Int. Immunol.18(1), 49–58 (2006).
  • Sivori S, Cantoni C, Parolini S et al. IL-21 induces both rapid maturation of human CD34+ cell precursors towards NK cells and acquisition of surface killer Ig-like receptors. Eur. J. Immunol.33(12), 3439–3447 (2003).
  • Strengell M, Julkunen I, Matikainen S. IFN-α regulates IL-21 and IL-21R expression in human NK and T cells. J. Leukocyte Biol.76(2), 416–422 (2004).
  • Ettinger R, Sims GP, Fairhurst AM et al. IL-21 induces differentiation of human naive and memory B cells into antibody-secreting plasma cells. J. Immunol.175(12), 7867–7879 (2005).
  • Tian Y, Yuan C, Ma D et al. IL-21 and IL-12 inhibit differentiation of Treg and Th17 cells and enhance cytotoxicity of peripheral blood mononuclear cells in patients with cervical cancer. Int. J. Gynecological Cancer: Official J. Int. Gynecological Cancer Society21(9), 1672–1678 (2011).
  • Geri G, Terrier B, Rosenzwajg M et al. Critical role of IL-21 in modulating Th17 and regulatory T cells in Behcet disease. J. Allergy Clin. Immunol.128(3), 655–664 (2011).
  • Liu Z, Yang L, Cui Y et al. Il-21 enhances NK cell activation and cytolytic activity and induces Th17 cell differentiation in inflammatory bowel disease. Inflamm. Bowel Dis.15(8), 1133–1144 (2009).
  • Bending D, Newland S, Krejci A, Phillips JM, Bray S, Cooke A. Epigenetic changes at Il12rb2 and Tbx21 in relation to plasticity behavior of Th17 cells. J. Immunol.186(6), 3373–3382 (2011).
  • Lexberg MH, Taubner A, Albrecht I et al. IFN-γ and IL-12 synergize to convert in vivo generated Th17 into Th1/Th17 cells. Eur. J. Immunol.40(11), 3017–3027 (2010).
  • Brand S. Crohn’s disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn’s disease. Gut58(8), 1152–1167 (2009).
  • Kobayashi T, Okamoto S, Hisamatsu T et al. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn’s disease. Gut57(12), 1682–1689 (2008).
  • Blaschitz C, Raffatellu M. Th17 cytokines and the gut mucosal barrier. J. Clin. Immunol.30(2), 196–203 (2010).
  • Kinugasa T, Sakaguchi T, Gu X, Reinecker HC. Claudins regulate the intestinal barrier in response to immune mediators. Gastroenterology118(6), 1001–1011 (2000).
  • Castellanos-Rubio A, Santin I, Irastorza I, Castano L, Carlos Vitoria J, Ramon Bilbao J. Th17 (and Th1) signatures of intestinal biopsies of CD patients in response to gliadin. Autoimmunity42(1), 69–73 (2009).
  • Heller F, Florian P, Bojarski C et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology129(2), 550–564 (2005).
  • Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol.9(5), 313–323 (2009).
  • Mcgovern D, Powrie F. The IL23 axis plays a key role in the pathogenesis of IBD. Gut56(10), 1333–1336 (2007).
  • Sarra M, Pallone F, Macdonald TT, Monteleone G. IL-23/IL-17 axis in IBD. Inflamm. Bowel Dis.16(10), 1808–1813 (2010).
  • Ivanov II, Frutos Rde L, Manel N et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe4(4), 337–349 (2008).
  • Raza A, Ahmed K, Navaneethan U et al. The levels of IL-17 secreted by lipopolysaccharide-stimulated peripheral blood mononuclear cells (PBMC) in inflammatory bowel disease (IBD) patients significantly correlated with disease severity in ulcerative colitis but not in Crohn’s disease. Am. J. Gastroenterol.106(Suppl. 2S), S486 (2011).
  • Monteleone G, Monteleone I, Fina D et al. Interleukin-21 enhances T-helper cell type 1 signaling and interferon-γ production in Crohn’s disease. Gastroenterology128(3), 687–694 (2005).
  • Brand S, Beigel F, Olszak T et al. IL-22 is increased in active Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am. J. Physiol. Gastrointest. Liver Physiol.290(4), G827–G838 (2006).
  • Kamada N, Hisamatsu T, Okamoto S et al. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-γ axis. J. Clin. Invest.118(6), 2269–2280 (2008).
  • Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell75(2), 263–274 (1993).
  • Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature453(7195), 620–625 (2008).
  • Taurog JD, Richardson JA, Croft JT et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J. Exp. Med.180(6), 2359–2364 (1994).
  • Cong Y, Brandwein SL, Mccabe RP et al. CD4+ T cells reactive to enteric bacterial antigens in spontaneously colitic C3H/HeJBir mice: increased T helper cell type 1 response and ability to transfer disease. J. Exp. Med.187(6), 855–864 (1998).
  • Ivanov II, Manel N. Induction of gut mucosal Th17 cells by segmented filamentous bacteria. Med. Sci. (Paris)26(4), 352–355 (2010).
  • Gaboriau-Routhiau V, Rakotobe S, Lecuyer E et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity31(4), 677–689 (2009).
  • Macfarlane GT, Blackett KL, Nakayama T, Steed H, Macfarlane S. The gut microbiota in inflammatory bowel disease. Curr. Pharm. Des.15(13), 1528–1536 (2009).
  • Macfarlane S, Steed H, Macfarlane GT. Intestinal bacteria and inflammatory bowel disease. Crit. Rev. Clin. Lab. Sci.46(1), 25–54 (2009).
  • Thompson AI, Lees CW. Genetics of ulcerative colitis. Inflamm. Bowel Dis.17(3), 831–848 (2011).
  • Winer S, Paltser G, Chan Y et al. Obesity predisposes to Th17 bias. Eur. J. Immunol.39(9), 2629–2635 (2009).
  • Cosnes J. Smoking, physical activity, nutrition and lifestyle: environmental factors and their impact on IBD. Dig. Dis.28(3), 411–417 (2010).
  • Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am. J. Gastroenterol.106(4), 563–573 (2011).
  • Steed H, Walsh S, Reynolds N. A brief report of the epidemiology of obesity in the inflammatory bowel disease population of Tayside, Scotland. Obes. Facts2(6), 370–372 (2009).

