501
|
Skin-draining lymph nodes contain dermis-derived CD103(-) dendritic cells that constitutively produce retinoic acid and induce Foxp3(+) regulatory T cells. Blood 2010; 115:1958-68. [PMID: 20068222 DOI: 10.1182/blood-2009-09-245274] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Small intestinal CD103(+) dendritic cells (DCs) have the selective ability to promote de novo generation of regulatory T cells via the production of retinoic acid (RA). Considering that aldehyde dehydrogenase (ALDH) activity controls the production of RA, we used a flow cytometry-based assay to measure ALDH activity at the single-cell level and to perform a comprehensive analysis of the RA-producing DC populations present in lymphoid and nonlymphoid mouse tissues. RA-producing DCs were primarily of the tissue-derived, migratory DC subtype and can be readily found in the skin and in the lungs as well as in their corresponding draining lymph nodes. The RA-producing skin-derived DCs were capable of triggering the generation of regulatory T cells, a finding demonstrating that the presence of RA-producing, tolerogenic DCs is not restricted to the intestinal tract as previously thought. Unexpectedly, the production of RA by skin DCs was restricted to CD103(-) DCs, indicating that CD103 expression does not constitute a "universal" marker for RA-producing mouse DCs. Finally, Toll-like receptor (TLR) triggering or the presence of a commensal microflora was not essential for the induction of ALDH activity in the discrete ALDH(+) DC subsets that characterize tissues constituting environmental interfaces.
Collapse
|
502
|
Nakao A. The role and potential use of oral transforming growth factor-beta in the prevention of infant allergy. Clin Exp Allergy 2010; 40:725-30. [PMID: 20067481 DOI: 10.1111/j.1365-2222.2009.03445.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The increasing prevalence of allergic diseases in infants and children as well as adults has become an important issue in public health in industrial countries. However, few preventive measures are available to reduce the risk of allergic diseases in infants; e.g. the avoidance of smoking and alcohol consumption during pregnancy and lactation. Therefore, there is an enthusiasm to identify certain factors in foods, nutrients, and environment responsible for the primary prevention of allergic diseases during infancy. In the last decade, TGF-beta in maternal milk has been implicated in the prevention of allergic diseases in infants and young children. This review summarizes the relevant epidemiological reports and highlights the recent animal studies to support the preventive role of orally administered TGF-beta, such as TGF-beta in human milk, in the development of allergic diseases in infants. We also provide suggestions for the potential use of dietary (oral) TGF-beta for the primary prevention of allergic diseases. Further studies to address the scientific validity and mechanistic insight to this Mother Nature-inspired concept are clearly required and will be important to develop new approaches to prevent allergic diseases.
Collapse
Affiliation(s)
- A Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Yamanashi, Japan.
| |
Collapse
|
503
|
Abstract
Since their discovery by Steinman and Cohn in 1973, dendritic cells (DCs) have become increasingly recognized for their crucial role as regulators of innate and adaptive immunity. DCs are exquisitely adept at acquiring, processing, and presenting antigens to T cells. They also adjust the context (and hence the outcome) of antigen presentation in response to a plethora of environmental inputs that signal the occurrence of pathogens or tissue damage. Such signals generally boost DC maturation, which promotes their migration from peripheral tissues into and within secondary lymphoid organs and their capacity to induce and regulate effector T cell responses. Conversely, more recent observations indicate that DCs are also crucial to ensure immunological peace. Indeed, DCs constantly present innocuous self- and nonself-antigens in a fashion that promotes tolerance, at least in part, through the control of regulatory T cells (Tregs). Tregs are specialized T cells that exert their immunosuppressive function through a variety of mechanisms affecting both DCs and effector cells. Here, we review recent advances in our understanding of the relationship between tolerogenic DCs and Tregs.
Collapse
|
504
|
Miloud T, Hämmerling GJ, Garbi N. Review of murine dendritic cells: types, location, and development. Methods Mol Biol 2010; 595:21-42. [PMID: 19941103 DOI: 10.1007/978-1-60761-421-0_2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Dendritic cells (DCs) are key coordinators of the immune response, governing the choice between tolerance and immunity. DCs are professional antigen-presenting cells capable of presenting antigen on MHC molecules and priming CD4 and CD8 T-cell responses. They form a heterogeneous group of cells based on phenotype, location, and function. In this review, murine DCs will be discussed regarding their function with special emphasis on their tissue distribution. Recent findings on DC homeostasis during cancer progression will be presented. Finally, the developmental pathways leading to DC differentiation from their precursors will be summarized.
Collapse
Affiliation(s)
- Tewfik Miloud
- Division of Molecular Immunology, German Cancer Research Center DKFZ, Heidelberg, Germany
| | | | | |
Collapse
|
505
|
Proinflammatory Th2 cytokines induce production of thymic stromal lymphopoietin in human colonic epithelial cells. Dig Dis Sci 2010; 55:1896-904. [PMID: 19757042 PMCID: PMC7087909 DOI: 10.1007/s10620-009-0979-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Accepted: 08/31/2009] [Indexed: 12/12/2022]
Abstract
PURPOSE Thymic stromal lymphopoietin (TSLP) is released by intestinal epithelial cells (IECs), and TSLP-conditioned dendritic cells appear to be involved in immune homeostasis of intestine and immunoglobulin A (IgA) class-switching in the physiological condition. In contrast, TSLP activates dendritic cells to induce strong T-cell proliferation and is involved in inflammatory T helper (Th) 2 responses in human allergic diseases. However, it is not clear how TSLP production by IECs is regulated in ulcerative colitis (UC), which appears to involve inflammatory Th2 responses. The aim of this study is to examine how TSLP production by IECs is regulated in ulcerative colitis. RESULTS We show here that expression of TSLP was enhanced in mucosal lesions from UC patients in which inflammatory Th2 cytokine production was predominant. In addition, using a human colonic epithelial cell line, we demonstrated that a combination of tumor necrosis factor-alpha (TNF-alpha) and interleukin-4 (IL-4) induced TSLP expression and that TSLP expression by TNF-alpha + IL-4 was further enhanced by either Toll-like receptor 3 ligand or interferon (IFN)-gamma. CONCLUSIONS Taken together, as in human allergic diseases, an inflammatory Th2 condition in the mucosal lesions of UC patients may trigger increased TSLP expression by IECs, resulting in exacerbation of UC.
Collapse
|
506
|
Abstract
The human intestine is colonized by an estimated 100 trillion bacteria. Some of these bacteria are essential for normal physiology, whereas others have been implicated in the pathogenesis of multiple inflammatory diseases including IBD and asthma. This review examines the influence of signals from intestinal bacteria on the homeostasis of the mammalian immune system in the context of health and disease. We review the bacterial composition of the mammalian intestine, known bacterial-derived immunoregulatory molecules, and the mammalian innate immune receptors that recognize them. We discuss the influence of bacterial-derived signals on immune cell function and the mechanisms by which these signals modulate the development and progression of inflammatory disease. We conclude with an examination of successes and future challenges in using bacterial communities or their products in the prevention or treatment of human disease.
