201
|
|
202
|
|
203
|
|
204
|
Vicente-Suarez I, Larange A, Reardon C, Matho M, Feau S, Chodaczek G, Park Y, Obata Y, Gold R, Wang-Zhu Y, Lena C, Zajonc DM, Schoenberger S, Kronenberg M, Cheroutre H. Unique lamina propria stromal cells imprint the functional phenotype of mucosal dendritic cells. Mucosal Immunol 2015; 8:141-51. [PMID: 24938743 PMCID: PMC4268120 DOI: 10.1038/mi.2014.51] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 05/13/2014] [Indexed: 02/04/2023]
Abstract
Mucosal dendritic cells (DCs) in the intestine acquire the unique capacity to produce retinoic acid (RA), a vitamin A metabolite that induces gut tropism and regulates the functional differentiation of the T cells they prime. Here, we identified a stromal cell (SC) population in the intestinal lamina propria (LP), which is capable of inducing RA production in DCs in a RA- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent fashion. Unlike DCs, LP SCs constitutively expressed the enzymatic machinery to produce RA even in the absence of dietary vitamin A, but were not able to do so in germ-free mice implying regulation by microbiota. Interestingly, DCs promoted GM-CSF production by the SCs indicating a two-way cross-talk between both cell types. Furthermore, RA-producing LP SCs and intestinal DCs localized closely in vivo suggesting that the interactions between both cell types might have an important role in the functional education of migratory DCs and therefore in the regulation of immune responses toward oral and commensal antigens.
Collapse
Affiliation(s)
- Ildefonso Vicente-Suarez
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Alexandre Larange
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Colin Reardon
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Michael Matho
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Sonia Feau
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Grzegorz Chodaczek
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Yunji Park
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Korea
| | - Yuuki Obata
- Division of Mucosal Barriology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan,Laboratory for immune regulation, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Rebecca Gold
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Yiran Wang-Zhu
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Chris Lena
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Dirk M. Zajonc
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Stephen Schoenberger
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| | - Hilde Cheroutre
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla CA 92037, USA
| |
Collapse
|
205
|
|
206
|
Systemic Manifestations of Mucosal Diseases. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00090-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
207
|
Cerovic V, Houston SA, Westlund J, Utriainen L, Davison ES, Scott CL, Bain CC, Joeris T, Agace WW, Kroczek RA, Mowat AM, Yrlid U, Milling SWF. Lymph-borne CD8α+ dendritic cells are uniquely able to cross-prime CD8+ T cells with antigen acquired from intestinal epithelial cells. Mucosal Immunol 2015; 8:38-48. [PMID: 24850430 PMCID: PMC4156465 DOI: 10.1038/mi.2014.40] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/18/2014] [Indexed: 02/04/2023]
Abstract
Cross-presentation of cellular antigens is crucial for priming CD8(+) T cells, and generating immunity to intracellular pathogens--particularly viruses. It is unclear which intestinal phagocytes perform this function in vivo. To address this, we examined dendritic cells (DCs) from the intestinal lymph of IFABP-tOVA 232-4 mice, which express ovalbumin in small intestinal epithelial cells (IECs). Among lymph DCs (LDCs) only CD103(+) CD11b(-) CD8α(+) DCs cross-present IEC-derived ovalbumin to CD8(+) OT-I T cells. Similarly, in the mesenteric lymph nodes (MLNs), cross-presentation of IEC-ovalbumin was limited to the CD11c(+) MHCII(hi) CD8α(+) migratory DCs, but absent from all other subsets, including the resident CD8α(hi) DCs. Crucially, delivery of purified CD8α(+) LDCs, but not other LDC subsets, into the MLN subcapsular lymphatic sinus induced proliferation of ovalbumin-specific, gut-tropic CD8(+) T cells in vivo. Finally, in 232-4 mice treated with R848, CD8α(+) LDCs were uniquely able to cross-prime interferon γ-producing CD8(+) T cells and drive their migration to the intestine. Our results clearly demonstrate that migrating CD8α(+) intestinal DCs are indispensable for cross-presentation of cellular antigens and, in conditions of inflammation, for the initial differentiation of effector CD8(+) T cells. They may therefore represent an important target for the development of antiviral vaccinations.
Collapse
Affiliation(s)
- V Cerovic
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK,()
| | - S A Houston
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - J Westlund
- Department of Medical Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - L Utriainen
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - E S Davison
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - C L Scott
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - C C Bain
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - T Joeris
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - W W Agace
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden,Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - R A Kroczek
- Department of Molecular Immunology, Robert Koch-Institute, Berlin, Germany
| | - A M Mowat
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - U Yrlid
- Department of Medical Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - S WF Milling
- Centre for Immunobiology, Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
208
|
Induction of Immune Tolerance to Dietary Antigens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 850:93-118. [DOI: 10.1007/978-3-319-15774-0_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
209
|
|
210
|
Rescigno M. Microbial Sensing and Regulation of Mucosal Immune Responses by Intestinal Epithelial Cells. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00028-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
211
|
Kayama H, Takeda K. Regulation of intestinal inflammation through interaction of intestinal environmental factors and innate immune cells. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Hisako Kayama
- Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kiyoshi Takeda
- Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
212
|
|
213
|
Clahsen T, Pabst O, Tenbrock K, Schippers A, Wagner N. Localization of dendritic cells in the gut epithelium requires MAdCAM-1. Clin Immunol 2015; 156:74-84. [DOI: 10.1016/j.clim.2014.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/21/2014] [Accepted: 11/12/2014] [Indexed: 11/24/2022]
|
214
|
Feehley T, Nagler CR. Cellular and molecular pathways through which commensal bacteria modulate sensitization to dietary antigens. Curr Opin Immunol 2014; 31:79-86. [PMID: 25458998 PMCID: PMC4255329 DOI: 10.1016/j.coi.2014.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/25/2014] [Accepted: 10/03/2014] [Indexed: 12/16/2022]
Abstract
Food allergies are a growing public health concern. The rapidly increasing prevalence of allergic disease cannot be explained by genetic variation alone, suggesting a role for gene-by-environment interactions. The bacteria that colonize barrier surfaces, often referred to as the commensal microbiota, are dramatically affected by environmental factors and have a major impact on host health and homeostasis. Increasing evidence suggests that alterations in the composition of the microbiota, caused by factors such as antibiotic use and diet, are contributing to increased sensitization to dietary antigens. This review will discuss the cellular and molecular pathways activated by commensal bacteria to protect against allergic sensitization. By understanding the interplay between the environment, the microbiota, and the host, we may uncover novel therapeutic targets that will allow us to control the allergy epidemic.
Collapse
Affiliation(s)
- Taylor Feehley
- Committee on Immunology, Department of Pathology, The University of Chicago, 924 E. 57th St. JFK R120, Chicago, IL 60637, USA
| | - Cathryn R Nagler
- Committee on Immunology, Department of Pathology, The University of Chicago, 924 E. 57th St. JFK R120, Chicago, IL 60637, USA.
| |
Collapse
|
215
|
Mann ER, Bernardo D, Ng SC, Rigby RJ, Al-Hassi HO, Landy J, Peake STC, Spranger H, English NR, Thomas LV, Stagg AJ, Knight SC, Hart AL. Human gut dendritic cells drive aberrant gut-specific t-cell responses in ulcerative colitis, characterized by increased IL-4 production and loss of IL-22 and IFNγ. Inflamm Bowel Dis 2014; 20:2299-307. [PMID: 25397892 DOI: 10.1097/mib.0000000000000223] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
: The pathogenesis of inflammatory bowel disease is incompletely understood but results from a dysregulated intestinal immune response to the luminal microbiota. CD4 T cells mediate tissue injury in the inflammatory bowel disease-associated immune response. Dendritic cells (DC) generate primary T-cell responses and mediate intestinal immune tolerance to prevent overt inflammation in response to the gut microbiota. However, most information regarding function of intestinal DC has come from mouse models, and information in humans is scarce. We show here that intestinal DC subsets are skewed in ulcerative colitis (UC) in humans, with a loss of CD103 lymph-node homing DC; this intestinal DC subset preferentially generates regulatory T cells in mice. We show infiltrates of DC negative for myeloid marker CD11c, with enhanced expression of Toll-like receptors for bacterial recognition. After mixed leukocyte reaction, DC from the inflamed UC colon had an enhanced ability to generate gut-specific CD4 T cells with enhanced production of interleukin-4 but a loss of interferon γ and interleukin-22 production. Conditioning intestinal DC with probiotic strain Lactobacillus casei Shirota in UC partially restored their normal function indicated by reduced Toll-like receptor 2/4 expression and restoration of their ability to imprint homing molecules on T cells and to generate interleukin-22 production by stimulated T cells. This study suggests that T-cell dysfunction in UC is driven by DC. T-cell responses can be manipulated indirectly through effects of bacterial conditioning on gut DC with implications for immunomodulatory effects of the commensal microbiota in vivo. Manipulation of DC to allow generation of DC-specific therapy may be beneficial in inflammatory bowel disease.
