1
|
Zhang T, Liu W, Yang YG. B cell development and antibody responses in human immune system mice: current status and future perspective. SCIENCE CHINA. LIFE SCIENCES 2024; 67:645-652. [PMID: 38270770 DOI: 10.1007/s11427-023-2462-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/28/2023] [Indexed: 01/26/2024]
Abstract
Humanized immune system (HIS) mice have been developed and used as a small surrogate model to study human immune function under normal or disease conditions. Although variations are found between models, most HIS mice show robust human T cell responses. However, there has been unsuccessful in constructing HIS mice that produce high-affinity human antibodies, primarily due to defects in terminal B cell differentiation, antibody affinity maturation, and development of primary follicles and germinal centers. In this review, we elaborate on the current knowledge about and previous attempts to improve human B cell development in HIS mice, and propose a potential strategy for constructing HIS mice with improved humoral immunity by transplantation of human follicular dendritic cells (FDCs) to facilitate the development of secondary follicles.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China.
- International Center of Future Science, Jilin University, Changchun, 130061, China.
| |
Collapse
|
2
|
Eken A, Cansever M, Okus FZ, Erdem S, Nain E, Azizoglu ZB, Haliloglu Y, Karakukcu M, Ozcan A, Devecioglu O, Aksu G, Arikan Ayyildiz Z, Topal E, Karakoc Aydiner E, Kiykim A, Metin A, Cipe F, Kaya A, Artac H, Reisli I, Guner SN, Uygun V, Karasu G, Dönmez Altuntas H, Canatan H, Oukka M, Ozen A, Chatila TA, Keles S, Baris S, Unal E, Patiroglu T. ILC3 deficiency and generalized ILC abnormalities in DOCK8-deficient patients. Allergy 2020; 75:921-932. [PMID: 31596517 DOI: 10.1111/all.14081] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dedicator of cytokinesis 8 (DOCK8) deficiency is the main cause of the autosomal recessive hyper-IgE syndrome (HIES). We previously reported the selective loss of group 3 innate lymphoid cell (ILC) number and function in a Dock8-deficient mouse model. In this study, we sought to test whether DOCK8 is required for the function and maintenance of ILC subsets in humans. METHODS Peripheral blood ILC1-3 subsets of 16 DOCK8-deficient patients recruited at the pretransplant stage, and seven patients with autosomal dominant (AD) HIES due to STAT3 mutations, were compared with those of healthy controls or post-transplant DOCK8-deficient patients (n = 12) by flow cytometry and real-time qPCR. Sorted total ILCs from DOCK8- or STAT3-mutant patients and healthy controls were assayed for survival, apoptosis, proliferation, and activation by IL-7, IL-23, and IL-12 by cell culture, flow cytometry, and phospho-flow assays. RESULTS DOCK8-deficient but not STAT3-mutant patients exhibited a profound depletion of ILC3s, and to a lesser extent ILC2s, in their peripheral blood. DOCK8-deficient ILC1-3 subsets had defective proliferation, expressed lower levels of IL-7R, responded less to IL-7, IL-12, or IL-23 cytokines, and were more prone to apoptosis compared with those of healthy controls. CONCLUSION DOCK8 regulates human ILC3 expansion and survival, and more globally ILC cytokine signaling and proliferation. DOCK8 deficiency leads to loss of ILC3 from peripheral blood. ILC3 deficiency may contribute to the susceptibility of DOCK8-deficient patients to infections.
Collapse
|
3
|
Pilarowski GO, Cazares T, Zhang L, Benjamin JS, Liu K, Jagannathan S, Mousa N, Kasten J, Barski A, Lindsley AW, Bjornsson HT. Abnormal Peyer patch development and B-cell gut homing drive IgA deficiency in Kabuki syndrome. J Allergy Clin Immunol 2020; 145:982-992. [DOI: 10.1016/j.jaci.2019.11.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/05/2019] [Accepted: 11/14/2019] [Indexed: 01/17/2023]
|
4
|
Flip the coin: IL-7 and IL-7R in health and disease. Nat Immunol 2019; 20:1584-1593. [PMID: 31745336 DOI: 10.1038/s41590-019-0479-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.
Collapse
|
5
|
Magrone T, Jirillo E. Development and Organization of the Secondary and Tertiary Lymphoid Organs: Influence of Microbial and Food Antigens. Endocr Metab Immune Disord Drug Targets 2019; 19:128-135. [DOI: 10.2174/1871530319666181128160411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 12/11/2022]
Abstract
Background:Secondary lymphoid organs (SLO) are distributed in many districts of the body and, especially, lymph nodes, spleen and gut-associated lymphoid tissue are the main cellular sites. On the other hand, tertiary lymphoid organs (TLO) are formed in response to inflammatory, infectious, autoimmune and neoplastic events. </P><P> Developmental Studies: In the present review, emphasis will be placed on the developmental differences of SLO and TLO between small intestine and colon and on the role played by various chemokines and cell receptors. Undoubtedly, microbiota is indispensable for the formation of SLO and its absence leads to their poor formation, thus indicating its strict interaction with immune and non immune host cells. Furthermore, food antigens (for example, tryptophan derivatives, flavonoids and byphenils) bind the aryl hydrocarbon receptor on innate lymphoid cells (ILCs), thus promoting the development of postnatal lymphoid tissues. Also retinoic acid, a metabolite of vitamin A, contributes to SLO development during embryogenesis. Vitamin A deficiency seems to account for reduction of ILCs and scarce formation of solitary lymphoid tissue. </P><P> Translational Studies: The role of lymphoid organs with special reference to intestinal TLO in the course of experimental and human disease will also be discussed. </P><P> Future Perspectives: Finally, a new methodology, the so-called “gut-in-a dish”, which has facilitated the in vitro interaction study between microbe and intestinal immune cells, will be described.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
6
|
|
7
|
Rodda LB, Lu E, Bennett ML, Sokol CL, Wang X, Luther SA, Barres BA, Luster AD, Ye CJ, Cyster JG. Single-Cell RNA Sequencing of Lymph Node Stromal Cells Reveals Niche-Associated Heterogeneity. Immunity 2018; 48:1014-1028.e6. [PMID: 29752062 PMCID: PMC5971117 DOI: 10.1016/j.immuni.2018.04.006] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/23/2017] [Accepted: 04/02/2018] [Indexed: 01/06/2023]
Abstract
Stromal cells (SCs) establish the compartmentalization of lymphoid tissues critical to the immune response. However, the full diversity of lymph node (LN) SCs remains undefined. Using droplet-based single-cell RNA sequencing, we identified nine peripheral LN non-endothelial SC clusters. Included are the established subsets, Ccl19hi T-zone reticular cells (TRCs), marginal reticular cells, follicular dendritic cells (FDCs), and perivascular cells. We also identified Ccl19lo TRCs, likely including cholesterol-25-hydroxylase+ cells located at the T-zone perimeter, Cxcl9+ TRCs in the T-zone and interfollicular region, CD34+ SCs in the capsule and medullary vessel adventitia, indolethylamine N-methyltransferase+ SCs in the medullary cords, and Nr4a1+ SCs in several niches. These data help define how transcriptionally distinct LN SCs support niche-restricted immune functions and provide evidence that many SCs are in an activated state.