Collapse
Affiliation(s)
- David A Hill
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, 19104-4539, USA
| | | |
Collapse
|
507
|
Abstract
Soon after birth, the neonatal intestine is confronted with a massive antigenic challenge of microbial colonization. Microbial signals are required for maturation of several physiological, anatomical, and biochemical functions of intestinal epithelial barrier (IEB) after birth. Commensal bacteria regulate intestinal innate and adaptive immunity and provide stimuli for ongoing repair and restitution of IEB. Colonization by pathogenic bacteria and/or dysmature response to microbial stimuli can result in flagrant inflammatory response as seen in necrotizing enterocolitis (NEC). Characterized by inflammation and hemorrhagic-ischemic necrosis, NEC is a devastating complication of prematurity. Although there is evidence that both prematurity and presence of bacteria, are proven contributing factors to the pathogenesis of NEC, the molecular mechanisms involved in IEB dysfunction associated with NEC have begun to emerge only recently. The metagenomic advances in the field of intestinal microecology are providing insight into the factors that are required for establishment of commensal bacteria that appear to provide protection against intestinal inflammation and NEC. Perturbations in achieving colonization by commensal bacteria such as premature birth or hospitalization in intensive care nursery can result in dysfunction of IEB and NEC. In this article, microbial modulation of functions of IEB and its relationship with barrier dysfunction and NEC are described.
Collapse
Affiliation(s)
- Renu Sharma
- Division of Neonatology, Department of Pediatrics, University of Florida at Jacksonville, 655 West 8th Street, Jacksonville, FL 32209, USA.
| | | |
Collapse
|
508
|
Takahashi I, Nochi T, Kunisawa J, Yuki Y, Kiyono H. The mucosal immune system for secretory IgA responses and mucosal vaccine development. Inflamm Regen 2010. [DOI: 10.2492/inflammregen.30.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
509
|
Abstract
The past 20 years have seen a growing interest over the control of adaptive immune responses by the innate immune system. In particular, considerable attention has been paid to the mechanisms by which antigen-primed dendritic cells orchestrate the differentiation of T cells. Additional studies have elucidated the pathways followed by T cells to initiate immunoglobulin responses in B cells. In this review, we discuss recent advances on the mechanisms by which intestinal bacteria, epithelial cells, dendritic cells, and macrophages cross talk with intestinal T cells and B cells to induce frontline immunoglobulin A class switching and production.
Collapse
Affiliation(s)
- Alejo Chorny
- Department of Medicine, The Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
510
|
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that orchestrate innate and adaptive immune responses. The intestinal mucosa contains numerous DCs that are highly specialized in function. Mucosal DCs display a unique response to toll-like receptor ligands, are capable of driving immunoglobulin isotype switching to IgA, can imprint gut-homing receptors on T and B cells, and drive either T regulatory or Th17 cells depending on the analyzed subtype. These functions are partly cell autonomous and partly conferred by the local microenvironment. In this review, we will summarize the different DC subtypes present in the intestine and in the gut-associated lymphoid tissue (GALT), the unique characteristics of these subtypes, and how the local microenvironment can shape DC function.
Collapse
Affiliation(s)
- Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
511
|
Nicoletti C, Arques JL, Bertelli E. CX₃CR1 is critical for Salmonella-induced migration of dendritic cells into the intestinal lumen. Gut Microbes 2010; 1:131-4. [PMID: 21327020 PMCID: PMC3023593 DOI: 10.4161/gmic.1.3.11711] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 02/26/2010] [Accepted: 03/04/2010] [Indexed: 02/03/2023] Open
Abstract
We have demonstrated that direct antigen sampling of bacteria by intestinal dendritic cells (DCs) is accompanied by a rapid migration of CD11c+CX3CR1+MHCII+CD8α-CD11b− DCs into the intestinal lumen upon exposure to non-invasive ΔSPI1-Salmonella. Importantly, intraluminal DCs internalized Salmonella but were not able to cross the epithelium to return into tissue, thus showing that these DCs do not function as antigen-presenting cells and participate in the conventional regulation of immune responses to intestinal pathogens. Here we show that the presence of the chemokine receptor CX3CR1, that plays a vital role in DC-mediated antigen sampling and clearance in the gut, is also instrumental for the transepithelial migration of DCs. The latter observation, along with the notion that CX3CR1-deficient mice displayed higher susceptibility to Salmonella infection compared to wild-type mice raises the possibility that Salmonella-induced migration of “bacteria-capturing” DCs into the lumen may be an important mechanism of mucosal defence and clearance.
Collapse
Affiliation(s)
- Claudio Nicoletti
- Laboratory of Mucosal Immunology; Programme of Integrated Biology of the GI Tract; Institute of Food Research; Norwich, UK
| | - Juan L Arques
- Laboratory of Mucosal Immunology; Programme of Integrated Biology of the GI Tract; Institute of Food Research; Norwich, UK,Department Tecnología de Alimentos; INIA; Madrid, Spain
| | - Eugenio Bertelli
- Department of Pharmacology “G. Segre”; University of Siena; Siena, Italy
| |
Collapse
|
512
|
Sung SS, Bolton WK. T cells and dendritic cells in glomerular disease: the new glomerulotubular feedback loop. Kidney Int 2009; 77:393-9. [PMID: 20032960 DOI: 10.1038/ki.2009.489] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A newly described glomerulotubular feedback loop may explain the relationship between glomerular damage, epitope spreading, tubulointerstitial nephritis, proteinuria as a progression factor, and the importance of the local milieu in kidney damage. It also opens the horizons for exciting innovative approaches to therapy of both acute and chronic kidney diseases.
Collapse
Affiliation(s)
- Sun-Sang Sung
- Division of Nephrology, University of Virginia, Charlottesville, Virginia 22908-0133, USA
| | | |
Collapse
|
513
|
Belyakov IM, Ahlers JD. What role does the route of immunization play in the generation of protective immunity against mucosal pathogens? THE JOURNAL OF IMMUNOLOGY 2009; 183:6883-92. [PMID: 19923474 DOI: 10.4049/jimmunol.0901466] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The route of vaccination is important in influencing immune responses at the initial site of pathogen invasion where protection is most effective. Immune responses required for mucosal protection can differ vastly depending on the individual pathogen. For some mucosal pathogens, including acute self-limiting infections, high-titer neutralizing Abs that enter tissue parenchyma or transude into the mucosal lumen are sufficient for clearing cell-free virus. However, for pathogens causing chronic infections such as HIV, hepatitis C virus, herpes viruses, mycobacteria, and fungal and parasitic infections, a single arm of the immune response generated by systemic vaccination may be insufficient for protection. Induction of the mucosal innate and adaptive immune systems, including CD4+ T help, Th17, high avidity CD8+ CTL, and secretory IgA and IgG1 neutralizing Abs, at the site of pathogen entry may be required for effective protection against highly invasive pathogens that lead to chronic infection and may be generated predominantly by mucosal vaccination.
Collapse
Affiliation(s)
- Igor M Belyakov
- Midwest Research Institute, 110 Thomas Johnson Drive, Frederick, MD 21702, USA.
| | | |
Collapse
|
514
|
Onodera T, Jang MH, Guo Z, Yamasaki M, Hirata T, Bai Z, Tsuji NM, Nagakubo D, Yoshie O, Sakaguchi S, Takikawa O, Miyasaka M. Constitutive expression of IDO by dendritic cells of mesenteric lymph nodes: functional involvement of the CTLA-4/B7 and CCL22/CCR4 interactions. THE JOURNAL OF IMMUNOLOGY 2009; 183:5608-14. [PMID: 19843945 DOI: 10.4049/jimmunol.0804116] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dendritic cells (DCs) express the immunoregulatory enzyme IDO in response to certain inflammatory stimuli, but it is unclear whether DCs express this enzyme under steady-state conditions in vivo. In this study, we report that the DCs in mesenteric lymph nodes (MLNs) constitutively express functional IDO, which metabolizes tryptophan to kynurenine. In line with a previous report that regulatory T cells (Tregs) can induce IDO in DCs via the CTLA-4/B7 interaction, a substantial proportion of the MLN DCs were located in juxtaposition to Tregs, whereas this tendency was not observed for splenic DCs, which do not express IDO constitutively. When CTLA-4 was selectively deleted in Tregs, the frequency of IDO-expressing DCs in MLNs decreased significantly, confirming CTLA-4's role in IDO expression by MLN DCs. We also found that the MLN DCs produced CCL22, which can attract Tregs via CCR4, and that the phagocytosis of autologous apoptotic cells induced CCL22 expression in CCL22 mRNA-negative DCs. Mice genetically deficient in the receptor for CCL22, CCR4, showed markedly reduced IDO expression in MLN-DCs, supporting the involvement of the CCL22/CCR4 axis in IDO induction. Together with our previous observation that MLN DCs contain much intracytoplasmic cellular debris in vivo, these results indicate that reciprocal interactions between the DCs and Tregs via both B7/CTLA-4 and CCL22/CCR4 lead to IDO induction in MLN DCs, which may be initiated and/or augmented by the phagocytosis of autologous apoptotic cells by intestinal DCs. Such a mechanism may help induce the specific milieu in MLNs that is required for the induction of oral tolerance.