Collapse
Affiliation(s)
- Elizabeth R Mann
- *Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, United Kingdom; †Gastrointestinal Division, Johns Hopkins University School of Medicine, Baltimore, Maryland; ‡Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, State Key Laboratory of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China; §Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom; ‖Department of Gastroenterology, St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, United Kingdom; ¶Yakult UK Ltd., South Ruislip, United Kingdom; and **Centre for Immunology and Infectious Disease, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Grainger JR, Askenase MH, Guimont-Desrochers F, da Fonseca DM, Belkaid Y. Contextual functions of antigen-presenting cells in the gastrointestinal tract. Immunol Rev 2014; 259:75-87. [PMID: 24712460 DOI: 10.1111/imr.12167] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The immune system of the gastrointestinal tract must be tightly regulated to limit pathologic responses toward innocuous antigens while simultaneously allowing for rapid development of effector responses against invading pathogens. Highly specialized antigen-presenting cell (APC) subsets present in the gut play a dominant role in balancing these seemingly disparate functions. In this review, we discuss new findings associated with the function of gut APCs and particularly the contextual role of these cells in both establishing tolerance to orally acquired antigens in the steady state and regulating acute inflammation during infection.
Collapse
Affiliation(s)
- John R Grainger
- Program in Barrier Immunity and Repair, Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
217
|
Goverse G, Olivier BJ, Molenaar R, Knippenberg M, Greuter M, Konijn T, Cook ECL, Beijer MR, Fedor DM, den Haan JMM, Napoli JL, Bouma G, Mebius RE. Vitamin A metabolism and mucosal immune function are distinct between BALB/c and C57BL/6 mice. Eur J Immunol 2014; 45:89-100. [DOI: 10.1002/eji.201343340] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 09/23/2014] [Accepted: 10/08/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Gera Goverse
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Brenda J. Olivier
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Rosalie Molenaar
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Marlene Knippenberg
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Mascha Greuter
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Tanja Konijn
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Emma C. L. Cook
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Marieke R. Beijer
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Dawn M. Fedor
- Department of Gastroenterology; VU University Medical Center; Amsterdam The Netherlands
| | - Joke M. M. den Haan
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| | - Joseph L. Napoli
- Department of Nutritional Science and Toxicology; University of California; Berkeley CA USA
| | - Gerd Bouma
- Department of Gastroenterology; VU University Medical Center; Amsterdam The Netherlands
| | - Reina E. Mebius
- Department of Molecular Cell Biology and Immunology; VU medical center; Amsterdam The Netherlands
| |
Collapse
|
218
|
Chewning JH, Weaver CT. Development and survival of Th17 cells within the intestines: the influence of microbiome- and diet-derived signals. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:4769-77. [PMID: 25381358 PMCID: PMC6007010 DOI: 10.4049/jimmunol.1401835] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Th17 cells have emerged as important mediators of host defense and homeostasis at barrier sites, particularly the intestines, where the greatest number and diversity of the microbiota reside. A critical balance exists between protection of the host from its own microbiota and pathogens and the development of immune-mediated disease. Breaches of local innate immune defenses provide critical stimuli for the induction of Th17 cell development, and additional cues within these tissues promote Th17 cell survival and/or plasticity. Normally, this results in eradication of the microbial threat and restitution of homeostasis. When dysregulated, however, Th17 cells can cause a range of immune-mediated diseases, whether directed against Ags derived from the microbiota, such as in inflammatory bowel disease, or against self-Ags in a range of autoimmune diseases. This review highlights recent discoveries that provide new insights into ways in which environmental signals impact Th17 cell development and function in the intestines.
Collapse
Affiliation(s)
- Joseph H Chewning
- Department of Pediatrics, Pediatric Blood and Marrow Transplantation Program, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
219
|
Hadley GA, Higgins JMG. Integrin αEβ7: molecular features and functional significance in the immune system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:97-110. [PMID: 25023170 DOI: 10.1007/978-94-017-9153-3_7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Alpha E beta 7 (αEβ7) is an α-I domain-containing integrin that is highly expressed by a variety of leukocyte populations at mucosal sites including intraepithelial T cells, dendritic cells, mast cells, and T regulatory cells (Treg). Expression depends largely or solely on transforming growth factor beta (TGF-β) isoforms. The best characterized ligand for αEβ7 is E-cadherin on epithelial cells, though there is evidence of a second ligand in the human system. An exposed acidic residue on the distal aspect of E-cadherin domain 1 interacts with the MIDAS site in the αE α-I domain. By binding to E-cadherin, αEβ7 contributes to mucosal specific retention of leukocytes within epithelia. Studies on αE knockout mice have identified an additional important function for this integrin in allograft rejection and have also indicated that it may have a role in immunoregulation. Recent studies point to a multifaceted role for αEβ7 in regulating both innate and acquired immune responses to foreign antigen.
Collapse
Affiliation(s)
- Gregg A Hadley
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA,
| | | |
Collapse
|
220
|
Bobat S, Cunningham AF. Bacterial infections and vaccines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 828:75-98. [PMID: 25253028 DOI: 10.1007/978-1-4939-1489-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- Saeeda Bobat
- The Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands, UK,
| | | |
Collapse
|
221
|
Wilson HL, Obradovic MR. Evidence for a common mucosal immune system in the pig. Mol Immunol 2014; 66:22-34. [PMID: 25242212 PMCID: PMC7132386 DOI: 10.1016/j.molimm.2014.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/03/2022]
Abstract
There is evidence that the common mucosal immune system exists in pigs. Vaccination at an easily accessible mucosal site may assist in providing protection at other mucosal sites. Local and distal mucosal sites should be sampled after vaccinations to define the optimal dose and formulation which promotes the common mucosal immune system in pigs.
The majority of lymphocytes activated at mucosal sites receive instructions to home back to the local mucosa, but a portion also seed distal mucosa sites. By seeding distal sites with antigen-specific effector or memory lymphocytes, the foundation is laid for the animal's mucosal immune system to respond with a secondary response should to this antigen be encountered at this site in the future. The common mucosal immune system has been studied quite extensively in rodent models but less so in large animal models such as the pig. Reasons for this paucity of reported induction of the common mucosal immune system in this species may be that distal mucosal sites were examined but no induction was observed and therefore it was not reported. However, we suspect that the majority of investigators simply did not sample distal mucosal sites and therefore there is little evidence of immune response induction in the literature. It is our hope that more pig immunologists and infectious disease experts who perform mucosal immunizations or inoculations on pigs will sample distal mucosal sites and report their findings, whether results are positive or negative. In this review, we highlight papers that show that immunization/inoculation using one route triggers mucosal immune system induction locally, systemically, and within at least one distal mucosal site. Only by understanding whether immunizations at one site triggers immunity throughout the common mucosal immune system can we rationally develop vaccines for the pig, and through these works we can gather evidence about the mucosal immune system that may be extrapolated to other livestock species or humans.