Collapse
Affiliation(s)
- Lauren B Rodda
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erick Lu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mariko L Bennett
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caroline L Sokol
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaoming Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Sanjiv A Luther
- Department of Biochemistry, Center for Immunity and Infection, University of Lausanne, 1066 Epalinges, Switzerland
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew D Luster
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Chun Jimmie Ye
- Institute for Human Genetics, Department of Epidemiology and Biostatistics, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Robinette ML, Cella M, Telliez JB, Ulland TK, Barrow AD, Capuder K, Gilfillan S, Lin LL, Notarangelo LD, Colonna M. Jak3 deficiency blocks innate lymphoid cell development. Mucosal Immunol 2018; 11:50-60. [PMID: 28513593 PMCID: PMC5693788 DOI: 10.1038/mi.2017.38] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 03/21/2017] [Indexed: 02/04/2023]
Abstract
Loss-of-function mutations in the tyrosine kinase JAK3 cause autosomal recessive severe combined immunodeficiency (SCID). Defects in this form of SCID are restricted to the immune system, which led to the development of immunosuppressive JAK inhibitors. We find that the B6.Cg-Nr1d1tm1Ven/LazJ mouse line purchased from Jackson Laboratories harbors a spontaneous mutation in Jak3, generating a SCID phenotype and an inability to generate antigen-independent professional cytokine-producing innate lymphoid cells (ILCs). Mechanistically, Jak3 deficiency blocks ILC differentiation in the bone marrow at the ILC precursor and the pre-NK cell progenitor. We further demonstrate that the pan-JAK inhibitor tofacitinib and the specific JAK3 inhibitor PF-06651600 impair the ability of human intraepithelial ILC1 (iILC1) to produce IFN-γ, without affecting ILC3 production of IL-22. Both inhibitors impaired the proliferation of iILC1 and ILC3 and differentiation of human ILC in vitro. Tofacitinib is currently approved for the treatment of moderate-to-severely active rheumatoid arthritis. Both tofacitinib and PF-06651600 are currently in clinical trials for several other immune-mediated conditions. Our data suggest that therapeutic inhibition of JAK may also impact ILCs and, to some extent, underlie clinical efficacy.
Collapse
Affiliation(s)
- Michelle L. Robinette
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marina Cella
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Tyler K. Ulland
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander D. Barrow
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kelly Capuder
- Division of Immunology, Harvard Medical School, Boston Children’s Hospital, Boston, MA
| | - Susan Gilfillan
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lih-Ling Lin
- Inflammation and Immunology Research Unit, Pfizer
| | - Luigi D. Notarangelo
- Division of Immunology, Harvard Medical School, Boston Children’s Hospital, Boston, MA,Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Marco Colonna
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
9
|
Withers DR, Hepworth MR. Group 3 Innate Lymphoid Cells: Communications Hubs of the Intestinal Immune System. Front Immunol 2017; 8:1298. [PMID: 29085366 PMCID: PMC5649144 DOI: 10.3389/fimmu.2017.01298] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
The maintenance of mammalian health requires the generation of appropriate immune responses against a broad range of environmental and microbial challenges, which are continually encountered at barrier tissue sites including the skin, lung, and gastrointestinal tract. Dysregulated barrier immune responses result in inflammation, both locally and systemically in peripheral organs. Group 3 innate lymphoid cells (ILC3) are constitutively present at barrier sites and appear to be highly specialized in their ability to sense a range of environmental and host-derived signals. Under homeostatic conditions, ILC3 respond to local cues to maintain tissue homeostasis and restrict inflammatory responses. In contrast, perturbations in the tissue microenvironment resulting from disease, infection, or tissue damage can drive dysregulated pro-inflammatory ILC3 responses and contribute to immunopathology. The tone of the ILC3 response is dictated by a balance of “exogenous” signals, such as dietary metabolites and commensal microbes, and “endogenous” host-derived signals from stromal cells, immune cells, and the nervous system. ILC3 must therefore have the capacity to simultaneously integrate a wide array of complex and dynamic inputs in order to regulate barrier function and tissue health. In this review, we discuss the concept of ILC3 as a “communications hub” in the intestinal tract and associated lymphoid tissues and address the variety of signals, derived from multiple biological systems, which are interpreted by ILC3 to modulate the release of downstream effector molecules and regulate cell–cell crosstalk. Successful integration of environmental cues by ILC3 and downstream propagation to the broader immune system is required to maintain a tolerogenic and anti-inflammatory tone and reinforce barrier function, whereas dysregulation of ILC3 responses can contribute to the onset or progression of clinically relevant chronic inflammatory diseases.
Collapse
Affiliation(s)
- David R Withers
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy (III), University of Birmingham, Birmingham, United Kingdom
| | - Matthew R Hepworth
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Gao J, Zhao L, Liu L, Yang Y, Guo B, Zhu B. Disrupted fibroblastic reticular cells and interleukin-7 expression in tumor draining lymph nodes. Oncol Lett 2017; 14:2954-2960. [PMID: 28928833 PMCID: PMC5588138 DOI: 10.3892/ol.2017.6537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/28/2017] [Indexed: 11/06/2022] Open
Abstract
The immune system of patients with cancer is usually in an inhibitory state. Lymph node (LN) draining of pathological sites provides a suitable microenvironment where adaptive immune responses mainly occur. However, the microenvironment in the tumor draining lymph nodes (TDLNs) of patients with cancer appears to be in favor of tolerance. The effects of tumor cells on TDLNs have not been elaborated clearly. The present results have indicated that tumor cells may directly affect TDLNs by decreasing the fibroblastic reticular cell population that led to less interleukin-7 secretion. As a result, the number of T cells in TDLNs declined with reduced survival signals. A decreased number of T cells in TDLNs means weakened ability of immune surveillance. Clinically, these results were also confirmed in LN biopsies from patients with colon cancer at different clinical stages. Results of the present study showed that tumor cells may directly inhibit the immunological function of TDLNs.