Collapse
Affiliation(s)
- Toshiharu Onodera
- Laboratory of Immunodynamics, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
515
|
Sonier B, Patrick C, Ajjikuttira P, Scott FW. Intestinal Immune Regulation as a Potential Diet-Modifiable Feature of Gut Inflammation and Autoimmunity. Int Rev Immunol 2009; 28:414-45. [DOI: 10.3109/08830180903208329] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
516
|
Abstract
Mucosal surfaces are lined by epithelial cells. These cells establish a barrier between sometimes hostile external environments and the internal milieu. However, mucosae are also responsible for nutrient absorption and waste secretion, which require a selectively permeable barrier. These functions place the mucosal epithelium at the centre of interactions between the mucosal immune system and luminal contents, including dietary antigens and microbial products. Recent advances have uncovered mechanisms by which the intestinal mucosal barrier is regulated in response to physiological and immunological stimuli. Here I discuss these discoveries along with evidence that this regulation shapes mucosal immune responses in the gut and, when dysfunctional, may contribute to disease.
Collapse
Affiliation(s)
- Jerrold R Turner
- Department of Pathology, The University of Chicago, 5841 South Maryland, MC 1089, Chicago, Illinois 60637, USA.
| |
Collapse
|
517
|
Oldenhove G, Bouladoux N, Wohlfert EA, Hall JA, Chou D, Dos Santos L, O'Brien S, Blank R, Lamb E, Natarajan S, Kastenmayer R, Hunter C, Grigg ME, Belkaid Y. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity 2009; 31:772-86. [PMID: 19896394 DOI: 10.1016/j.immuni.2009.10.001] [Citation(s) in RCA: 473] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/25/2009] [Accepted: 09/15/2009] [Indexed: 01/08/2023]
Abstract
Using a model of lethal oral infection with Toxoplasma gondii, we examined the fate of both induced and natural regulatory T (Treg) cells in the face of strong inflammatory responses occurring in a tolerogenic-prone environment. We found that during highly T helper 1 (Th1) cell-polarized mucosal immune responses, Treg cell numbers collapsed via multiple pathways, including blockade of Treg cell induction and disruption of endogenous Treg cell homeostasis. In particular, shutdown of interleukin 2 (IL-2) in the highly Th1 cell-polarized environment triggered by infection directly contributes to Treg cell incapacity to suppress effector responses and eventually leads to immunopathogenesis. Furthermore, we found that environmental cues provided by both local dendritic cells and effector T cells can induce the expression of T-bet transcription factor and IFN-gamma by Treg cells. These data reveal a mechanism for Th1 cell pathogenicity that extends beyond their proinflammatory program to limit Treg cell survival.
Collapse
Affiliation(s)
- Guillaume Oldenhove
- Mucosal Immunology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
518
|
Rescigno M, Iliev ID. Interleukin-23: linking mesenteric lymph node dendritic cells with Th1 immunity in Crohn's disease. Gastroenterology 2009; 137:1566-70. [PMID: 19789081 DOI: 10.1053/j.gastro.2009.09.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
519
|
The intestinal microbiota in health and disease: the influence of microbial products on immune cell homeostasis. Curr Opin Gastroenterol 2009; 25:496-502. [PMID: 19770652 PMCID: PMC4737592 DOI: 10.1097/mog.0b013e328331b6b4] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW A vast and diverse array of microbes colonizes the mammalian gastrointestinal tract. These microorganisms are integral in shaping the development and function of the immune system. Metagenomic sequencing analysis has revealed alterations in intestinal microbiota in patients suffering from chronic inflammatory diseases, including inflammatory bowel disease and asthma. This review will discuss the mechanisms through which the innate immune system recognizes and responds to the intestinal microbiota as well as the effect of specific microbiota-derived signals on immune cell homeostasis. RECENT FINDINGS Recent studies in murine model systems have demonstrated that manipulation of the intestinal microbiota can alter mammalian immune cell homeostasis. Specific microbial signals have been identified that can impact immune cell function both within the intestinal tract and in peripheral tissues. These microbiota-derived signals can either have an immunoregulatory effect, creating an immune state that is refractory to inflammation, or conversely, act as an adjuvant, aiding in the propagation of an immune response. SUMMARY Associations between alterations in the microbiota and human disease implicate intestinal microbial signals in shaping immune responses. These signals are recognized by innate immune cells and influence the ability of these cells to modulate both the local and systemic immune response.
Collapse
|
520
|
Hirata N, Yanagawa Y, Satoh M, Ogura H, Ebihara T, Noguchi M, Matsumoto M, Togashi H, Seya T, Onoé K, Iwabuchi K. Dendritic cell-derived TNF-alpha is responsible for development of IL-10-producing CD4+ T cells. Cell Immunol 2009; 261:37-41. [PMID: 19931858 DOI: 10.1016/j.cellimm.2009.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 10/21/2009] [Indexed: 11/26/2022]
Abstract
Immature dendritic cells (DCs) appear to be involved in peripheral immune tolerance via induction of IL-10-producing CD4(+) T cells. We examined the role of TNF-alpha in generation of the IL-10-producing CD4(+) T cells by immature DCs. Immature bone marrow-derived DCs from wild type (WT) or TNF-alpha(-/-) mice were cocultured with CD4(+) T cells from OVA specific TCR transgenic mice (OT-II) in the presence of OVA(323-339) peptide. The WT DCs efficiently induced the antigen-specific IL-10-producing CD4(+) T cells, while the ability of the TNF-alpha(-/-) DCs to induce these CD4(+) T cells was considerably depressed. Addition of exogenous TNF-alpha recovered the impaired ability of the TNF-alpha(-/-) DCs to induce IL-10-producing T cells. However, no difference in this ability was observed between TNF-alpha(-/-) and WT DCs after their maturation by LPS. Thus, TNF-alpha appears to be critical for the generation of IL-10-producing CD4(+) T cells during the antigen presentation by immature DCs.