Collapse
Affiliation(s)
- Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO), Home of the International Vaccine Centre (InterVac), 120 Veterinary Road, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.
| | - Milan R Obradovic
- Vaccine and Infectious Disease Organization (VIDO), Home of the International Vaccine Centre (InterVac), 120 Veterinary Road, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.
| |
Collapse
|
222
|
Dave M, Papadakis KA, Faubion WA. Immunology of inflammatory bowel disease and molecular targets for biologics. Gastroenterol Clin North Am 2014; 43:405-24. [PMID: 25110250 PMCID: PMC4480636 DOI: 10.1016/j.gtc.2014.05.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated disease and involves a complex interplay of host genetics and environmental influences. Recent advances in the field, including data from genome-wide association studies and microbiome analysis, have started to unravel the complex interaction between host genetics and environmental influences in the pathogenesis of IBD. A drawback of current clinical trials is inadequate or lack of immune phenotyping of patients. However, recent advances in high-throughput technologies provide an opportunity to monitor the dynamic and complex immune system, which may to lead to a more personalized treatment approach in IBD.
Collapse
|
223
|
Figueiredo MM, Deoti B, Amorim IF, Pinto AJW, Moraes A, Carvalho CS, da Silva SM, de Assis ACB, de Faria AMC, Tafuri WL. Expression of regulatory T cells in jejunum, colon, and cervical and mesenteric lymph nodes of dogs naturally infected with Leishmania infantum. Infect Immun 2014; 82:3704-12. [PMID: 24935975 PMCID: PMC4187817 DOI: 10.1128/iai.01862-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Using flow cytometry, we evaluated the frequencies of CD4(+) and CD8(+) T cells and Foxp3(+) regulatory T cells (Tregs) in mononuclear cells in the jejunum, colon, and cervical and mesenteric lymph nodes of dogs naturally infected with Leishmania infantum and in uninfected controls. All infected dogs showed chronic lymphadenitis and enteritis. Despite persistent parasite loads, no erosion or ulcers were evident in the epithelial mucosa. The colon harbored more parasites than the jejunum. Frequencies of total CD4(+), total Foxp3, and CD4(+) Foxp3(+) cells were higher in the jejunum than in the colon. Despite negative enzyme-linked immunosorbent assay (ELISA) serum results for cytokines, levels of interleukin-10 (IL-10), gamma interferon (IFN-γ), transforming growth factor beta (TGF-β), and tumor necrosis factor alpha (TNF-α) were higher in the jejunum than in the colon for infected dogs. However, IL-4 levels were higher in the colon than in the jejunum for infected dogs. There was no observed correlation between clinical signs and histopathological changes or immunological and parasitological findings in the gastrointestinal tract (GIT) of canines with visceral leishmaniasis. However, distinct segments of the GIT presented different immunological and parasitological responses. The jejunum showed a lower parasite load, with increased frequencies and expression of CD4, Foxp3, and CD8 receptors and IL-10, TGF-β, IFN-γ, and TNF-α cytokines. The colon showed a higher parasite load, with increasing expression of IL-4. Leishmania infantum infection increased expression of CD4, Foxp3, IL-10, TGF-β, IFN-γ, and TNF-α and reduced CD8 and IL-4 expression in both the jejunum and the colon.
Collapse
Affiliation(s)
- Maria M Figueiredo
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Beatriz Deoti
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Izabela F Amorim
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Aldair J W Pinto
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Andrea Moraes
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Carolina S Carvalho
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Sydnei Magno da Silva
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Ana C B de Assis
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Ana M C de Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Wagner L Tafuri
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| |
Collapse
|
224
|
Mann ER, Li X. Intestinal antigen-presenting cells in mucosal immune homeostasis: Crosstalk between dendritic cells, macrophages and B-cells. World J Gastroenterol 2014; 20:9653-9664. [PMID: 25110405 PMCID: PMC4123356 DOI: 10.3748/wjg.v20.i29.9653] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 02/26/2014] [Accepted: 04/29/2014] [Indexed: 02/06/2023] Open
Abstract
The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance of the commensal microbiota. Inflammatory bowel disease (IBD) involves a breakdown in tolerance towards the microbiota. Dendritic cells (DC), macrophages (MΦ) and B-cells are known as professional antigen-presenting cells (APC) due to their specialization in presenting processed antigen to T-cells, and in turn shaping types of T-cell responses generated. Intestinal DC are migratory cells, unique in their ability to generate primary T-cell responses in mesenteric lymph nodes or Peyer’s patches, whilst MΦ and B-cells contribute to polarization and differentiation of secondary T-cell responses in the gut lamina propria. The antigen-sampling function of gut DC and MΦ enables them to sample bacterial antigens from the gut lumen to determine types of T-cell responses generated. The primary function of intestinal B-cells involves their secretion of large amounts of immunoglobulin A, which in turn contributes to epithelial barrier function and limits immune responses towards to microbiota. Here, we review the role of all three types of APC in intestinal immunity, both in the steady state and in inflammation, and how these cells interact with one another, as well as with the intestinal microenvironment, to shape mucosal immune responses. We describe mechanisms of maintaining intestinal immune tolerance in the steady state but also inappropriate responses of APC to components of the gut microbiota that contribute to pathology in IBD.
Collapse
|
225
|
Becker M, Güttler S, Bachem A, Hartung E, Mora A, Jäkel A, Hutloff A, Henn V, Mages HW, Gurka S, Kroczek RA. Ontogenic, Phenotypic, and Functional Characterization of XCR1(+) Dendritic Cells Leads to a Consistent Classification of Intestinal Dendritic Cells Based on the Expression of XCR1 and SIRPα. Front Immunol 2014; 5:326. [PMID: 25120540 PMCID: PMC4112810 DOI: 10.3389/fimmu.2014.00326] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/27/2014] [Indexed: 12/20/2022] Open
Abstract
In the past, lack of lineage markers confounded the classification of dendritic cells (DC) in the intestine and impeded a full understanding of their location and function. We have recently shown that the chemokine receptor XCR1 is a lineage marker for cross-presenting DC in the spleen. Now, we provide evidence that intestinal XCR1+ DC largely, but not fully, overlap with CD103+ CD11b− DC, the hypothesized correlate of “cross-presenting DC” in the intestine, and are selectively dependent in their development on the transcription factor Batf3. XCR1+ DC are located in the villi of the lamina propria of the small intestine, the T cell zones of Peyer’s patches, and in the T cell zones and sinuses of the draining mesenteric lymph node. Functionally, we could demonstrate for the first time that XCR1+/CD103+ CD11b− DC excel in the cross-presentation of orally applied antigen. Together, our data show that XCR1 is a lineage marker for cross-presenting DC also in the intestinal immune system. Further, extensive phenotypic analyses reveal that expression of the integrin SIRPα consistently demarcates the XCR1− DC population. We propose a simplified and consistent classification system for intestinal DC based on the expression of XCR1 and SIRPα.
Collapse
Affiliation(s)
- Martina Becker
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Steffen Güttler
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Annabell Bachem
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Evelyn Hartung
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Ahmed Mora
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Anika Jäkel
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | - Andreas Hutloff
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany ; German Rheumatism Research Centre , Berlin , Germany
| | - Volker Henn
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | | | - Stephanie Gurka
- Molecular Immunology, Robert Koch-Institute , Berlin , Germany
| | | |
Collapse
|
226
|
Spasova DS, Surh CD. Blowing on embers: commensal microbiota and our immune system. Front Immunol 2014; 5:318. [PMID: 25120539 PMCID: PMC4112811 DOI: 10.3389/fimmu.2014.00318] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/24/2014] [Indexed: 12/19/2022] Open
Abstract
Vertebrates have co-evolved with microorganisms resulting in a symbiotic relationship, which plays an important role in health and disease. Skin and mucosal surfaces are colonized with a diverse population of commensal microbiota, over 1000 species, outnumbering the host cells by 10-fold. In the past 40 years, studies have built on the idea that commensal microbiota is in constant contact with the host immune system and thus influence immune function. Recent studies, focusing on mutualism in the gut, have shown that commensal microbiota seems to play a critical role in the development and homeostasis of the host immune system. In particular, the gut microbiota appears to direct the organization and maturation of lymphoid tissues and acts both locally and systemically to regulate the recruitment, differentiation, and function of innate and adaptive immune cells. While the pace of research in the area of the mucosal–immune interface has certainly intensified over the last 10 years, we are still in the early days of this field. Illuminating the mechanisms of how gut microbes shape host immunity will enhance our understanding of the causes of immune-mediated pathologies and improve the design of next-generation vaccines. This review discusses the recent advances in this field, focusing on the close relationship between the adaptive immune system and commensal microbiota, a constant and abundant source of foreign antigens.