Th17 cells: interactions with predisposing factors in the immunopathogenesis of inflammatory bowel disease

To obtain credit, you should first read the journal article. After reading the article, you should be able to answer the following, related, multiple-choice questions. To complete the questions (with a minimum 70% passing score) and earn continuing medical education (CME) credit, please go to http://www.medscape.org/journal/expertimmunology. Credit cannot be obtained for tests completed on paper, although you may use the worksheet below to keep a record of your answers. You must be a registered user on Medscape.org. If you are not registered on Medscape.org, please click on the New Users: Free Registration link on the left hand side of the website to register. Only one answer is correct for each question. Once you successfully answer all post-test questions you will be able to view and/or print your certificate. For questions regarding the content of this activity, contact the accredited provider, [email protected]. For technical assistance, contact [email protected]. American Medical Association’s Physician’s Recognition Award (AMA PRA) credits are accepted in the US as evidence of participation in CME activities. For further information on this award, please refer to http://www.ama-assn.org/ama/pub/category/2922.html. The AMA has determined that physicians not licensed in the US who participate in this CME activity are eligible for AMA PRA Category 1 Credits™. Through agreements that the AMA has made with agencies in some countries, AMA PRA credit may be acceptable as evidence of participation in CME activities. If you are not licensed in the US, please complete the questions online, print the AMA PRA CME credit certificate and present it to your national medical association for review.

Activity Evaluation: Where 1 is strongly disagree and 5 is strongly agree

1. Your patient is a 42-year-old woman with Crohn disease (CD). Based on the review by Dr. Raza and colleagues, which of the following statements about the role of T-helper 17 (Th17) cells in regulating immune responses and in the gut mucosal immune barrier is most likely correct?

  • A Th17 cells stimulate expansion of regulatory T (Treg) cells

  • B Treg cells stimulate expansion of Th17 cells

  • C Plasticity is an important feature of Th17 cells

  • D Th17 cells contribute to the gut mucosal barrier by means of a single mechanism

2. The patient described in question 1 asks you what factors affect the development and course of irritable bowel disease (IBD). Based on the review by Dr. Raza and colleagues, which of the following statements about the pathogenesis of IBD is most likely correct?

  • A The host immune system is the only known contributor to the pathogenesis of IBD

  • B Intestinal flora in the large intestine may prime the immune system and stimulate the development and migration of Th17 cells to the small intestine

  • C Genetic factors have been implicated in ulcerative colitis (UC) but not in CD

  • D High fiber and fruit intake increase the risk for IBD

3. Based on the review by Dr. Raza and colleagues, the regulatory cytokines of Th17 cells could potentially provide future strategies to treat IBD. Which of the following statements about the regulatory cytokines of Th17 cells is most likely correct?

  • A Levels of IL-17 are lower in patients with UC and CD than in both subjects without UC and CD and patients with ischemic colitis

  • B Disease severity is correlated with the levels of IL-17 secreted by peripheral blood mononuclear cells from patients with CD but not from those with UC

  • C Increased production of Th17 cytokines (IL-21 and IL-22) increases inflammation in IBD by promoting Th1 responses

  • D Research to date has not examined the effect on autoimmune diseases of inhibiting secretion of Th17 cytokines

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.