Collapse
Affiliation(s)
- Noriyuki Hirata
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
521
|
Ochoa-Repáraz J, Mielcarz DW, Ditrio LE, Burroughs AR, Foureau DM, Haque-Begum S, Kasper LH. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2009; 183:6041-50. [PMID: 19841183 DOI: 10.4049/jimmunol.0900747] [Citation(s) in RCA: 419] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mucosal tolerance has been considered a potentially important pathway for the treatment of autoimmune disease, including human multiple sclerosis and experimental conditions such as experimental autoimmune encephalomyelitis (EAE). There is limited information on the capacity of commensal gut bacteria to induce and maintain peripheral immune tolerance. Inbred SJL and C57BL/6 mice were treated orally with a broad spectrum of antibiotics to reduce gut microflora. Reduction of gut commensal bacteria impaired the development of EAE. Intraperitoneal antibiotic-treated mice showed no significant decline in the gut microflora and developed EAE similar to untreated mice, suggesting that reduction in disease activity was related to alterations in the gut bacterial population. Protection was associated with a reduction of proinflammatory cytokines and increases in IL-10 and IL-13. Adoptive transfer of low numbers of IL-10-producing CD25(+)CD4(+) T cells (>75% FoxP3(+)) purified from cervical lymph nodes of commensal bacteria reduced mice and in vivo neutralization of CD25(+) cells suggested the role of regulatory T cells maintaining peripheral immune homeostasis. Our data demonstrate that antibiotic modification of gut commensal bacteria can modulate peripheral immune tolerance that can protect against EAE. This approach may offer a new therapeutic paradigm in the treatment of multiple sclerosis and perhaps other autoimmune conditions.
Collapse
|
522
|
Gut commensal bacteria direct a protective immune response against Toxoplasma gondii. Cell Host Microbe 2009; 6:187-96. [PMID: 19683684 DOI: 10.1016/j.chom.2009.06.005] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 03/15/2009] [Accepted: 06/01/2009] [Indexed: 01/07/2023]
Abstract
Toxoplasma gondii is a universally distributed pathogen that infects over one billion people worldwide. Host resistance to this protozoan parasite depends on a Th1 immune response with potent production of the cytokines interleukin-12 and interferon gamma. Although Toll-like receptor 11 (TLR11) plays a major role in controlling Th1 immunity to this pathogen in mice, this innate immune receptor is nonfunctional in humans, and the mechanisms of TLR11-independent sensing of T. gondii remain elusive. Here, we show that oral infection by T. gondii triggers a TLR11-independent but MyD88-dependent Th1 response that is impaired in TLR2xTLR4 double knockout and TLR9 single knockout mice. These mucosal innate and adaptive immune responses to T. gondii rely on the indirect stimulation of dendritic cells by normal gut microflora. Thus, our results reveal that gut commensal bacteria can serve as molecular adjuvants during parasitic infection, providing indirect immunostimulation that protects against T. gondii in the absence of TLR11.
Collapse
|
523
|
Accumulation of DC in Lamina Propria Induced by FMS-Like Tyrosine Kinase 3 Ligand Aggravates the Intestinal Inflammatory Response During Endotoxemia. Inflammation 2009; 33:34-45. [DOI: 10.1007/s10753-009-9156-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
524
|
T cell cross-talk with kidney dendritic cells in glomerulonephritis. J Mol Med (Berl) 2009; 88:19-26. [DOI: 10.1007/s00109-009-0541-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 09/04/2009] [Accepted: 09/07/2009] [Indexed: 12/28/2022]
|
525
|
Abstract
Regulatory T cells help maintain intestinal homeostasis by preventing inappropriate innate and adaptive immune responses. CD4(+) T cells that express Foxp3 and Tr1-like cells that produce IL-10 comprise the major regulatory populations in the intestine. CD4(+)Foxp3(+) T cells play an important functional role in promoting tolerance of the flora and dietary proteins. Tr1-like cells can be generated in conditions that also promote effector T cell responses and may serve a similar function. In this review, we discuss the signals specific to the gastrointestinal tract that support both regulatory cell types and their distinct modes of action in the mesenteric lymph nodes and intestinal tissues. Dysregulation of intestinal immune homeostasis occurs in inflammatory bowel disease and can also be observed in graft-versus-host disease, tumor immunotherapy regimens, and acute HIV infection.
Collapse
Affiliation(s)
- Michael J Barnes
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK
| | | |
Collapse
|
526
|
Abstract
Over the last decade, it has become apparent that the complex interactions between components of the mucosal microflora and the mucosal immune system can involve either direct contact with dendritic cells in the lamina propria or, alternatively, contact with epithelial cells lining the mucosa that then influence the function of dendritic cells. Although in some cases these interactions involve signaling specific to particular organisms and in others, to classes of organisms, a common theme is that signaling is invariably channeled through receptors that address many organisms or all organisms such as the pattern-recognition receptors TLR and NLR. Here, I review this information with the intention of identifying how the mucosal microflora influences specific functions of the mucosal immune system such the production of particular cytokines as well as broader functions such as the maintenance of mucosal immune homeostasis and host defense.
Collapse
Affiliation(s)
- Warren Strober
- Mucosal Immunity Section, Laboratory of Host Defenses, NIAID, NIH Bethesda, MD, USA.
| |
Collapse
|
527
|
Westendorf AM, Fleissner D, Hansen W, Buer J. T cells, dendritic cells and epithelial cells in intestinal homeostasis. Int J Med Microbiol 2009; 300:11-8. [PMID: 19781991 DOI: 10.1016/j.ijmm.2009.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mucosal immune system of the intestinal tract is continuously exposed to both potential pathogens and beneficial commensal microorganism. A variety of mechanisms contribute to the ability of the gut to either react or remain tolerant to antigen present in the intestinal lumen. Antigens of the gut commensals are not simply ignored, but rather trigger an active immunosuppressive process, which prevents the outcome of immunopathology. The aim of this review is to provide an update on the mechanism of intestinal homeostasis, with particular focus on the complex crosstalk between T cells, dendritic cells and intestinal epithelial cells.
Collapse
Affiliation(s)
- Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, Hufelandstrasse 55, D-45122 Essen, Germany.
| | | | | | | |
Collapse
|
528
|
Balic A, Smith KA, Harcus Y, Maizels RM. Dynamics of CD11c(+) dendritic cell subsets in lymph nodes draining the site of intestinal nematode infection. Immunol Lett 2009; 127:68-75. [PMID: 19766674 PMCID: PMC2789245 DOI: 10.1016/j.imlet.2009.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 08/21/2009] [Accepted: 09/08/2009] [Indexed: 11/13/2022]
Abstract
Helminth parasites drive dominant Th2 responses through an as yet unidentified pathway. We have previously shown that the rodent gastrointestinal nematode Nippostrongylus brasiliensis secretes products which selectively activate in vitro-derived dendritic cells to induce Th2 responses on in vivo transfer. We now show that, during active infection with this parasite, the draining mesenteric lymph node dendritic cell population is altered significantly. Although there is substantial expansion of DC numbers during infection, the CD86hi-CD8αint-CD11b− subset is markedly diminished, and expression levels of CD40, CD86 and CD103 are reduced. Notably, the reduced frequency of CD8αint DCs is evident only in those mesenteric lymph nodes draining the anterior site of infestation. In infections with the longer lived Heligmosomoides polygyrus, the proportion of CD8αint DCs in the MLNC falls to below 10% of total DC numbers by 35 days post-infection. Further, infection alters TLR responsiveness, as IL-12 production (as measured by ex vivo intracellular staining of CD11c+ DCs) in response to LPS stimulation is reduced, while IL-6, TNF-α and in particular, IL-10 all increase following infection with either nematode parasite. These changes suggest the possibility that helminth parasites modulate gastrointestinal immunity both by inhibiting migration of CD8αint DCs to the draining lymph nodes, and modifying DC responsiveness in a manner which favours a Th2 outcome.