Collapse
Affiliation(s)
- Darina S Spasova
- Kellog School of Science and Technology Doctoral Program in Chemical and Biological Sciences and the Department of Immunology and Microbial Science, The Scripps Research Institute , La Jolla, CA , USA ; Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| | - Charles D Surh
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA ; Academy of Immunology and Microbiology, Institute of Basic Science , Pohang , South Korea ; Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology , Pohang , South Korea
| |
Collapse
|
227
|
Envelope glycoprotein binding to the integrin α4β7 is not a general property of most HIV-1 strains. J Virol 2014; 88:10767-77. [PMID: 25008916 DOI: 10.1128/jvi.03296-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The HIV-1 surface glycoprotein gp120 has been reported to bind and signal through α4β7 by means of a tripeptide motif in the V2 loop that mimics structures present in the natural ligands for α4β7, suggesting that α4β7 may facilitate HIV-1 infection of CD4(+) T cells in the gut. Furthermore, immune correlates in the RV144 vaccine efficacy trial generated the hypothesis that V1V2 antibodies to an epitope near the putative α4β7 binding motif may play a role in protection against HIV-1 infection. In the interest of developing an assay to detect antibodies that block gp120 binding to α4β7, we used retinoic acid (RA)-activated human peripheral blood mononuclear cells (PBMCs) and transfected HEK293T (293T) cells expressing the integrin complex to study the α4β7 binding properties of 16 HIV-1 envelope glycoproteins. The natural ligand for α4β7, mucosal addressin cell adhesion molecule-1 (MAdCAM-1), bound efficiently to RA-activated PBMCs and transfected 293T cells, and this binding was blocked by antibodies to α4. gp120 from multiple HIV-1 subtypes bound to RA-activated PBMCs from three donors in a CD4-dependent manner, but little or no α4β7 binding was detected. Similarly, little or no binding to α4β7 on transfected 293T cells was detected with multiple gp120s and gp140s, including gp120s from transmitted/founder strains, or when gp120 was produced in CHO, 293T, and 293S/GnT1(-/-) cells. Finally, we found no evidence that infectious HIV-1 virions produced in either PBMCs or 293T cells could bind α4β7 on transfected 293T cells. Infectious HIV-1 virions and most gp120s/gp140s appear to be poor ligands for the α4β7 integrin complex under the conditions tested here. IMPORTANCE Certain HIV-1 gp120 envelope glycoproteins have been shown to bind the gut-homing receptor α4β7, and it has been suggested that this binding facilitates mucosal transmission and virus replication in the gut mucosa. Additional evidence has generated the hypothesis that antibodies that bind near the putative α4β7 binding motif in the V2 loop of gp120, possibly disrupting gp120-α4β7 binding, may be important for HIV-1 vaccines. Our evidence indicates that infectious HIV-1 virions and many gp120s lack detectable α4β7 binding activity, suggesting that this homing receptor may play a limited role in direct HIV-1 infection of cells.
Collapse
|
228
|
Abstract
Classical dendritic cells (cDCs) form a critical interface between innate and adaptive immunity. As myeloid immune cell sentinels, cDCs are specialized in the sensing of pathogen challenges and cancer. They translate the latter for T cells into peptide form. Moreover, cDCs provide additional critical information on the original antigen context to trigger a diverse spectrum of appropriate protective responses. Here we review recent progress in our understanding of cDC subsets in mice. We will discuss cDC subset ontogeny and transcription factor dependencies, as well as emerging functional specializations within the cDC compartment in lymphoid and nonlymphoid tissues.
Collapse
Affiliation(s)
- Alexander Mildner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
229
|
Magnusson MK, Wick MJ. Intestinal dendritic cell and macrophage subsets: Tipping the balance to Crohn's disease? Eur J Microbiol Immunol (Bp) 2014; 1:19-24. [PMID: 24466433 DOI: 10.1556/eujmi.1.2011.1.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells and macrophages play an essential role in immune homeostasis in the intestine. They have the critical task of maintaining the balance between tolerance to the intestinal microflora and potential food antigens while retaining the ability to initiate immunity against pathogens. For patients with Crohn's Disease, the tolerance/immunity balance is disturbed and these individuals suffer from chronic intestinal inflammation driven by aberrant T cell reactivity to intestinal bacteria. As antigen presenting cells are required for T cell activation, intestinal phagocytes with the capacity to present antigens from intestinal bacteria to T cells are likely involved in initiating and propagating Crohn's Disease. Recent data describe unique subsets of human intestinal phagocytes that may be involved in the aberrant reactivity to commensal flora that drives Crohn's Disease pathogenesis. This review summarizes the current knowledge of phagocyte subsets in the intestine and mesenteric lymph nodes in healthy individuals and Crohn's Disease patients. Deciphering the function of intestinal phagocytes in health and disease is crucial to advance our understanding of the cellular mechanisms underlying this debilitating disease, provides a potential way to improve treatment for patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- M K Magnusson
- Department of Microbiology and Immunology, University of Gothenburg Gothenburg Sweden
| | - M J Wick
- Department of Microbiology and Immunology, University of Gothenburg Gothenburg Sweden
| |
Collapse
|
230
|
Geem D, Medina-Contreras O, McBride M, Newberry RD, Koni PA, Denning TL. Specific microbiota-induced intestinal Th17 differentiation requires MHC class II but not GALT and mesenteric lymph nodes. THE JOURNAL OF IMMUNOLOGY 2014; 193:431-8. [PMID: 24899505 DOI: 10.4049/jimmunol.1303167] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IL-17-expressing CD4+ T lymphocytes (Th17 cells) naturally reside in the intestine where specific cytokines and microbiota, such as segmented filamentous bacteria (SFB), promote their differentiation. Intestinal Th17 cells are thought to initially differentiate in the GALT and/or mesenteric lymph nodes upon Ag encounter and subsequently home to the lamina propria (LP) where they mediate effector functions. However, whether GALT and/or mesenteric lymph nodes are required for intestinal Th17 differentiation as well as how microbiota containing SFB regulate Ag-specific intestinal Th17 cells remain poorly defined. In this study, we observed that naive CD4+ T cells were abundant in the intestinal LP prior to weaning and that the accumulation of Th17 cells in response to microbiota containing SFB occurred in the absence of lymphotoxin-dependent lymphoid structures and the spleen. Furthermore, the differentiation of intestinal Th17 cells in the presence of microbiota containing SFB was dependent on MHC class II expression by CD11c+ cells. Lastly, the differentiation of Ag-specific Th17 cells required both the presence of cognate Ag and microbiota containing SFB. These findings suggest that microbiota containing SFB create an intestinal milieu that may induce Ag-specific Th17 differentiation against food and/or bacterial Ags directly in the intestinal LP.