Collapse
Affiliation(s)
- Jianbao Gao
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Lintao Zhao
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China.,Department of Oncology, The People's Liberation Army No. 324 Hospital, Chongqing 404000, P.R. China
| | - Lina Liu
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yang Yang
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Guo
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China.,Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
11
|
Robinette ML, Bando JK, Song W, Ulland TK, Gilfillan S, Colonna M. IL-15 sustains IL-7R-independent ILC2 and ILC3 development. Nat Commun 2017; 8:14601. [PMID: 28361874 PMCID: PMC5380969 DOI: 10.1038/ncomms14601] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/16/2017] [Indexed: 12/18/2022] Open
Abstract
The signals that maintain tissue-resident innate lymphoid cells (ILC) in different microenvironments are incompletely understood. Here we show that IL-7 receptor (IL-7R) is not strictly required for the development of any ILC subset, as residual cells persist in the small intestinal lamina propria (siLP) of adult and neonatal Il7ra-/- mice. Il7ra-/- ILC2 primarily express an ST2- phenotype, but are not inflammatory ILC2. CCR6+ ILC3, which express higher Bcl-2 than other ILC3, are the most abundant subset in Il7ra-/- siLP. All ILC subsets are functionally competent in vitro, and are sufficient to provide enhanced protection to infection with C. rodentium. IL-15 equally sustains wild-type and Il7ra-/- ILC survival in vitro and compensates for IL-7R deficiency, as residual ILCs are depleted in mice lacking both molecules. Collectively, these data demonstrate that siLP ILCs are not completely IL-7R dependent, but can persist partially through IL-15 signalling.
Collapse
Affiliation(s)
- Michelle L. Robinette
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| | - Jennifer K. Bando
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| | - Wilbur Song
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| | - Tyler K. Ulland
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| | - Susan Gilfillan
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| | - Marco Colonna
- Department of Pathology & Immunology, Washington University School of Medicine, 509 S. Euclid Ave Box 8118, St Louis, Missouri 63110, USA
| |
Collapse
|
12
|
Li Z, Shang C. Where have the organizers gone? – The growth control system as a foundation of physiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 123:42-47. [DOI: 10.1016/j.pbiomolbio.2016.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 09/04/2016] [Indexed: 01/24/2023]
|
13
|
Buettner M, Lochner M. Development and Function of Secondary and Tertiary Lymphoid Organs in the Small Intestine and the Colon. Front Immunol 2016; 7:342. [PMID: 27656182 PMCID: PMC5011757 DOI: 10.3389/fimmu.2016.00342] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023] Open
Abstract
The immune system of the gut has evolved a number of specific lymphoid structures that contribute to homeostasis in the face of microbial colonization and food-derived antigenic challenge. These lymphoid organs encompass Peyer’s patches (PP) in the small intestine and their colonic counterparts that develop in a programed fashion before birth. In addition, the gut harbors a network of lymphoid tissues that is commonly designated as solitary intestinal lymphoid tissues (SILT). In contrast to PP, SILT develop strictly after birth and consist of a dynamic continuum of structures ranging from small cryptopatches (CP) to large, mature isolated lymphoid follicles (ILF). Although the development of PP and SILT follow similar principles, such as an early clustering of lymphoid tissue inducer (LTi) cells and the requirement for lymphotoxin beta (LTβ) receptor-mediated signaling, the formation of CP and their further maturation into ILF is associated with additional intrinsic and environmental signals. Moreover, recent data also indicate that specific differences exist in the regulation of ILF formation between the small intestine and the colon. Importantly, intestinal inflammation in both mice and humans is associated with a strong expansion of the lymphoid network in the gut. Recent experiments in mice suggest that these structures, although they resemble large, mature ILF in appearance, may represent de novo-induced tertiary lymphoid organs (TLO). While, so far, it is not clear whether intestinal TLO contribute to the exacerbation of inflammatory pathology, it has been shown that ILF provide the critical microenvironment necessary for the induction of an effective host response upon infection with enteric bacterial pathogens. Regarding the importance of ILF for intestinal immunity, interfering with the development and maturation of these lymphoid tissues may offer novel means for manipulating the immune response during intestinal infection or inflammation.
Collapse
Affiliation(s)
- Manuela Buettner
- Central Animal Facility, Institute of Laboratory Animal Science, Hannover Medical School , Hannover , Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI) , Hannover , Germany
| |
Collapse
|
14
|
Yin J, Sheng B, Qiu Y, Yang K, Xiao W, Yang H. Role of AhR in positive regulation of cell proliferation and survival. Cell Prolif 2016; 49:554-60. [PMID: 27523394 DOI: 10.1111/cpr.12282] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/29/2016] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is an important nuclear transcription factor that is best known for mediating toxic responses by adjusting numbers of metabolism-related enzymes, including CYP1A1 and CYP1B1. Previous findings have revealed that, in addition to negatively regulating cell proliferation and survival, AhR may also positively regulate these pathways. Here, we review these findings and summarize distinct mechanisms by which AhR promotes cell proliferation and survival, including modulation of receptor expression, growth factor signalling and apoptosis, regulating the cell cycle and promoting cytokine expression. This review will aid better understanding the role of AhR in positive regulation of cell proliferation and survival.
Collapse
Affiliation(s)
- Jiuheng Yin
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Baifa Sheng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Kunqiu Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
15
|
Gronke K, Kofoed-Nielsen M, Diefenbach A. Innate lymphoid cells, precursors and plasticity. Immunol Lett 2016; 179:9-18. [PMID: 27394700 DOI: 10.1016/j.imlet.2016.07.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 07/05/2016] [Indexed: 12/30/2022]
Abstract
Innate lymphoid cells (ILC) have only recently been recognized as a separate entity of the lymphoid lineage. Their subpopulations share common characteristics in terms of early development and major transcriptional circuitry with their related cousins of the T cell world. It is currently hypothesized that ILCs constitute an evolutionary older version of the lymphoid immune system. They are found at all primary entry points for pathogens such as mucosal surfaces of the lung and gastrointestinal system, the skin and the liver, which is the central contact point for pathogens that breach the intestinal barrier and enter the circulation. There, ILC contribute to the first line defense as well as to organ homeostasis. However, ILC are not only involved in classical defense tasks, but also contribute to the organogenesis of lymphoid organs as well as tissue remodeling and even stem cell regeneration. ILC may, therefore, implement different functions according to their emergence in ontogeny, their development and their final tissue location. We will review here their early development from precursors of the fetal liver and the adult bone marrow as well as their late plasticity in adaptation to their environment.