Collapse
Affiliation(s)
- Adam Balic
- Institute of Immunology and Infection Research, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | | | | | | |
Collapse
|
529
|
Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M. Origin of the lamina propria dendritic cell network. Immunity 2009; 31:513-25. [PMID: 19733489 DOI: 10.1016/j.immuni.2009.08.010] [Citation(s) in RCA: 681] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/27/2009] [Accepted: 08/17/2009] [Indexed: 12/11/2022]
Abstract
CX(3)CR1(+) and CD103(+) dendritic cells (DCs) in intestinal lamina propria play a key role in mucosal immunity. However, the origin and the developmental pathways that regulate their differentiation in the lamina propria remain unclear. We showed that monocytes gave rise exclusively to CD103(-)CX(3)CR1(+) lamina propria DCs under the control of macrophage-colony-stimulating factor receptor (M-CSFR) and Fms-like thyrosine kinase 3 (Flt3) ligands. In contrast, common DC progenitors (CDP) and pre-DCs, which give rise to lymphoid organ DCs but not to monocytes, differentiated exclusively into CD103(+)CX(3)CR1(-) lamina propria DCs under the control of Flt3 and granulocyte-macrophage-colony-stimulating factor receptor (GM-CSFR) ligands. CD103(+)CX(3)CR1(-) DCs but not CD103(-)CX(3)CR1(+) DCs in the lamina propria constitutively expressed CCR7 and were the first DCs to transport pathogenic Salmonella from the intestinal tract to the mesenteric lymph nodes. Altogether, these results underline the diverse origin of the lamina propria DC network and identify mucosal DCs that arise from pre-DCs as key sentinels of the gut immune system.
Collapse
Affiliation(s)
- Milena Bogunovic
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
530
|
Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity 2009; 31:502-12. [PMID: 19733097 DOI: 10.1016/j.immuni.2009.06.025] [Citation(s) in RCA: 569] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 05/31/2009] [Accepted: 06/22/2009] [Indexed: 12/12/2022]
Abstract
The intestinal immune system discriminates between tolerance toward the commensal microflora and robust responses to pathogens. Maintenance of this critical balance is attributed to mucosal dendritic cells (DCs) residing in organized lymphoid tissue and dispersed in the subepithelial lamina propria. In situ parameters of lamina propria DCs (lpDCs) remain poorly understood. Here, we combined conditional cell ablation and precursor-mediated in vivo reconstitution to establish that lpDC subsets have distinct origins and functions. CD103(+) CX(3)CR1(-) lpDCs arose from macrophage-DC precursors (MDPs) via DC-committed intermediates (pre-cDCs) through a Flt3L growth-factor-mediated pathway. CD11b(+) CD14(+) CX(3)CR1(+) lpDCs were derived from grafted Ly6C(hi) but not Ly6C(lo) monocytes under the control of GM-CSF. Mice reconstituted exclusively with CX(3)CR1(+) lpDCs when challenged in an innate colitis model developed severe intestinal inflammation that was driven by graft-derived TNF-alpha-secreting CX(3)CR1(+) lpDCs. Our results highlight the critical importance of the lpDC subset balance for robust gut homeostasis.
Collapse
Affiliation(s)
- Chen Varol
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel 76100
| | | | | | | | | | | | | | | | | | | |
Collapse
|
531
|
Gorfu G, Rivera-Nieves J, Ley K. Role of β7 Integrins in Intestinal Lymphocyte Homing and Retention. Curr Mol Med 2009; 9:836-50. [DOI: 10.2174/156652409789105525] [Citation(s) in RCA: 225] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
532
|
Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A 2009; 106:15813-8. [PMID: 19805227 DOI: 10.1073/pnas.0907722106] [Citation(s) in RCA: 448] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the Nod2 gene are among the strongest genetic risk factors in the pathogenesis of ileal Crohn's disease, but the exact contributions of Nod2 to intestinal mucosal homeostasis are not understood. Here we show that Nod2 plays an essential role in controlling commensal bacterial flora in the intestine. Analysis of intestinal bacteria from the terminal ilea of Nod2-deficient mice showed that they harbor an increased load of commensal resident bacteria. Furthermore, Nod2-deficient mice had a diminished ability to prevent intestinal colonization of pathogenic bacteria. In vitro, intestinal crypts isolated from terminal ilea of Nod2-deficient mice were unable to kill bacteria effectively, suggesting an important role of Nod2 signaling in crypt function. Interestingly, the expression of Nod2 is dependent on the presence of commensal bacteria, because mice re-derived into germ-free conditions expressed significantly less Nod2 in their terminal ilea, and complementation of commensal bacteria into germ-free mice induced Nod2 expression. Therefore, Nod2 and intestinal commensal bacterial flora maintain a balance by regulating each other through a feedback mechanism. Dysfunction of Nod2 results in a break-down of this homeostasis.
Collapse
|
533
|
Rescigno M, Di Sabatino A. Dendritic cells in intestinal homeostasis and disease. J Clin Invest 2009; 119:2441-50. [PMID: 19729841 DOI: 10.1172/jci39134] [Citation(s) in RCA: 230] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DCs are specialized APCs that orchestrate innate and adaptive immune responses. The intestinal mucosa contains numerous DCs, which induce either protective immunity to infectious agents or tolerance to innocuous antigens, including food and commensal bacteria. Several subsets of mucosal DCs have been described that display unique functions, dictated in part by the local microenvironment. In this review, we summarize the distinct subtypes of DCs and their distribution in the gut; examine how DC dysfunction contributes to intestinal disease development, including inflammatory bowel disease and celiac disease; and discuss manipulation of DCs for therapy.
Collapse
Affiliation(s)
- Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.
| | | |
Collapse
|
534
|
Fortin G, Raymond M, Van VQ, Rubio M, Gautier P, Sarfati M, Franchimont D. A role for CD47 in the development of experimental colitis mediated by SIRPalpha+CD103- dendritic cells. ACTA ACUST UNITED AC 2009; 206:1995-2011. [PMID: 19703989 PMCID: PMC2737153 DOI: 10.1084/jem.20082805] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenteric lymph node (mLN) CD103 (alphaE integrin)(+) dendritic cells (DCs) induce regulatory T cells and gut tolerance. However, the function of intestinal CD103(-) DCs remains to be clarified. CD47 is the ligand of signal regulatory protein alpha (SIRPalpha) and promotes SIRPalpha(+) myeloid cell migration. We first show that mucosal CD103(-) DCs selectively express SIRPalpha and that their frequency was augmented in the lamina propria and mLNs of mice that developed Th17-biased colitis in response to trinitrobenzene sulfonic acid. In contrast, the percentage of SIRPalpha(+)CD103(-) DCs and Th17 responses were decreased in CD47-deficient (CD47 knockout [KO]) mice, which remained protected from colitis. We next demonstrate that transferring wild-type (WT), but not CD47 KO, SIRPalpha(+)CD103(-) DCs in CD47 KO mice elicited severe Th17-associated wasting disease. CD47 expression was required on the SIRPalpha(+)CD103(-) DCs for efficient trafficking to mLNs in vivo, whereas it was dispensable on both DCs and T cells for Th17 polarization in vitro. Finally, administration of a CD47-Fc molecule resulted in reduced SIRPalpha(+)CD103(-) DC-mediated Th17 responses and the protection of WT mice from colitis. We thus propose SIRPalpha(+)CD103(-) DCs as a pathogenic DC subset that drives Th17-biased responses and colitis, and the CD47-SIRPalpha axis as a potential therapeutic target for inflammatory bowel disease.