Collapse
Affiliation(s)
- Duke Geem
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; Department of Pediatrics, Emory University, Atlanta, GA 30322; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322
| | - Oscar Medina-Contreras
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; Department of Pediatrics, Emory University, Atlanta, GA 30322; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322
| | - Michelle McBride
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Pandelakis A Koni
- Cancer Immunology, Inflammation, and Tolerance Program, Georgia Regents University, Augusta, GA 30912
| | - Timothy L Denning
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; Department of Pediatrics, Emory University, Atlanta, GA 30322; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322;
| |
Collapse
|
231
|
Nerreter T, Köchel C, Jesper D, Eichelbrönner I, Putz E, Einsele H, Seggewiss-Bernhardt R. Dasatinib enhances migration of monocyte-derived dendritic cells by reducing phosphorylation of inhibitory immune receptors Siglec-9 and Siglec-3. Exp Hematol 2014; 42:773-82.e1-3. [PMID: 24882272 DOI: 10.1016/j.exphem.2014.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 04/18/2014] [Accepted: 05/19/2014] [Indexed: 11/19/2022]
Abstract
The SRC family of kinases (SFKs) is crucial to malignant growth, but also important for signaling in immune cells such as dendritic cells (DCs). These specialized antigen-presenting cells are essential for inducing and boosting specific T-cell responses against pathogens and malignancies. Targeted therapy with SFK inhibitors holds great promise as a direct anti-cancer treatment, but potentially also as an indirect treatment via immunomodulation. Here, we investigated whether the BCR-ABL/SRC inhibitor dasatinib would modulate the major effector functions of DCs, especially their migration, a prerequisite to interaction with lymphocytes in secondary lymphoid organs. We report for the first time that dasatinib more than doubled the number of mature human monocyte-derived DCs (moDCs) migrating toward a CCL19 gradient despite unchanged CCR7 expression when used for pretreatment. These effects were caused by dephosphorylation of SFKs, as confirmed by the specific SFK inhibitor SRC inhibitor 1, leading to dephosphorylation of the inhibitory immunoreceptors Siglec-9 and Siglec-3. The specific blocking of the latter also enhanced migration and underlined the importance of these SFK-dependent receptor systems for migration of moDCs. Dasatinib hampered the secretion of interleukin-12 by moDCs at clinically relevant concentrations. In contrast, endocytosis or boosting of cytomegalovirus-specific CD8(+) T-cell responses remained unaltered when applying dasatinib-pretreated moDCs, in line with minor effects on the expression of co-stimulatory molecules essential for DC-T cell interaction. The induction of enhanced migration of moDCs may potentially be useful in chemo-immunotherapeutic applications. Thus, the use of dasatinib or blocking Siglec antibodies as adjuvants in this setting to induce stronger immune responses is worthy of further study.
Collapse
Affiliation(s)
- Thomas Nerreter
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken (CCC MF), University Hospital of Würzburg, Würzburg, Germany
| | - Christoph Köchel
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Daniel Jesper
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Irina Eichelbrönner
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Evelyn Putz
- Department of Transfusion Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Ruth Seggewiss-Bernhardt
- Comprehensive Cancer Center Mainfranken (CCC MF), University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
232
|
CX3CR1⁺ cells facilitate the activation of CD4 T cells in the colonic lamina propria during antigen-driven colitis. Mucosal Immunol 2014; 7:533-48. [PMID: 24129164 DOI: 10.1038/mi.2013.70] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 08/14/2013] [Indexed: 02/04/2023]
Abstract
Dendritic cells (DCs) and macrophages populate the intestinal lamina propria to initiate immune responses required for the maintenance of intestinal homeostasis. To investigate whether CX3CR1(+) phagocytes communicate with CD4 T cells during the development of transfer colitis, we established an antigen-driven colitis model induced by the adoptive transfer of DsRed OT-II cells in CX3CR1(GFP/+) × RAG(-/-) recipients challenged with Escherichia coli expressing ovalbumin (OVA) fused to a cyan fluorescent protein (CFP). After colonization of CX3CR1(GFP/+) × RAG(-/-) animals with red fluorescent E. coli pCherry-OVA, colonic CX3CR1(+) cells but not CD103(+) DCs phagocytosed E. coli pCherry-OVA. Degraded bacterial-derived antigens are transported by CD103(+) DCs to mesenteric lymph nodes (MLNs), where CD103(+) DCs prime naive T cells. In RAG(-/-) recipients reconstituted with OT II cells and gavaged with OVA-expressing E. coli, colonic CX3CR1(+) phagocytes are in close contact with CD4 T cells and presented bacterial-derived antigens to CD4 T cells to activate and expand effector T cells.
Collapse
|
233
|
Cerovic V, Bain CC, Mowat AM, Milling SWF. Intestinal macrophages and dendritic cells: what's the difference? Trends Immunol 2014; 35:270-7. [PMID: 24794393 DOI: 10.1016/j.it.2014.04.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 03/31/2014] [Accepted: 04/03/2014] [Indexed: 02/07/2023]
Abstract
Mononuclear phagocytes (MPs) in the murine intestine, comprising dendritic cells (DCs) and macrophages (Mϕs), perform disparate yet complementary immunological functions. Functional analyses of these distinct MP subsets have been complicated by the substantial overlap in their surface phenotypes. Here, we review recent findings that have enabled more accurate definition of these MP subsets. We discuss these recent advances in the context of the current understanding of the functions of DCs and Mϕs in the maintenance of intestinal homeostasis, and how their functions may alter when homeostasis is disrupted.
Collapse
Affiliation(s)
- Vuk Cerovic
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Calum C Bain
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Allan M Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Simon W F Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
234
|
Depletion of host CCR7(+) dendritic cells prevented donor T cell tissue tropism in anti-CD3-conditioned recipients. Biol Blood Marrow Transplant 2014; 20:920-8. [PMID: 24691220 DOI: 10.1016/j.bbmt.2014.03.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/25/2014] [Indexed: 11/23/2022]
Abstract
We reported previously that anti-CD3 mAb treatment before hematopoietic cell transplantation (HCT) prevented graft-versus-host disease (GVHD) and preserved graft-versus-leukemia (GVL) effects in mice. These effects were associated with downregulated donor T cell expression of tissue-specific homing and chemokine receptors, marked reduction of donor T cell migration into GVHD target tissues, and deletion of CD103(+) dendritic cells (DCs) in mesenteric lymph nodes (MLN). MLN CD103(+) DCs and peripheral lymph node (PLN) DCs include CCR7(+) and CCR7(-) subsets, but the role of these DC subsets in regulating donor T cell expression of homing and chemokine receptors remain unclear. Here, we show that recipient CCR7(+), but not CCR7(-), DCs in MLN induced donor T cell expression of gut-specific homing and chemokine receptors in a retinoid acid-dependent manner. CCR7 regulated activated DC migration from tissue to draining lymph node, but it was not required for the ability of DCs to induce donor T cell expression of tissue-specific homing and chemokine receptors. Finally, anti-CD3 treatment depleted CCR7(+) but not CCR7(-) DCs by inducing sequential expansion and apoptosis of CCR7(+) DCs in MLN and PLN. Apoptosis of CCR7(+) DCs was associated with DC upregulation of Fas expression and natural killer cell but not T, B, or dendritic cell upregulation of FasL expression in the lymph nodes. These results suggest that depletion of CCR7(+) host-type DCs, with subsequent inhibition of donor T cell migration into GVHD target tissues, can be an effective approach in prevention of acute GVHD and preservation of GVL effects.
Collapse
|
235
|
Affiliation(s)
- Lesley E. Smythies
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John R. Smythies
- Center for Brain and Cognition, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
236
|
Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol 2014; 14:141-53. [PMID: 24566914 DOI: 10.1038/nri3608] [Citation(s) in RCA: 2104] [Impact Index Per Article: 191.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The abundance of innate and adaptive immune cells that reside together with trillions of beneficial commensal microorganisms in the mammalian gastrointestinal tract requires barrier and regulatory mechanisms that conserve host-microbial interactions and tissue homeostasis. This homeostasis depends on the diverse functions of intestinal epithelial cells (IECs), which include the physical segregation of commensal bacteria and the integration of microbial signals. Hence, IECs are crucial mediators of intestinal homeostasis that enable the establishment of an immunological environment permissive to colonization by commensal bacteria. In this Review, we provide a comprehensive overview of how IECs maintain host-commensal microbial relationships and immune cell homeostasis in the intestine.