Collapse
Affiliation(s)
- Konrad Gronke
- Research Centre Immunology, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany; Institute of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, D79108 Freiburg, Germany
| | - Michael Kofoed-Nielsen
- Research Centre Immunology, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany; Institute of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, D79108 Freiburg, Germany
| | - Andreas Diefenbach
- Research Centre Immunology, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany; Institute of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany.
| |
Collapse
|
16
|
Baerenwaldt A, von Burg N, Kreuzaler M, Sitte S, Horvath E, Peter A, Voehringer D, Rolink AG, Finke D. Flt3 Ligand Regulates the Development of Innate Lymphoid Cells in Fetal and Adult Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:2561-71. [PMID: 26851220 DOI: 10.4049/jimmunol.1501380] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 01/03/2016] [Indexed: 11/19/2022]
Abstract
Flt3 ligand (Flt3L) promotes survival of lymphoid progenitors in the bone marrow and differentiation of dendritic cells (DCs), but its role in regulating innate lymphoid cells (ILCs) during fetal and adult life is not understood. By using Flt3L knockout and transgenic mice, we demonstrate that Flt3L controls ILC numbers by regulating the pool of α4β7(-) and α4β7(+) lymphoid tissue inducer cell progenitors in the fetal liver and common lymphoid progenitors in the bone marrow. Deletion of flt3l severely reduced the number of fetal liver progenitors and lymphoid tissue inducer cells in the neonatal intestine, resulting in impaired development of Peyer's patches. In the adult intestine, NK cells and group 2 and 3 ILCs were severely reduced. This effect occurred independently of DCs as ILC numbers were normal in mice in which DCs were constitutively deleted. Finally, we could show that administration of Flt3L increased the number of NKp46(-) group 3 ILCs in wild-type and even in Il7(-/-) mice, which generally have reduced numbers of ILCs. Taken together, Flt3L significantly contributes to ILC and Peyer's patches development by targeting lymphoid progenitor cells during fetal and adult life.
Collapse
Affiliation(s)
- Anne Baerenwaldt
- University of Basel Children's Hospital, 4056 Basel, Switzerland; Developmental Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Nicole von Burg
- University of Basel Children's Hospital, 4056 Basel, Switzerland; Developmental Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Matthias Kreuzaler
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland; and
| | - Selina Sitte
- Department of Infection Biology, University Clinic of Erlangen, Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Edit Horvath
- University of Basel Children's Hospital, 4056 Basel, Switzerland; Developmental Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Annick Peter
- University of Basel Children's Hospital, 4056 Basel, Switzerland; Developmental Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - David Voehringer
- Department of Infection Biology, University Clinic of Erlangen, Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Antonius G Rolink
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland; and
| | - Daniela Finke
- University of Basel Children's Hospital, 4056 Basel, Switzerland; Developmental Immunology, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland;
| |
Collapse
|
17
|
Differentiation of human innate lymphoid cells (ILCs). Curr Opin Immunol 2016; 38:75-85. [DOI: 10.1016/j.coi.2015.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/25/2023]
|
18
|
Montaldo E, Juelke K, Romagnani C. Group 3 innate lymphoid cells (ILC3s): Origin, differentiation, and plasticity in humans and mice. Eur J Immunol 2015; 45:2171-82. [PMID: 26031799 DOI: 10.1002/eji.201545598] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/18/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022]
Abstract
Since their discovery, innate lymphoid cells (ILCs) have been the subject of intense research. As their name implies, ILCs are innate cells of lymphoid origin, and can be grouped into subsets based on their cytotoxic activity, cytokine profile, and the transcriptional requirements during ILC differentiation. The main ILC groups are "killer" ILCs, comprising NK cells, and "helper-like" ILCs (including ILC1s, ILC2s, and ILC3s). This review examines the origin, differentiation stages, and plasticity of murine and human ILC3s. ILC3s express the retinoic acid receptor (RAR) related orphan receptor RORγt and the signature cytokines IL-22 and IL-17. Fetal ILC3s or lymphoid tissue inducer cells are required for lymphoid organogenesis, while postnatally developing ILC3s are important for the generation of intestinal cryptopatches and isolated lymphoid follicles as well as for the defence against pathogens and epithelial homeostasis. Here, we discuss the transcription factors and exogenous signals (including cytokines, nutrients and cell-to-cell interaction) that drive ILC3 lineage commitment and acquisition of their distinctive effector program.
Collapse
Affiliation(s)
| | - Kerstin Juelke
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Berlin, Germany
| |
Collapse
|
19
|
Group 3 innate lymphoid cells accumulate and exhibit disease-induced activation in the meninges in EAE. Cell Immunol 2015; 297:69-79. [PMID: 26163773 DOI: 10.1016/j.cellimm.2015.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/31/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells are immune cells that reside in tissues that interface with the external environment and contribute to the first line defense against pathogens. However, they also have roles in promoting chronic inflammation. Here we demonstrate that group 3 ILCs, (ILC3s - CD45+Lin-IL-7Rα+RORγt+), are normal residents of the meninges and exhibit disease-induced accumulation and activation in EAE. In addition to production of the pro-inflammatory cytokines IL-17 and GM-CSF, ILC3s constitutively express CD30L and OX40L, molecules required for memory T cell survival. We show that disease-induced trafficking of transferred wild type T cells to the meninges is impaired in ILC3-deficient Rorc-/- mice. Furthermore, lymphoid tissue inducer cells, a c-kit+ ILC3 subset that promotes ectopic lymphoid follicle development, a hallmark of many autoimmune diseases, are reduced in the meninges of EAE-resistant c-kit mutant Kit(W/Wv) mice. We propose that ILC3s sustain neuroinflammation by supporting T cell survival and reactivation in the meninges.
Collapse
|
20
|
Hematopoietic Kit Deficiency, rather than Lack of Mast Cells, Protects Mice from Obesity and Insulin Resistance. Cell Metab 2015; 21:678-91. [PMID: 25955205 DOI: 10.1016/j.cmet.2015.04.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/20/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022]
Abstract
Obesity, insulin resistance, and related pathologies are associated with immune-mediated chronic inflammation. Kit mutant mice are protected from diet-induced obesity and associated co-morbidities, and this phenotype has previously been attributed to their lack of mast cells. We performed a comprehensive metabolic analysis of Kit-dependent Kit(W/Wv) and Kit-independent Cpa3(Cre/+) mast-cell-deficient mouse strains, employing diet-induced or genetic (Lep(Ob/Ob) background) models of obesity. Our results show that mast cell deficiency, in the absence of Kit mutations, plays no role in the regulation of weight gain or insulin resistance. Moreover, we provide evidence that the metabolic phenotype observed in Kit mutant mice, while independent of mast cells, is immune regulated. Our data underscore the value of definitive mast cell deficiency models to conclusively test the involvement of this enigmatic cell in immune-mediated pathologies and identify Kit as a key hematopoietic factor in the pathogenesis of metabolic syndrome.