Collapse
Affiliation(s)
- Genevieve Fortin
- Research Institute of McGill University Health Centre, McGill University, Montreal H3H 2R9, Canada
| | | | | | | | | | | | | |
Collapse
|
535
|
Type II collagen oral tolerance; mechanism and role in collagen-induced arthritis and rheumatoid arthritis. Mod Rheumatol 2009; 19:581-9. [PMID: 19697097 DOI: 10.1007/s10165-009-0210-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 07/08/2009] [Indexed: 12/24/2022]
Abstract
Oral tolerance means a diminished immune response to previously fed antigens. Repeated oral administrations of type II collagen (CII) induce oral tolerance and inhibit the development of collagen-induced arthritis (CIA). Dendritic cells (DCs) in the gut-associated lymphoid tissue (GALT) take up the CII and then present it to T cells to generate regulatory T cells (Tregs), which induce systemic immune tolerance to CII. Inhibitory cytokines, such as transforming growth factor (TGF)-beta and interleukin (IL)-10, and several immune regulatory molecules, including indoleamine 2,3-dioxygenase (IDO) and retinoic acid, play an important role in Treg generation. Each DC subset may play different roles, and CD11c+CD11b+DCs and IDO+DCs are important in the generation of antigen-inducible Tregs in CII oral tolerance. Upon stimulation with the antigen involved in its generation, Treg is activated and regulates the immune response through inhibitory cytokine production, cell-to-cell contact-dependent mechanisms, DC modification, and bystander suppression. The DCs and Tregs are deeply involved in oral tolerance through reciprocal interactions. Several clinical trials have been conducted in RA patients to examine the efficacy of CII oral tolerance. An understanding the mechanism of oral tolerance to CII would give clinicians new insights into the development of natural immune tolerance and new therapeutic approaches for the treatment of autoimmune diseases.
Collapse
|
536
|
Schiffrin EJ, Morley JE, Donnet-Hughes A, Guigoz Y. The inflammatory status of the elderly: the intestinal contribution. Mutat Res 2009; 690:50-6. [PMID: 19666034 DOI: 10.1016/j.mrfmmm.2009.07.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 07/22/2009] [Accepted: 07/31/2009] [Indexed: 12/23/2022]
Abstract
A common finding in the elderly population is a chronic subclinical inflammatory status that coexists with immune dysfunction. These interconnected processes are of sufficient magnitude to impact health and survival time. In this review we discuss the different signals that may stimulate the inflammatory process in the aging population as well as the molecular and cellular components that can participate in the initiation, the modulation or termination of the said process. A special interest has been devoted to the intestine as a source of signals that can amplify local and systemic inflammation. Sentinel cells in the splanchnic area are normally exposed to more than one stimulus at a given time. In the intestine of the elderly, endogenous molecules produced by the cellular aging process and stress as well as exogenous evolutionarily conserved molecules from bacteria, are integrated into a network of receptors and molecular signalling pathways that result in chronic inflammatory activation. It is thus possible that nutritional interventions which modify the intestinal ecology can diminish the pro-inflammatory effects of the microbiota and thereby reinforce the mucosal barrier or modulate the cellular activation pathways.
Collapse
Affiliation(s)
- Eduardo J Schiffrin
- Nestlé Nutrition, HealthCare Nutrition, Route des Avouillons 30, CH-1196 Gland, Nestec Ltd., Vevey, Switzerland
| | | | | | | |
Collapse
|
537
|
|
538
|
McNeilly TN, Devaney E, Matthews JB. Teladorsagia circumcincta in the sheep abomasum: defining the role of dendritic cells in T cell regulation and protective immunity. Parasite Immunol 2009; 31:347-56. [PMID: 19527450 DOI: 10.1111/j.1365-3024.2009.01110.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Parasitic nematodes of the small-ruminant gastrointestinal tract pose a problem worldwide. The impact of these pathogens is worsened by the emergence of anthelmintic resistance to all three available classes of drugs. In addition to causing considerable economic loss, these parasites are detrimental to the health and welfare of sheep and goats. Vaccination offers an alternative approach to drug-based control and a great deal of investment has gone into the investigation of protective antigens for some of these nematode species. However, attempts at vaccination are hindered by a lack of understanding of how best to promote protective immunity to nematode species, such as Teladorsagia circumcincta, which inhabits the abomasum of sheep. This situation contrasts with that in murine models of gastrointestinal nematode infection, where the basis of protective immunity is increasingly well understood. In this review, we discuss the current knowledge of the immune effector mechanisms elicited by T. circumcincta and consider the probable role of dendritic cells in the initiation of both effector and regulatory responses in the abomasum.
Collapse
Affiliation(s)
- T N McNeilly
- Moredun Research Institute, Pentlands Science Park, Midlothian, UK
| | | | | |
Collapse
|
539
|
Current World Literature. Curr Opin Allergy Clin Immunol 2009; 9:386-90. [DOI: 10.1097/aci.0b013e32832eb836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
540
|
Boge T, Rémigy M, Vaudaine S, Tanguy J, Bourdet-Sicard R, van der Werf S. A probiotic fermented dairy drink improves antibody response to influenza vaccination in the elderly in two randomised controlled trials. Vaccine 2009; 27:5677-84. [PMID: 19615959 DOI: 10.1016/j.vaccine.2009.06.094] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/12/2009] [Accepted: 06/29/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND Influenza vaccination is recommended for the elderly in many countries, but immune responses are weaker compared to younger adults. OBJECTIVE To investigate the impact of daily consumption of a probiotic dairy drink on the immune response to influenza vaccination in an elderly population of healthy volunteers over 70 years of age. DESIGN Two randomised, multicentre, double-blind, controlled studies were conducted during two vaccination seasons in 2005-2006 (pilot) and 2006-2007 (confirmatory). Eighty-six and 222 elderly volunteers consumed either a fermented dairy drink, containing the probiotic strain Lactobacillus casei DN-114 001 and yoghurt ferments (Actimel, or a non-fermented control dairy product twice daily for a period of 7 weeks (pilot) or 13 weeks (confirmatory). Vaccination occurred after 4 weeks of product consumption. Geometric mean antibody titres (GMT) against the 3 viral strains composing the vaccine (H1N1, H3N2, and B) were measured at several time intervals post-vaccination by haemagglutination inhibition test. RESULTS In the pilot study, the influenza-specific antibody titres increased after vaccination, being consistently higher in the probiotic product group compared to the control group under product consumption. Similarly, in the confirmatory study, titres against the B strain increased significantly more in the probiotic group than in the control group at 3, 6 and 9 weeks post-vaccination under product consumption (p=0.020). Significant differences in seroconversion between the groups by intended to treat analysis were still found 5 months after vaccination. Similar GMT results were observed for the H3N2 strain and H1N1 strain, confirming the results of the pilot study. CONCLUSION These studies demonstrate that daily consumption of this particular probiotic product increased relevant specific antibody responses to influenza vaccination in individuals of over 70 years of age and may therefore provide a health benefit in this population.
Collapse
Affiliation(s)
- Thierry Boge
- Centre Hospitalier de Bourg en Bresse, 01012 Bourg en Bresse, France
| | | | | | | | | | | |
Collapse
|
541
|
Noti M, Sidler D, Brunner T. Extra-adrenal glucocorticoid synthesis in the intestinal epithelium: more than a drop in the ocean? Semin Immunopathol 2009; 31:237-48. [PMID: 19495759 DOI: 10.1007/s00281-009-0159-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 05/14/2009] [Indexed: 12/31/2022]
Abstract
Glucocorticoids (GC) are lipophilic hormones commonly used as therapeutics in acute and chronic inflammatory disorders such as inflammatory bowel disease due to their attributed anti-inflammatory and immunosuppressive actions. Although the adrenal glands are the major source of endogenous GC, there is increasing evidence for the production of extra-adrenal GC in the brain, thymus, skin, vasculature, and the intestine. However, the physiological relevance of extra-adrenal-produced GC remains still ambiguous. Therefore, this review attracts attention to discuss possible biological benefits of extra-adrenal-synthesized GC, especially focusing on the impact of locally synthesized GC in the regulation of intestinal immune responses.