Collapse
Affiliation(s)
- Lance W Peterson
- Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - David Artis
- 1] Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania. [2] Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
237
|
Osteopontin expression by CD103- dendritic cells drives intestinal inflammation. Proc Natl Acad Sci U S A 2014; 111:E856-65. [PMID: 24550510 DOI: 10.1073/pnas.1316447111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intestinal CD103(-) dendritic cells (DCs) are pathogenic for colitis. Unveiling molecular mechanisms that render these cells proinflammatory is important for the design of specific immunotherapies. In this report, we demonstrated that mesenteric lymph node CD103(-) DCs express, among other proinflammatory cytokines, high levels of osteopontin (Opn) during experimental colitis. Opn expression by CD103(-) DCs was crucial for their immune profile and pathogenicity, including induction of T helper (Th) 1 and Th17 cell responses. Adoptive transfer of Opn-deficient CD103(-) DCs resulted in attenuated colitis in comparison to transfer of WT CD103(-) DCs, whereas transgenic CD103(-) DCs that overexpress Opn were highly pathogenic in vivo. Neutralization of secreted Opn expressed exclusively by CD103(-) DCs restrained disease severity. Also, Opn deficiency resulted in milder disease, whereas systemic neutralization of secreted Opn was therapeutic. We determined a specific domain of the Opn protein responsible for its CD103(-) DC-mediated proinflammatory effect. We demonstrated that disrupting the interaction of this Opn domain with integrin α9, overexpressed on colitic CD103(-) DCs, suppressed the inflammatory potential of these cells in vitro and in vivo. These results add unique insight into the biology of CD103(-) DCs and their function during inflammatory bowel disease.
Collapse
|
238
|
de Kivit S, Tobin MC, Forsyth CB, Keshavarzian A, Landay AL. Regulation of Intestinal Immune Responses through TLR Activation: Implications for Pro- and Prebiotics. Front Immunol 2014; 5:60. [PMID: 24600450 PMCID: PMC3927311 DOI: 10.3389/fimmu.2014.00060] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/03/2014] [Indexed: 12/12/2022] Open
Abstract
The intestinal mucosa is constantly facing a high load of antigens including bacterial antigens derived from the microbiota and food. Despite this, the immune cells present in the gastrointestinal tract do not initiate a pro-inflammatory immune response. Toll-like receptors (TLRs) are pattern recognition receptors expressed by various cells in the gastrointestinal tract, including intestinal epithelial cells (IEC) and resident immune cells in the lamina propria. Many diseases, including chronic intestinal inflammation (e.g., inflammatory bowel disease), irritable bowel syndrome (IBS), allergic gastroenteritis (e.g., eosinophilic gastroenteritis and allergic IBS), and infections are nowadays associated with a deregulated microbiota. The microbiota may directly interact with TLR. In addition, differences in intestinal TLR expression in health and disease may suggest that TLRs play an essential role in disease pathogenesis and may be novel targets for therapy. TLR signaling in the gut is involved in either maintaining intestinal homeostasis or the induction of an inflammatory response. This mini review provides an overview of the current knowledge regarding the contribution of intestinal epithelial TLR signaling in both tolerance induction or promoting intestinal inflammation, with a focus on food allergy. We will also highlight a potential role of the microbiota in regulating gut immune responses, especially through TLR activation.
Collapse
Affiliation(s)
- Sander de Kivit
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA
| | - Mary C Tobin
- Department of Immunology/Microbiology, Rush University Medical Center , Chicago, IL , USA
| | - Christopher B Forsyth
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA ; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Utrecht , Netherlands
| | - Alan L Landay
- Department of Immunology/Microbiology, Rush University Medical Center , Chicago, IL , USA ; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Utrecht , Netherlands
| |
Collapse
|
239
|
Caricilli AM, Castoldi A, Câmara NOS. Intestinal barrier: A gentlemen’s agreement between microbiota and immunity. World J Gastrointest Pathophysiol 2014; 5:18-32. [PMID: 24891972 PMCID: PMC4024517 DOI: 10.4291/wjgp.v5.i1.18] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/26/2013] [Accepted: 01/14/2014] [Indexed: 02/07/2023] Open
Abstract
Our body is colonized by more than a hundred trillion commensals, represented by viruses, bacteria and fungi. This complex interaction has shown that the microbiome system contributes to the host’s adaptation to its environment, providing genes and functionality that give flexibility of diet and modulate the immune system in order not to reject these symbionts. In the intestine, specifically, the microbiota helps developing organ structures, participates of the metabolism of nutrients and induces immunity. Certain components of the microbiota have been shown to trigger inflammatory responses, whereas others, anti-inflammatory responses. The diversity and the composition of the microbiota, thus, play a key role in the maintenance of intestinal homeostasis and explain partially the link between intestinal microbiota changes and gut-related disorders in humans. Tight junction proteins are key molecules for determination of the paracellular permeability. In the context of intestinal inflammatory diseases, the intestinal barrier is compromised, and decreased expression and differential distribution of tight junction proteins is observed. It is still unclear what is the nature of the luminal or mucosal factors that affect the tight junction proteins function, but the modulation of the immune cells found in the intestinal lamina propria is hypothesized as having a role in this modulation. In this review, we provide an overview of the current understanding of the interaction of the gut microbiota with the immune system in the development and maintenance of the intestinal barrier.
Collapse
|
240
|
Bates J, Diehl L. Dendritic cells in IBD pathogenesis: an area of therapeutic opportunity? J Pathol 2014; 232:112-20. [PMID: 24122796 PMCID: PMC4285849 DOI: 10.1002/path.4277] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 09/03/2013] [Accepted: 09/22/2013] [Indexed: 12/15/2022]
Abstract
Dysfunction of the mucosal immune system plays an important role in inflammatory bowel disease (IBD) pathogenesis. Dendritic cells are emerging as central players based on both our increasing understanding of how genetic susceptibility impacts the mucosal immune system and the key role of dendritic cells in regulating response to gut microflora. We discuss areas of therapeutic opportunity in this evolving landscape. © 2013 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
|
241
|
Berer K, Boziki M, Krishnamoorthy G. Selective accumulation of pro-inflammatory T cells in the intestine contributes to the resistance to autoimmune demyelinating disease. PLoS One 2014; 9:e87876. [PMID: 24504092 PMCID: PMC3913661 DOI: 10.1371/journal.pone.0087876] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/30/2013] [Indexed: 12/17/2022] Open
Abstract
Myelin-specific, pro-inflammatory TH17 cells are widely regarded as the drivers of experimental autoimmune encephalomyelitis (EAE), an animal model for Multiple sclerosis (MS). The factors, responsible for the generation and maintenance of TH17 cells as well as their participation in the pathogenic cascade leading to the demyelinating disease, have been studied extensively. However, how these harmful autoreactive cells are controlled in vivo remains unclear. By comparing TCR transgenic mice on a disease susceptible and a disease resistant genetic background, we show here that pathogenic TH17 cells are sequestered within the intestine of spontaneous EAE resistant B10.S mice. Disease resistant B10.S mice harbored higher frequencies of TH17 cells in the intestine compared to EAE susceptible SJL/J mice. Moreover, transferred TH17 cells selectively migrated to intestinal lymphoid organs of B10.S mice. The sequestration of TH17 cells in the gut was partially dependent on the gut homing receptor α4β7-mediated adhesion to the intestine. Administration of α4β7 blocking-antibodies increased the peripheral availability of TH17 cells, resulting in increased EAE severity after immunization in B10.S mice. Together, these results support the concept that the intestine is a check-point for controlling pathogenic, organ-specific T cells.