Collapse
|
21
|
Klose CSN, Diefenbach A. Transcription factors controlling innate lymphoid cell fate decisions. Curr Top Microbiol Immunol 2015; 381:215-55. [PMID: 25038936 DOI: 10.1007/82_2014_381] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mucosal epithelium is in direct contact with symbiotic and pathogenic microorganisms. Therefore, the mucosal surface is the principal portal of entry for invading pathogens and immune cells accumulated in the intestine to prevent infections. In addition to these conventional immune system functions, it has become clear that immune cells during steady-state continuously integrate microbial and nutrient-derived signals from the environment to support organ homeostasis. A major role in both processes is played by a recently discovered group of lymphocytes referred to as innate lymphoid cells (ILCs) Innate lymphoid cells (ILCs) that are specifically enriched at mucosal surfaces but are rather rare in secondary lymphoid organs. In analogy to the dichotomy between CD8 and CD4 T cells, we propose to classify ILCs into interleukin-7 receptor α-negative cytotoxic ILCs and IL-7Rα(+) helper-like ILCs. Dysregulated immune responses triggered by the various ILC subsets have been linked to inflammatory diseases such as inflammatory bowel disease, atopic dermatitis and airway hyperresponsiveness. Here, we will review recent progress in determining the transcriptional and developmental programs that control ILC fate decisions.
Collapse
Affiliation(s)
- Christoph S N Klose
- Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | | |
Collapse
|
22
|
Development of Gut-Associated Lymphoid Tissues. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
23
|
Montaldo E, Teixeira-Alves LG, Glatzer T, Durek P, Stervbo U, Hamann W, Babic M, Paclik D, Stölzel K, Gröne J, Lozza L, Juelke K, Matzmohr N, Loiacono F, Petronelli F, Huntington ND, Moretta L, Mingari MC, Romagnani C. Human RORγt(+)CD34(+) cells are lineage-specified progenitors of group 3 RORγt(+) innate lymphoid cells. Immunity 2014; 41:988-1000. [PMID: 25500367 DOI: 10.1016/j.immuni.2014.11.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 10/27/2014] [Indexed: 12/11/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) are defined by the expression of the transcription factor RORγt, which is selectively required for their development. The lineage-specified progenitors of ILC3s and their site of development after birth remain undefined. Here we identified a population of human CD34(+) hematopoietic progenitor cells (HPCs) that express RORγt and share a distinct transcriptional signature with ILC3s. RORγt(+)CD34(+) HPCs were located in tonsils and intestinal lamina propria (LP) and selectively differentiated toward ILC3s. In contrast, RORγt(-)CD34(+) HPCs could differentiate to become either ILC3s or natural killer (NK) cells, with differentiation toward ILC3 lineage determined by stem cell factor (SCF) and aryl hydrocarbon receptor (AhR) signaling. Thus, we demonstrate that in humans RORγt(+)CD34(+) cells are lineage-specified progenitors of IL-22(+) ILC3s and propose that tonsils and intestinal LP, which are enriched both in committed precursors and mature ILC3s, might represent preferential sites of ILC3 lineage differentiation.
Collapse
Affiliation(s)
- Elisa Montaldo
- Department of Experimental Medicine, University of Genova, Via LB Alberti 2, 16132 Genova, Italy; UOC Immunologia, IRCCS-AOU-San Martino-IST, L.go R. Benzi 10, 16132 Genova, Italy
| | - Luiz Gustavo Teixeira-Alves
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Timor Glatzer
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Pawel Durek
- Cell Biology, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Ulrik Stervbo
- Cell Biology, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Wiebke Hamann
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Marina Babic
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany; Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, B. Branchetta 20a, 51000 Rijeka, Croatia
| | - Daniela Paclik
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany
| | - Katharina Stölzel
- HNO-Klinik-Charité-Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jörn Gröne
- Klinik für Allgemein-, Gefäß- und Thoraxchirurgie Charité-Universitätsmedizin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Laura Lozza
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Kerstin Juelke
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany; Immune System, Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Charité-Universitätsmedizin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nadine Matzmohr
- Immune Regeneration and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Charité-Universitätsmedizin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | - Nicholas David Huntington
- Department of Medical Biology, The Walter and Eliza Hall Institute of Medical Research, The University of Melbourne, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Lorenzo Moretta
- Giannina Gaslini Institute, Via G. Gaslini 5, 16147 Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine, University of Genova, Via LB Alberti 2, 16132 Genova, Italy; UOC Immunologia, IRCCS-AOU-San Martino-IST, L.go R. Benzi 10, 16132 Genova, Italy
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma Forschungszentrum (DRFZ) Berlin, Leibniz-Gemeinschaft, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
24
|
Activated group 3 innate lymphoid cells promote T-cell-mediated immune responses. Proc Natl Acad Sci U S A 2014; 111:12835-40. [PMID: 25136120 DOI: 10.1073/pnas.1406908111] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) have emerged as important cellular players in tissue repair and innate immunity. Whether these cells meaningfully regulate adaptive immune responses upon activation has yet to be explored. Here we show that upon IL-1β stimulation, peripheral ILC3s become activated, secrete cytokines, up-regulate surface MHC class II molecules, and express costimulatory molecules. ILC3s can take up latex beads, process protein antigen, and consequently prime CD4(+) T-cell responses in vitro. The cognate interaction of ILC3s and CD4(+) T cells leads to T-cell proliferation both in vitro and in vivo, whereas its disruption impairs specific T-cell and T-dependent B-cell responses in vivo. In addition, the ILC3-CD4(+) T-cell interaction is bidirectional and leads to the activation of ILC3s. Taken together, our data reveal a novel activation-dependent function of peripheral ILC3s in eliciting cognate CD4(+) T-cell immune responses.