Collapse
Affiliation(s)
- Mario Noti
- Division of Immunopathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
542
|
Ungaro R, Fukata M, Hsu D, Hernandez Y, Breglio K, Chen A, Xu R, Sotolongo J, Espana C, Zaias J, Elson G, Mayer L, Kosco-Vilbois M, Abreu MT. A novel Toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1167-79. [PMID: 19359427 PMCID: PMC2697943 DOI: 10.1152/ajpgi.90496.2008] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dysregulated innate immune responses to commensal bacteria contribute to the development of inflammatory bowel disease (IBD). TLR4 is overexpressed in the intestinal mucosa of IBD patients and may contribute to uncontrolled inflammation. However, TLR4 is also an important mediator of intestinal repair. The aim of this study is to examine the effect of a TLR4 antagonist on inflammation and intestinal repair in two murine models of IBD. Colitis was induced in C57BL/6J mice with dextran sodium sulfate (DSS) or by transferring CD45Rb(hi) T cells into RAG1-/- mice. An antibody (Ab) against the TLR4/MD-2 complex or isotype control Ab was administered intraperitoneally during DSS treatment, recovery from DSS colitis, or induction of colitis in RAG1-/- mice. Colitis severity was assessed by disease activity index (DAI) and histology. The effect of the Ab on the inflammatory infiltrate was determined by cell isolation and immunohistochemistry. Mucosal expression of inflammatory mediators was analyzed by real-time PCR and ELISA. Blocking TLR4 at the beginning of DSS administration delayed the development of colitis with significantly lower DAI scores. Anti-TLR4 Ab treatment decreased macrophage and dendritic cell infiltrate and reduced mucosal expression of CCL2, CCL20, TNF-alpha, and IL-6. Anti-TLR4 Ab treatment during recovery from DSS colitis resulted in defective mucosal healing with lower expression of COX-2, PGE(2), and amphiregulin. In contrast, TLR4 blockade had minimal efficacy in ameliorating inflammation in the adoptive transfer model of chronic colitis. Our findings suggest that anti-TLR4 therapy may decrease inflammation in IBD but may also interfere with colonic mucosal healing.
Collapse
Affiliation(s)
- Ryan Ungaro
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Masayuki Fukata
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - David Hsu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Yasmin Hernandez
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Keith Breglio
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Anli Chen
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Ruliang Xu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - John Sotolongo
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Cecillia Espana
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Julia Zaias
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Greg Elson
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Lloyd Mayer
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Marie Kosco-Vilbois
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Maria T. Abreu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| |
Collapse
|
543
|
New horizon of mucosal immunity and vaccines. Curr Opin Immunol 2009; 21:352-8. [PMID: 19493665 DOI: 10.1016/j.coi.2009.04.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 04/27/2009] [Indexed: 11/23/2022]
Abstract
Progress in the past quarter-century on understanding the molecular, cellular, and in vivo components of the mucosal immune system have allowed us to develop a practical strategy for a novel mucosal vaccine. The mucosal immune system can induce secretory IgA (SIgA) and serum IgG responses to provide two layers of defense against mucosal pathogens. For SIgA-mediated immunity in the gastrointestinal tract, the gut-associated lymphoid tissue contains both the tissue-dependent and tissue-independent IgA components. Harnessing the mucosal immune system for vaccine development may help prevent the global health problems caused by enteric infectious diseases. We have therefore combined mucosal immunology and plant biology to create a rice-based mucosal vaccine that requires neither needles and syringes nor refrigeration.
Collapse
|
544
|
Barnes MJ, Krebs P, Harris N, Eidenschenk C, Gonzalez-Quintial R, Arnold CN, Crozat K, Sovath S, Moresco EM, Theofilopoulos AN, Beutler B, Hoebe K. Commitment to the regulatory T cell lineage requires CARMA1 in the thymus but not in the periphery. PLoS Biol 2009; 7:e51. [PMID: 19260764 PMCID: PMC2650725 DOI: 10.1371/journal.pbio.1000051] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Accepted: 01/19/2009] [Indexed: 12/11/2022] Open
Abstract
Regulatory T (Treg) cells expressing forkhead box P3 (Foxp3) arise during thymic selection among thymocytes with modestly self-reactive T cell receptors. In vitro studies suggest Foxp3 can also be induced among peripheral CD4+ T cells in a cytokine dependent manner. Treg cells of thymic or peripheral origin may serve different functions in vivo, but both populations are phenotypically indistinguishable in wild-type mice. Here we show that mice with a Carma1 point mutation lack thymic CD4+Foxp3+ Treg cells and demonstrate a cell-intrinsic requirement for CARMA1 in thymic Foxp3 induction. However, peripheral Carma1-deficient Treg cells could be generated and expanded in vitro in response to the cytokines transforming growth factor beta (TGFβ) and interleukin-2 (IL-2). In vivo, a small peripheral Treg pool existed that was enriched at mucosal sites and could expand systemically after infection with mouse cytomegalovirus (MCMV). Our data provide genetic evidence for two distinct mechanisms controlling regulatory T cell lineage commitment. Furthermore, we show that peripheral Treg cells are a dynamic population that may expand to limit immunopathology or promote chronic infection. In mammals, CD4+ T cells are essential for controlling infections, but have the potential to attack host tissues as well, resulting in autoimmune disease. A subset of CD4+ T cells, regulatory T cells (Treg)—identified by the expression of the forkhead transcription factor Foxp3—serve to prevent immunopathology by dampening immune responses. These cells are unique among CD4+ T cell subsets, as only the Treg lineage can develop in both the thymus and periphery. Using a genetic approach, we identified a mutation in the gene Carma1, a key component of T and B cell signaling, which in mice distinguishes Treg cells derived from the periphery from thymic-derived regulatory T cells. The mutation caused an absence of thymic Treg cells. However, a small population of Treg cells was observed in the spleen, lymph nodes, and colon of Carma1-mutant mice that expanded after viral infection, suggesting that peripheral development of Treg cells could still occur. Indeed, Carma1-mutant CD4+ T cells could be converted into the Treg lineage in vitro. Our results demonstrate an organ-specific requirement for the CARMA1 signaling pathway that developing thymocytes need in order to become Treg cells, but that naïve CD4+ T cells can bypass in the periphery. This dichotomy suggests that Treg cells of thymic or peripheral origin may have different specificities or functions in vivo. The organ-specific requirement for CARMA1-dependent signaling in the thymus suggests that regulatory T cells of thymic or peripheral origin may have different roles in vivo.
Collapse
Affiliation(s)
- Michael J Barnes
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
- * To whom correspondence should be addressed. E-mail: (MJB); (KH)
| | - Philippe Krebs
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Nathaniel Harris
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Celine Eidenschenk
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Rosana Gonzalez-Quintial
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Carrie N Arnold
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Karine Crozat
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Sosathya Sovath
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Eva Marie Moresco
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Argyrios N Theofilopoulos
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Bruce Beutler
- Department of Genetics, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kasper Hoebe
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, United States of America
- * To whom correspondence should be addressed. E-mail: (MJB); (KH)
| |
Collapse
|
545
|
Abstract
The immune system is pivotal in mediating the interactions between host and microbiota that shape the intestinal environment. Intestinal homeostasis arises from a highly dynamic balance between host protective immunity and regulatory mechanisms. This regulation is achieved by a number of cell populations acting through a set of shared regulatory pathways. In this review, we summarize the main lymphocyte subsets controlling immune responsiveness in the gut and their mechanisms of control, which involve maintenance of intestinal barrier function and suppression of chronic inflammation. CD4(+)Foxp3(+) T cells play a nonredundant role in the maintenance of intestinal homeostasis through IL-10- and TGF-beta-dependent mechanisms. Their activity is complemented by other T and B lymphocytes. Because breakdown in immune regulatory networks in the intestine leads to chronic inflammatory diseases of the gut, such as inflammatory bowel disease and celiac disease, regulatory lymphocytes are an attractive target for therapies of intestinal inflammation.