Collapse
Affiliation(s)
- Kerstin Berer
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Marina Boziki
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | | |
Collapse
|
242
|
Goodyear AW, Kumar A, Dow S, Ryan EP. Optimization of murine small intestine leukocyte isolation for global immune phenotype analysis. J Immunol Methods 2014; 405:97-108. [PMID: 24508527 DOI: 10.1016/j.jim.2014.01.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/24/2013] [Accepted: 01/28/2014] [Indexed: 02/07/2023]
Abstract
New efforts to understand complex interactions between diet, gut microbiota, and intestinal immunity emphasize the need for a standardized murine protocol that has been optimized for the isolation of lamina propria immune cells. In this study multiple mouse strains including BALB/c, 129S6/Sv/EvTac and ICR mice were utilized to develop an optimal protocol for global analysis of lamina propria leukocytes. Incubation temperature was found to significantly improve epithelial cell removal, while changes in media formulation had minor effects. Tissue weight was an effective method for normalization of solution volumes and incubation times. Collagenase digestion in combination with thermolysin was identified as the optimal method for release of leukocytes from tissues and global immunophenotyping, based on the criteria of minimizing marker cleavage, improving cell viability, and reagent cost. The effects of collagenase in combination with dispase or thermolysin on individual cell surface markers revealed diverse marker specific effects. Aggressive formulations cleaved CD8α, CD138, and B220 from the cell surface, and resulted in relatively higher expression levels of CD3, γδ TCR, CD5, DX5, Ly6C, CD11b, CD11c, MHC-II and CD45. Improved collagenase digestion significantly improved viability and reduced debris formation, eliminating the need for density gradient purification. Finally, we demonstrate that two different digestion protocols yield significant differences in detection of CD4(+) and CD8(+) T cells, NK cells, monocytes and interdigitating DC (iDC) populations, highlighting the importance and impact of cell collection protocols on assay outputs. The optimized protocol described herein will help assure the reproducibility and robustness of global assessment of lamina propria immune responses. Moreover, this technique may be applied to isolation of leukocytes from the entire gastrointestinal tract.
Collapse
Affiliation(s)
- Andrew W Goodyear
- Department of Clinical Sciences, Colorado State University, 300 W. Drake Street, Fort Collins, CO 80523, USA.
| | - Ajay Kumar
- Department of Clinical Sciences, Colorado State University, 300 W. Drake Street, Fort Collins, CO 80523, USA.
| | - Steven Dow
- Department of Clinical Sciences, Colorado State University, 300 W. Drake Street, Fort Collins, CO 80523, USA.
| | - Elizabeth P Ryan
- Department of Clinical Sciences, Colorado State University, 300 W. Drake Street, Fort Collins, CO 80523, USA; Department of Environmental and Radiological Health Sciences, Colorado State University, 200 W. Lake Street, Fort Collins, CO 80523, USA.
| |
Collapse
|
243
|
Gordon JR, Ma Y, Churchman L, Gordon SA, Dawicki W. Regulatory dendritic cells for immunotherapy in immunologic diseases. Front Immunol 2014; 5:7. [PMID: 24550907 PMCID: PMC3907717 DOI: 10.3389/fimmu.2014.00007] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/07/2014] [Indexed: 12/12/2022] Open
Abstract
We recognize well the abilities of dendritic cells to activate effector T cell (Teff cell) responses to an array of antigens and think of these cells in this context as pre-eminent antigen-presenting cells, but dendritic cells are also critical to the induction of immunologic tolerance. Herein, we review our knowledge on the different kinds of tolerogenic or regulatory dendritic cells that are present or can be induced in experimental settings and humans, how they operate, and the diseases in which they are effective, from allergic to autoimmune diseases and transplant tolerance. The primary conclusions that arise from these cumulative studies clearly indicate that the agent(s) used to induce the tolerogenic phenotype and the status of the dendritic cell at the time of induction influence not only the phenotype of the dendritic cell, but also that of the regulatory T cell responses that they in turn mobilize. For example, while many, if not most, types of induced regulatory dendritic cells lead CD4+ naïve or Teff cells to adopt a CD25+Foxp3+ Treg phenotype, exposure of Langerhans cells or dermal dendritic cells to vitamin D leads in one case to the downstream induction of CD25+Foxp3+ regulatory T cell responses, while in the other to Foxp3− type 1 regulatory T cells (Tr1) responses. Similarly, exposure of human immature versus semi-mature dendritic cells to IL-10 leads to distinct regulatory T cell outcomes. Thus, it should be possible to shape our dendritic cell immunotherapy approaches for selective induction of different types of T cell tolerance or to simultaneously induce multiple types of regulatory T cell responses. This may prove to be an important option as we target diseases in different anatomic compartments or with divergent pathologies in the clinic. Finally, we provide an overview of the use and potential use of these cells clinically, highlighting their potential as tools in an array of settings.
Collapse
Affiliation(s)
- John R Gordon
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Yanna Ma
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Laura Churchman
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Sara A Gordon
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Wojciech Dawicki
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| |
Collapse
|
244
|
Kayama H, Nishimura J, Takeda K. Regulation of intestinal homeostasis by innate immune cells. Immune Netw 2013; 13:227-34. [PMID: 24385940 PMCID: PMC3875780 DOI: 10.4110/in.2013.13.6.227] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 02/07/2023] Open
Abstract
The intestinal immune system has an ability to distinguish between the microbiota and pathogenic bacteria, and then activate pro-inflammatory pathways against pathogens for host defense while remaining unresponsive to the microbiota and dietary antigens. In the intestine, abnormal activation of innate immunity causes development of several inflammatory disorders such as inflammatory bowel diseases (IBD). Thus, activity of innate immunity is finely regulated in the intestine. To date, multiple innate immune cells have been shown to maintain gut homeostasis by preventing inadequate adaptive immune responses in the murine intestine. Additionally, several innate immune subsets, which promote Th1 and Th17 responses and are implicated in the pathogenesis of IBD, have recently been identified in the human intestinal mucosa. The demonstration of both murine and human intestinal innate immune subsets contributing to regulation of adaptive immunity emphasizes the conserved innate immune functions across species and might promote development of the intestinal innate immunity-based clinical therapy.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan. ; Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan. ; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan. ; Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan. ; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| |
Collapse
|
245
|
Strbo N, Garcia-Soto A, Schreiber TH, Podack ER. Secreted heat shock protein gp96-Ig: next-generation vaccines for cancer and infectious diseases. Immunol Res 2013; 57:311-25. [PMID: 24254084 DOI: 10.1007/s12026-013-8468-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Over the past decade, our laboratory has developed a secreted heat shock protein (HSP), chaperone gp96, cell-based vaccine that generates effective anti-tumor and anti-infectious immunity in vivo. Gp96-peptide complexes were identified as an extremely efficient stimulator of MHC I-mediated antigen cross-presentation, generating CD8 cytotoxic T-lymphocyte responses detectable in blood, spleen, gut and reproductive tract to femto-molar concentrations of antigen. These studies provided the first evidence that cell-based gp96-Ig-secreting vaccines may serve as a potent modality to induce both systemic and mucosal immunity. This approach takes advantage of the combined adjuvant and antigen delivery capacity of gp96 for the generation of cytotoxic immunity against a wide range of antigens in both anti-vial and anti-cancer vaccination. Here, we review the vaccine design that utilizes the unique property/ability of endoplasmic HSP gp96 to bind antigenic peptides and deliver them to antigen-presenting cells.