Collapse
|
25
|
Terada T. Huge clusters of embryonic stem cells in human embryos: a morphologic study. Microsc Res Tech 2014; 77:825-31. [PMID: 25091607 DOI: 10.1002/jemt.22405] [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] [Received: 05/19/2014] [Accepted: 06/22/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Nothing is known about huge clusters (HC) of embryonic stem cells (ESC) in human fetal organs (HFO). AIM To know the status of HC-ESC in HFO. METHODS Morphology and immunohistochemistry (IHC) in 32 HFO of 7-40 gestational weeks (GW). RESULTS HC-ESC were seen in many HFO including central nervous system, spinal cords, spine, soft tissue, bone, skin, thyroid, lung, liver, pancreas, gall bladder, extrahepatic bile duct, adrenal, kidney, bladder, foregut, midgut, hindgut, female and male genital organs, and neurons. HC-ESC's were composed of two populations depending on constituting cells. One were large cells with ample acidophilic cytoplasms with vesicular nuclei and nucleoli. The other were small cells with scant cytoplasm with hyperchromatic nuclei without nucleoli, resembling lymphocytes. The HC-ESC were frequently showed neuronal differentiation. HC-ESC were positive for NCAM, synaptophysin, NSE, chromogranin, PDGFRA, AFP, ErbB2, bcl-2, KIT, MET. They were negative for CD45, CD3, CD20, EMA, CEA, CA19-9, cytokeratin (CK) 7, CK8, CK18, CK19, MUC1, MUC2, MUC5AC, and MUC6. The mean Ki-67 labeling index (LI) was 13% ± 7%. HC-ESC showed a little glycogen but lacked mucins. These HC-ESC were seen in 7-25 GW, and they were rarely seen in 26-40 GW. CONCLUSIONS The morphology, IHC, and ontogeny of HC-ESC were described.
Collapse
Affiliation(s)
- Tadashi Terada
- Department of Pathology, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| |
Collapse
|
26
|
Montaldo E, Vacca P, Moretta L, Mingari MC. Development of human natural killer cells and other innate lymphoid cells. Semin Immunol 2014; 26:107-13. [DOI: 10.1016/j.smim.2014.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 01/21/2014] [Indexed: 12/20/2022]
|
27
|
Toward understanding the role of aryl hydrocarbon receptor in the immune system: current progress and future trends. BIOMED RESEARCH INTERNATIONAL 2014; 2014:520763. [PMID: 24527450 PMCID: PMC3914515 DOI: 10.1155/2014/520763] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/14/2013] [Indexed: 01/03/2023]
Abstract
The immune system is regulated by distinct signaling pathways that control the development and function of the immune cells. Accumulating evidence suggest that ligation of aryl hydrocarbon receptor (Ahr), an environmentally responsive transcription factor, results in multiple cross talks that are capable of modulating these pathways and their downstream responsive genes. Most of the immune cells respond to such modulation, and many inflammatory response-related genes contain multiple xenobiotic-responsive elements (XREs) boxes upstream. Active research efforts have investigated the physiological role of Ahr in inflammation and autoimmunity using different animal models. Recently formed paradigm has shown that activation of Ahr by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 3,3′-diindolylmethane (DIM) prompts the differentiation of CD4+Foxp3+ regulatory T cells (Tregs) and inhibits T helper (Th)-17 suggesting that Ahr is an innovative therapeutic strategy for autoimmune inflammation. These promising findings generate a basis for future clinical practices in humans. This review addresses the current knowledge on the role of Ahr in different immune cell compartments, with a particular focus on inflammation and autoimmunity.
Collapse
|
28
|
Ahn YO, Blazar BR, Miller JS, Verneris MR. Lineage relationships of human interleukin-22-producing CD56+ RORγt+ innate lymphoid cells and conventional natural killer cells. Blood 2013; 121:2234-43. [PMID: 23327921 PMCID: PMC3606063 DOI: 10.1182/blood-2012-07-440099] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 01/06/2013] [Indexed: 12/15/2022] Open
Abstract
Human interleukin (IL)-22-producing RORγt(+) innate lymphoid cells (ILC22) and conventional natural killer (cNK) cells are present in secondary lymphoid tissues. Both have an immunophenotype corresponding to stage III NK progenitors (CD56(+/-)CD117(high)CD94(-)). Using an in vitro differentiation and primary human tissues, we investigated their developmental relationships. cNK cells showed a CD56(+)CD117(+)CD7(+/-)LFA-1(high) phenotype and expressed surface receptors, cytokines, and transcription factors found on mature cNK cells. In contrast, ILC22 cells were contained within the CD56(+)CD117(high)CD94(-)CD7(-)LFA-1(-) fraction and produced IL-22, IL-8, and granulocyte macrophage colony stimulating factor. Although ILC22 cells expressed NKp44 and CD161, they lacked most other NK receptors and NK-associated transcription factors (T-bet and Eomes) and were incapable of interferon-γ production or cytotoxic responses. Most purified CD56(+)CD117(+)CD7(+/-)LFA-1(-) remained as ILC22 cells and never became cNK cells. In the absence of IL-15, CD34(+) cells showed a complete block in cNK differentiation and instead gave rise to a CD56(+) population of ILC22 cells. Conversely, in the absence of IL-7 and stem cell factor, cNK cells were generated but ILC22 cells showed minimal differentiation. Although human ILC22 cells and cNK progenitors have a phenotype that overlaps with stage III NK progenitors, they have unique cytokine requirements and can be distinguished by LFA-1 expression.
Collapse
Affiliation(s)
- Yong-Oon Ahn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | | | | | | |
Collapse
|
29
|
Nakagawa R, Togawa A, Nagasawa T, Nishikawa SI. Peyer’s Patch Inducer Cells Play a Leading Role in the Formation of B and T Cell Zone Architecture. THE JOURNAL OF IMMUNOLOGY 2013; 190:3309-18. [DOI: 10.4049/jimmunol.1202766] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Sanos SL, Diefenbach A. Innate lymphoid cells: from border protection to the initiation of inflammatory diseases. Immunol Cell Biol 2013; 91:215-24. [PMID: 23357882 DOI: 10.1038/icb.2013.3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Innate lymphoid cells (ILC) are a recently discovered group of innate lymphocytes found at mucosal surfaces. The transcriptional and effector programs of ILC strikingly resemble those of the various T-helper (Th) cell fates (that is, Th1, Th2, Th9, Th17, Th22). ILC are involved in protecting the mucosal borders by producing tissue protective factors. More recently, evidence has been provided that inappropriately activated ILC can be drivers of various inflammatory disorders. Here, we will highlight recent developments in our understanding of the transcriptional and developmental programs controlling ILC specification and fate decisions. We will also review the roles assigned to ILC in protecting barriers and in promoting inflammatory diseases. Finally, we will outline how the power of ILC may be harnessed for clinical application, and how interference with ILC function may be used as a new strategy to treat inflammatory diseases.