Collapse
Affiliation(s)
- Ana Izcue
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | | | | |
Collapse
|
546
|
Shiokawa A, Tanabe K, Tsuji NM, Sato R, Hachimura S. IL-10 and IL-27 producing dendritic cells capable of enhancing IL-10 production of T cells are induced in oral tolerance. Immunol Lett 2009; 125:7-14. [PMID: 19446579 DOI: 10.1016/j.imlet.2009.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 04/29/2009] [Accepted: 05/04/2009] [Indexed: 01/10/2023]
Abstract
Oral tolerance is a key feature of intestinal immunity, generating systemic tolerance to ingested antigens (Ag). Dendritic cells (DC) have been revealed as important immune regulators, however, the precise role of DC in oral tolerance induction remains unclear. We investigated the characteristics of DC in spleen, mesenteric lymph node (MLN), and Peyer's patch (PP) after oral Ag administration in a TCR-transgenic mouse model. DC from PP and MLN of tolerized mice induced IL-10 production but not Foxp3 expression in cocultured T cells. IL-10 production was markedly increased after 5-7-day Ag administration especially in PP DC. On the other hand, IL-27 production was increased after 2-5-day Ag administration. CD11b(+) DC, which increased after ingestion of Ag, prominently expressed IL-10 and IL-27 compared with CD11b(-) DC. These results suggest that IL-10 and IL-27 producing DC are increased by interaction with antigen specific T cells in PP, and these DC act as an inducer of IL-10 producing T cells in oral tolerance.
Collapse
Affiliation(s)
- Aya Shiokawa
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
547
|
Abstract
The small and large intestine contain the largest number of macrophages in the body and these cells are strategically located directly underneath the epithelial layer, enabling them to sample the lumen. Such intestinal macrophages have a different phenotype from other tissue macrophages in that they ingest and may kill microbes but they do not mediate strong pro-inflammatory responses upon microbial recognition. These properties are essential for maintaining a healthy intestine. It is generally accepted that tolerance to the intestinal flora is lost in inflammatory bowel diseases, and genes involved in microbial recognition, killing and macrophage activation have already been associated with these diseases. In this review, we shed light on the intestinal macrophage and how it influences intestinal immunity.
Collapse
|
548
|
von Hertzen LC, Savolainen J, Hannuksela M, Klaukka T, Lauerma A, Mäkelä MJ, Pekkanen J, Pietinalho A, Vaarala O, Valovirta E, Vartiainen E, Haahtela T. Scientific rationale for the Finnish Allergy Programme 2008-2018: emphasis on prevention and endorsing tolerance. Allergy 2009; 64:678-701. [PMID: 19383025 DOI: 10.1111/j.1398-9995.2009.02024.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In similarity to many other western countries, the burden of allergic diseases in Finland is high. Studies worldwide have shown that an environment rich in microbes in early life reduces the subsequent risk of developing allergic diseases. Along with urbanization, such exposure has dramatically reduced, both in terms of diversity and quantity. Continuous stimulation of the immune system by environmental saprophytes via the skin, respiratory tract and gut appears to be necessary for activation of the regulatory network including regulatory T-cells and dendritic cells. Substantial evidence now shows that the balance between allergy and tolerance is dependent on regulatory T-cells. Tolerance induced by allergen-specific regulatory T-cells appears to be the normal immunological response to allergens in non atopic healthy individuals. Healthy subjects have an intact functional allergen-specific regulatory T-cell response, which in allergic subjects is impaired. Evidence on this exists with respect to atopic dermatitis, contact dermatitis, allergic rhinitis and asthma. Restoration of impaired allergen-specific regulatory T-cell response and tolerance induction has furthermore been demonstrated during allergen-specific subcutaneous and sublingual immunotherapy and is crucial for good therapeutic outcome. However, tolerance can also be strengthened unspecifically by simple means, e.g. by consuming farm milk and spending time in nature. Results so far obtained from animal models indicate that it is possible to restore tolerance by administering the allergen in certain circumstances both locally and systemically. It has become increasingly clear that continuous exposure to microbial antigens as well as allergens in foodstuffs and the environment is decisive, and excessive antigen avoidance can be harmful and weaken or even prevent the development of regulatory mechanisms. Success in the Finnish Asthma Programme was an encouraging example of how it is possible to reduce both the costs and morbidity of asthma. The time, in the wake of the Asthma Programme, is now opportune for a national allergy programme, particularly as in the past few years, fundamentally more essential data on tolerance and its mechanisms have been published. In this review, the scientific rationale for the Finnish Allergy Programme 2008-2018 is outlined. The focus is on tolerance and how to endorse tolerance at the population level.
Collapse
Affiliation(s)
- L C von Hertzen
- Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
549
|
Abstract
The intestinal mucosa harbors a complexly organized immune system, which accomplishes important, partially contradictory surveillance and protective functions. It has to protect the host from potential external (microbial and nonmicrobial) aggression, whereas at the same time avoiding an inflammatory reaction toward harmless antigenic structures of microbial (commensal microflora) or alimentary origin. Professional sentinels (of the innate immune system) are in close and interactive contact with the intestinal microflora, which plays via this exchange an important role during the postnatal period in the maturation of the intestinal immune system. The innate immune system is a key regulator of local and systemic immune responses, including the adaptive immune system. Distinct signals induce tolerance to specific antigens presented by intestinal dendritic cells, whereas proinflammatory mediators easily switch these tolerogenic responses toward protective or destructive inflammatory reactions. Given the intense exchange between the intestinal microflora and the mucosal immune system, a permanent but actively controlled and downregulated physiological inflammation characterizes the intestinal mucosa. Therefore, it is easily conceivable that perturbation of the intestinal microflora or immune control mechanisms can initiate or contribute to pathological and potentially chronic inflammatory reactions, such as seen in inflammatory bowel diseases.
Collapse
|
550
|
Abstract
In the intestinal tract, only a single layer of epithelial cells separates innate and adaptive immune effector cells from a vast amount of antigens. Here, the immune system faces a considerable challenge in tolerating commensal flora and dietary antigens while preventing the dissemination of potential pathogens. Failure to tightly control immune reactions may result in detrimental inflammation. In this respect, 'conventional' regulatory CD4(+) T cells, including naturally occurring and adaptive CD4(+) CD25(+) Foxp3(+) T cells, Th3 and Tr1 cells, have recently been the focus of considerable attention. However, regulatory mechanisms in the intestinal mucosa are highly complex, including adaptations of nonhaematopoietic cells and innate immune cells as well as the presence of unconventional T cells with regulatory properties such as resident TCRgammadelta or TCRalphabeta CD8(+) intraepithelial lymphocytes. This review aims to summarize the currently available knowledge on conventional and unconventional regulatory T cell subsets (Tregs), with special emphasis on clinical data and the potential role or malfunctioning of Tregs in four major human gastrointestinal diseases, i.e. inflammatory bowel diseases, coeliac disease, food allergy and colorectal cancer. We conclude that the clinical data confirms some but not all of the findings derived from experimental animal models.
Collapse
Affiliation(s)
- L Saurer
- Institute of Pathology, University of Bern, Switzerland
| | | |
Collapse
|