Collapse
Affiliation(s)
- Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3008, 1600 NW 10th Ave, Miami, FL, 33136, USA,
| | | | | | | |
Collapse
|
246
|
Ogino T, Nishimura J, Barman S, Kayama H, Uematsu S, Okuzaki D, Osawa H, Haraguchi N, Uemura M, Hata T, Takemasa I, Mizushima T, Yamamoto H, Takeda K, Doki Y, Mori M. Increased Th17-inducing activity of CD14+ CD163 low myeloid cells in intestinal lamina propria of patients with Crohn's disease. Gastroenterology 2013; 145:1380-91.e1. [PMID: 23993972 DOI: 10.1053/j.gastro.2013.08.049] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 08/13/2013] [Accepted: 08/22/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Abnormal activity of innate immune cells and T-helper (Th) 17 cells has been implicated in the pathogenesis of autoimmune and inflammatory diseases, including Crohn's disease (CD). Intestinal innate immune (myeloid) cells have been found to induce development of Th17 cells in mice, but it is not clear if this occurs in humans or in patients with CD. We investigated whether human intestinal lamina propria cells (LPCs) induce development of Th17 cells and whether these have a role in the pathogenesis of CD. METHODS Normal intestinal mucosa samples were collected from patients with colorectal cancer and noninflamed and inflamed regions of mucosa were collected from patients with CD. LPCs were isolated by enzymatic digestion and analyzed for expression of HLA-DR, lineage markers CD14 and CD163 using flow cytometry. RESULTS Among HLA-DR(high) Lin(-) cells, we identified a subset of CD14(+) CD163(low) cells in intestinal LPCs; this subset expressed Toll-like receptor (TLR) 2, TLR4, and TLR5 mRNAs and produced interleukin (IL)-6, IL-1β, and tumor necrosis factor in response to lipopolysaccharide. In vitro co-culture with naïve T cells revealed that CD14(+) CD163(low) cells induced development of Th17 cells. CD14(+) CD163(low) cells from inflamed regions of mucosa of patients with CD expressed high levels of IL-6, IL-23p19, and tumor necrosis factor mRNAs, and strongly induced Th17 cells. CD14(+) CD163(low) cells from the noninflamed mucosa of patients with CD also had increased abilities to induce Th17 cells compared with those from normal intestinal mucosa. CONCLUSIONS CD14(+) CD163(low) cells in intestinal LPCs from normal intestinal mucosa induce differentiation of naive T cells into Th17 cells; this activity is increased in mucosal samples from patients with CD. These findings show how intestinal myeloid cell types could contribute to pathogenesis of CD and possibly other Th17-associated diseases.
Collapse
Affiliation(s)
- Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Thaiss CA, Levy M, Suez J, Elinav E. The interplay between the innate immune system and the microbiota. Curr Opin Immunol 2013; 26:41-8. [PMID: 24556399 DOI: 10.1016/j.coi.2013.10.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/21/2013] [Accepted: 10/28/2013] [Indexed: 01/03/2023]
Abstract
The human gastrointestinal tract harbors one of the highest densities of microorganisms on earth, called the microbiota. In fact, the number of microbial cells in the intestine outnumbers the amount of human cells of the entire organism by a factor of 10. As such, a human being is more and more perceived as a super-organism consisting of a eukaryotic and a prokaryotic part. The compartment mediating the communication between both parts is the innate immune system and its various microbe-sensing pattern-recognition receptors. Co-evolution of the microbiota with the innate immune system has resulted in elaborate interdependency and feedback mechanisms by which both systems control mutual homeostasis. Here, we review the most important innate immune-microbiota interdependencies known to date. While microbial sensing by pattern-recognition receptors is required for stable microbial composition, the presence of the microbiota, in turn, is necessary for proper development and function of the immune system.
Collapse
Affiliation(s)
- Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
248
|
Do JS, Visperas A, Freeman ML, Iwakura Y, Oukka M, Min B. Colitogenic effector T cells: roles of gut-homing integrin, gut antigen specificity and γδ T cells. Immunol Cell Biol 2013; 92:90-8. [PMID: 24189163 PMCID: PMC3947309 DOI: 10.1038/icb.2013.70] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/24/2013] [Accepted: 09/30/2013] [Indexed: 02/08/2023]
Abstract
Disturbance of T cell homeostasis could lead to intestinal inflammation. Naïve CD4 T cells undergoing spontaneous proliferation, a robust proliferative response that occurs under severe lymphopenic conditions, differentiate into effector cells producing Th1 and/or Th17 type cytokines and induce a chronic inflammation in the intestine that resembles human inflammatory bowel disease. In this study, we investigated key properties of CD4 T cells necessary to induce experimental colitis. α4β7 upregulation was primarily induced by mLN resident CD11b+ dendritic cell subsets via TGFβ/retinoic acid-dependent mechanism. Interestingly, α4β7 expression was essential but not sufficient to induce inflammation. In addition to gut homing specificity, expression of gut Ag specificity was also crucial. T cell acquisition of the specificity was dramatically enhanced by the presence of γδ T cells, a population previously shown to exacerbate T cell mediated colitis. Importantly, IL-23-mediated γδ T cell stimulation was necessary to enhance colitogenicity but not gut antigen reactivity of proliferating CD4 T cells. These findings demonstrate that T cell colitogenicity is achieved through multiple processes, offering a therapeutic rationale by intervening these pathways.
Collapse
Affiliation(s)
- Jeong-Su Do
- Department of Immunology/NB30, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Anabelle Visperas
- 1] Department of Immunology/NB30, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA [2] Department of Molecular Medicine, Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - Michael L Freeman
- Department of Immunology/NB30, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Yoichiro Iwakura
- Center of Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Mohamed Oukka
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Booki Min
- 1] Department of Immunology/NB30, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA [2] Department of Molecular Medicine, Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
249
|
Bamias G, Clark DJ, Rivera-Nieves J. Leukocyte traffic blockade as a therapeutic strategy in inflammatory bowel disease. Curr Drug Targets 2013; 14:1490-1500. [PMID: 23621509 PMCID: PMC3779486 DOI: 10.2174/13894501113149990158] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/17/2013] [Accepted: 04/23/2013] [Indexed: 12/16/2022]
Abstract
Dysregulated recruitment of leukocytes into the intestine is a characteristic feature of IBD. Several families of molecules regulate the influx of these cells into sites of inflammation within the gastrointestinal tract. Pharmacological blockade of interactions between molecules that mediate the formation of stable bonds (integrins) and their endothelial ligands has already shown clinical efficacy. Antibodies that target participant molecules have been approved by the US Federal Drug Administration for use in Crohn's, multiple sclerosis (MS) (i.e. natalizumab) and psoriasis (i.e. efalizumab). A more recent additional family of drugs, which might also interfere with lymphocyte traffic (i.e. sphingosine-1- phosphate receptor agonists: fingolimod) is in clinical use for MS and just recently entered the clinical trial stage for ulcerative colitis. In the present review we discuss basic aspects of clinically relevant molecules and compile the clinical studies that support the targeting of specific steps of the leukocyte adhesion cascade for therapeutic purposes in IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- Inflammatory Bowel Disease Center, Division of Gastroenterology, 9500 Gilman Drive, San Diego, California 92093-0063, USA.
| | | | | |
Collapse
|
250
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize recent advances in the fields of intestinal T-regulatory cell (Treg) and tolerogenic dendritic cell subsets. Under homeostatic conditions, CD4(+) Tregs and tolerogenic dendritic cells function to maintain mucosal tolerance. Loss of immune homeostasis is the primary cause of intestinal abnormalities, including inflammatory bowel disease. Thus, an improved understanding of cellular mechanisms promoting tolerance will be critical for the development of more efficacious therapies to treat chronic intestinal inflammation. RECENT FINDINGS Significant progress has been made in the past year in the study of mucosal Treg and dendritic cell populations. In particular, efforts have focused on the migration and differentiation of these cells in the intestinal mucosa, the functional consequences of cross-talk with the intestinal microbiome, mechanisms by which tolerogenic dendritic cells take up antigen, and regulation of retinoic acid synthesis. SUMMARY Recent studies examining tolerogenic cell populations of the intestinal mucosa highlight the progress in understanding the function, regulation, and cross-talk of Treg and dendritic cell populations, and their interactions with the gut microbiota. Scientific advances in these areas will undoubtedly lead to the development of more effective therapeutic strategies for intestinal abnormalities such as inflammatory bowel disease.
Collapse
|