Collapse
Affiliation(s)
- Stephanie L Sanos
- IMMH, Institute of Medical Microbiology and Hygiene, University of Freiburg Medical Centre, Freiburg, Germany.
| | | |
Collapse
|
31
|
Volk A, Hartmann S, Muik A, Geiss Y, Königs C, Dietrich U, von Laer D, Kimpel J. Comparison of three humanized mouse models for adoptive T cell transfer. J Gene Med 2013; 14:540-8. [PMID: 22847974 DOI: 10.1002/jgm.2652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Humanized mouse models for adoptive T cell transfer are important for preclinical efficacy and toxicity studies. However, common xenograft models using immunodeficient mice have so far failed to efficiently support the homing of human T cells to secondary lymphoid tissues. METHODS We established a new mouse model for the adoptive transfer of genetically-modified (gm) T cells using conditioned BALB/c mice. Conditioning involved cyclophosphamide injections, lethal irradiation and radioprotection with bone marrow from immunodeficient mice. We compared repopulation kinetics and the quality of grafts in these modified Trimera (mT3) mice with immunodeficient BALB/c Rag2(-/-) interleukin (IL)2 receptor gamma (rg) knockout (DKO) and NOD/LtSz-scid IL2rg(-/-) (NSG) recipient mouse strains. RESULTS DKO mice showed only marginal engraftment until onset of graft-versus-host disease, whereas mT3 and NSG were repopulated with comparable kinetics. However, T cell repertoire and human cytokine profiles suggest a xenoreactivity-driven gm T cell expansion in mT3 mice, whereas NSG mice were characterized by an initial homeostatic proliferation. Morphological analysis revealed high levels of human gm T cell infiltration in the spleen and liver of all three mouse strains. However, mT3 mice provided the strongest homing of human gm T cells to mucosal sites. Additionally, mT3 mice were the only model with macroscopically visible superficial inguinal lymph nodes. These lymph nodes strongly supported the homing of gm T cells. CONCLUSIONS In the present study, we give proof-of-concept that wild-type mice can accept gm T cell grafts while providing secondary lymphoid structures. Despite limitations, mT3 mice are a valid alternative for applications that specifically rely on improved secondary lymphoid structures.
Collapse
Affiliation(s)
- Andreas Volk
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kiss EA, Vonarbourg C. Aryl hydrocarbon receptor: a molecular link between postnatal lymphoid follicle formation and diet. Gut Microbes 2012; 3:577-82. [PMID: 22909905 PMCID: PMC3495797 DOI: 10.4161/gmic.21865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Intestinal homeostasis results from a complex mutualism between gut microbiota and host cells. Defining the molecular network regulating such mutualism is currently of increasing interest, as its deregulation is reported to lead to increased susceptibility to infections, chronic inflammatory bowel diseases and cancer. Until now, the focus has been on the mechanism, by which the composition of indigenous microbiota shapes the immune system. In a recent study, we have shown that dietary compounds have also the ability to affect innate immune system. This regulation involves aryl hydrocarbon receptor (AhR), a sensor of plant-derived phytochemicals, which mediates the maintenance of Retinoic acid related orphan receptor γ t-expressing innate lymphoid cells (RORγt(+) ILC) in the gut and consequently formation of postnatal lymphoid follicles. Thus, AhR represents the first evidence of a molecular link between diet and immunity at intestinal mucosal surfaces.
Collapse
Affiliation(s)
- Elina A. Kiss
- Institute of Medical Microbiology and Hygiene (IMMH); University of Freiburg Medical Center; Freiburg, Germany,Spemann Graduate School of Biology and Medicine; Freiburg, Germany.,Correspondence to: Elina A. Kiss,
| | - Cedric Vonarbourg
- Institute of Medical Microbiology and Hygiene (IMMH); University of Freiburg Medical Center; Freiburg, Germany
| |
Collapse
|
33
|
Kiss EA, Diefenbach A. Role of the Aryl Hydrocarbon Receptor in Controlling Maintenance and Functional Programs of RORγt(+) Innate Lymphoid Cells and Intraepithelial Lymphocytes. Front Immunol 2012; 3:124. [PMID: 22666222 PMCID: PMC3364460 DOI: 10.3389/fimmu.2012.00124] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 05/02/2012] [Indexed: 12/14/2022] Open
Abstract
Mucosal retinoic receptor-related orphan receptor (ROR)γt-expressing innate lymphoid cells (ILC) play an important role in the defense against intestinal pathogens and in promoting epithelial homeostasis and adaptation, thereby effectively protecting the vertebrate host against intestinal inflammatory disorders. The functional activity of RORγt(+) ILC is under the control of environmental cues. However, the molecular sensors for such environmental signals are largely unknown. Recently, the aryl hydrocarbon receptor (AhR) has emerged as a master regulator for the postnatal maintenance of intestinal RORγt(+) ILC and intraepithelial lymphocytes. AhR is a highly conserved transcription factor whose activity is regulated by environmental and dietary small molecule ligands. Here, we review the role of AhR signaling for the maintenance of intestinal immune cells and its impact on the immunological protection against intestinal infections and debilitating chronic inflammatory disorders.
Collapse
Affiliation(s)
- Elina A Kiss
- Institute of Medical Microbiology and Hygiene, University of Freiburg Freiburg, Germany
| | | |
Collapse
|
34
|
Vonarbourg C, Diefenbach A. Multifaceted roles of interleukin-7 signaling for the development and function of innate lymphoid cells. Semin Immunol 2012; 24:165-74. [PMID: 22541512 DOI: 10.1016/j.smim.2012.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 03/22/2012] [Indexed: 12/20/2022]
Abstract
Recently, additional innate lymphocyte subsets have been identified that express germline encoded immunoreceptors and respond to cytokine cues. Among these, innate lymphoid cells (ILC) at mucosal surfaces are of significant interest because they were found to play important roles for lymphoid organogenesis, tissue homeostasis and repair, for immunity to various infections but also have been involved as disease-promoting cells in models of chronic inflammatory diseases and of autoimmunity. Their functional and transcriptional programs strikingly resemble that of the various T helper cell subsets suggesting that these programs are already pre-formed in the innate immune system and that these may be more conserved than previously appreciated. Interestingly, all ILC subsets express the interleukin 7 receptor α chain and IL-7 signaling has been involved in various aspects of their developmental and functional programs. Here, we will review the role of IL-7 signaling for the differentiation, maintenance and function of two important ILC subsets, lymphoid tissue inducer cells (i.e., RORγt(+) ILC) and natural helper cells (i.e., type 2 ILC). We will also put emphasis on the recently discovered role of IL-7 in controlling plasticity of RORγt(+) ILC.
Collapse
Affiliation(s)
- Cedric Vonarbourg
- Institute of Medical Microbiology & Hygiene, University of Freiburg, Hermann-Herder-Strasse 11, Freiburg, Germany
| | | |
Collapse
|
35
|
Expression and function of interleukin-7 in secondary and tertiary lymphoid organs. Semin Immunol 2012; 24:175-89. [PMID: 22444422 DOI: 10.1016/j.smim.2012.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 01/06/2012] [Accepted: 02/15/2012] [Indexed: 12/23/2022]
Abstract
Interleukin-7 (IL-7) is known since many years as stromal-cell derived cytokine that plays a key role for the adaptive immune system. It promotes lymphocyte development in the bone marrow and thymus as well as naive and memory T cell homeostasis in the periphery. More recently, IL-7 reporter mice and other approaches have led to the further characterization of the various stromal cell sources of IL-7 in secondary lymphoid organs (SLO) and other tissues. We will review these advances along with a discussion of the regulation of IL-7 and its receptor, and compare the biological effects IL-7 has on adaptive as well as innate immune cells in SLO. Finally, we will review the role of IL-7 in development of SLO and tertiary lymphoid tissues that frequently are associated with sites of chronic inflammation.
Collapse
|
36
|
Lee JS, Cella M, Colonna M. AHR and the Transcriptional Regulation of Type-17/22 ILC. Front Immunol 2012; 3:10. [PMID: 22566896 PMCID: PMC3342302 DOI: 10.3389/fimmu.2012.00010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/16/2012] [Indexed: 12/30/2022] Open
Abstract
Mucosal innate lymphoid cells (ILCs) are an emerging population of diverse and heterogeneous immune cells, all with the unique ability to mount a rapid response against invading pathogens. They are further divided into subsets based on their differing cell surface markers as well as in their functional specialization. In this review, we summarize recent reports describing the importance of the transcription factor aryl hydrocarbon receptor (AHR) in regulating the development of one of these subsets, the Type-17/22 ILCs, as well as in the organization of postnatal lymphoid structures. We discuss the mechanisms behind the AHR dependence for development in Type-17/22 ILCs as well as reviewing the proposed physiological ligands that are mediating this effect.
Collapse
Affiliation(s)
- Jacob S Lee
- Department of Pathology and Immunology, Washington University School of Medicine St. Louis, MO, USA
| | | | | |
Collapse
|
37
|
Yoshino N, Kanno H, Takahashi K, Endo M, Sato S. Mucosal Immune Responses in W/Wv and Sl/Sld Mutant Mice. Exp Anim 2012; 61:407-16. [DOI: 10.1538/expanim.61.407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Naoto Yoshino
- Department of Microbiology, Iwate Medical University
| | | | | | - Masahiro Endo
- Department of Microbiology, Iwate Medical University
| | | |
Collapse
|
38
|
Mortha A, Diefenbach A. Natural killer cell receptor-expressing innate lymphocytes: more than just NK cells. Cell Mol Life Sci 2011; 68:3541-55. [PMID: 21904914 PMCID: PMC11114688 DOI: 10.1007/s00018-011-0803-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 08/08/2011] [Accepted: 08/08/2011] [Indexed: 12/17/2022]
Abstract
Recently, additional subsets that extend the family of innate lymphocytes have been discovered. Among these newly identified innate lymphoid cells is a subset sharing phenotypic characteristics of natural killer cells and lymphoid tissue inducer cells. These cells co-express the transcription factor RORγt and activating NK cell receptors (NKR), but their lineage and functional qualities remain poorly defined. Here, we discuss recent proposals to place these NKR(+)RORγt(+) innate lymphocytes on hematopoietic lineage maps. An overview of the transcriptional circuitry determining fate decisions of innate lymphocytes and a summary of current concepts concerning plasticity and stability of innate lymphocyte effector fates are provided. We will conclude by discussing the function of RORγt-expressing innate lymphocytes during inflammatory bowel diseases and in the immune response to tumors.
Collapse
Affiliation(s)
- Arthur Mortha
- IMMH, Institute of Medical Microbiology and Hygiene, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, 79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, 79104 Freiburg, Germany
| | - Andreas Diefenbach
- IMMH, Institute of Medical Microbiology and Hygiene, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, 79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, 79104 Freiburg, Germany
| |
Collapse
|
39
|
Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, Diefenbach A. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 2011; 334:1561-5. [PMID: 22033518 DOI: 10.1126/science.1214914] [Citation(s) in RCA: 633] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Innate lymphoid cells (ILC) expressing the transcription factor RORγt induce the postnatal formation of intestinal lymphoid follicles and regulate intestinal homeostasis. RORγt(+) ILC express the aryl hydrocarbon receptor (AhR), a highly conserved, ligand-inducible transcription factor believed to control adaptation of multicellular organisms to environmental challenges. We show that AhR is required for the postnatal expansion of intestinal RORγt(+) ILC and the formation of intestinal lymphoid follicles. AhR activity within RORγt(+) ILC could be induced by dietary ligands such as those contained in vegetables of the family Brassicaceae. AhR-deficient mice were highly susceptible to infection with Citrobacter rodentium, a mouse model for attaching and effacing infections. Our results establish a molecular link between nutrients and the formation of immune system components required to maintain intestinal homeostasis and resistance to infections.
Collapse
Affiliation(s)
- Elina A Kiss
- Institute of Medical Microbiology and Hygiene, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Wang Z, Jing ZW, Zhou CX, Zhang L, Cheng J, Zhang ZJ, Liu J, Xu CS, Li PT, Wang YY. Fusion of core pathways reveals a horizontal synergistic mechanism underlying combination therapy. Eur J Pharmacol 2011; 667:278-86. [DOI: 10.1016/j.ejphar.2011.05.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 05/01/2011] [Accepted: 05/17/2011] [Indexed: 01/04/2023]
|
41
|
Heijmans J, Büller NV, Muncan V, van den Brink GR. Role of mast cells in colorectal cancer development, the jury is still out. Biochim Biophys Acta Mol Basis Dis 2010; 1822:9-13. [PMID: 21146606 DOI: 10.1016/j.bbadis.2010.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 11/22/2010] [Accepted: 12/01/2010] [Indexed: 12/11/2022]
Abstract
The link between inflammation and colorectal cancer development is becoming increasingly clear. It had long been recognized that patients with inflammatory bowel disease are at an increased risk of colon cancer. Evidence from experimental animals now also implicates the innate immune system in the development of sporadically occurring intestinal adenomas, the precursors to colorectal cancer. Here we discuss the interaction between the immune system and the adenoma to carcinoma sequence with a special emphasis on the role of mast cells which may play a key role in adenoma development. This article is part of a Special Issue entitled: Mast cells in inflammation.
Collapse
Affiliation(s)
- J Heijmans
- Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|