1
|
Li Y, Lopez GE, Lindner PN, Parrella L, Larson M, Sun Y, Stanic AK. The role of RORγt at maternal-fetal interface during murine pregnancy. Am J Reprod Immunol 2020; 84:e13250. [PMID: 32314428 PMCID: PMC8261794 DOI: 10.1111/aji.13250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/15/2020] [Accepted: 04/04/2020] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Innate lymphoid cells (ILCs, including NK cells) and their subsets are the most frequent lymphocytes at the maternal-fetal interface (decidua). Recent recognition of extensive ILC subset diversity at mucosal sites and the possible role they might play at different stages of pregnancy poses questions about their composition and lineage stability. Namely, RORγt-dependent ILC3s have been recognized as a key cellular mediator of tissue organization in the gut and secondary lymphoid organs, prompting examination of their distribution and role in decidua during pregnancy. METHOD OF STUDY We employed highly polychromatic flow cytometry with conventional and machine learning-aided analysis to map ILC subsets and dissected the role of canonical transcription factor RORγt using fate-mapping animals and RORγt-/- animals. RESULTS We demonstrate a comprehensive immunome map of ILCs/NKs, revealing a dynamic interface even in the absence of antigenic or allogeneic challenge. Strikingly, we demonstrate plasticity of RORγt expression in decidual ILCs with across gestation. However, gross reproductive efficiency is not affected in RORγt-/- animals. CONCLUSION These results indicated that RORγt+ ILCs are highly plastic at the maternal-fetal interface, but dispensable for normal pregnancy, revealing a novel mechanism of transcriptional immunoregulation in pregnancy.
Collapse
Affiliation(s)
- Yan Li
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Gladys E. Lopez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Payton N. Lindner
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Luke Parrella
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Mariah Larson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Yan Sun
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
- Reproductive Medicine Center, Fujian Provincial Maternity and Children’s Hospital, Affiliated Hospital of Fujian Medical University
| | - Aleksandar K. Stanic
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
2
|
Chu C, Moriyama S, Li Z, Zhou L, Flamar AL, Klose CSN, Moeller JB, Putzel GG, Withers DR, Sonnenberg GF, Artis D. Anti-microbial Functions of Group 3 Innate Lymphoid Cells in Gut-Associated Lymphoid Tissues Are Regulated by G-Protein-Coupled Receptor 183. Cell Rep 2019; 23:3750-3758. [PMID: 29949760 PMCID: PMC6209103 DOI: 10.1016/j.celrep.2018.05.099] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/25/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
The intestinal tract is constantly exposed to various stimuli. Group 3 innate lymphoid cells (ILC3s) reside in lymphoid organs and in the intestinal tract and are required for immunity to enteric bacterial infection. However, the mechanisms that regulate the ILC3s in vivo remain incompletely defined. Here, we show that GPR183, a chemotactic receptor expressed on murine and human ILC3s, regulates ILC3 migration toward its ligand 7α,25-dihydroxycholesterol (7α,25-OHC) in vitro, and GPR183 deficiency in vivo leads to a disorganized distribution of ILC3s in mesenteric lymph nodes and decreased ILC3 accumulation in the intestine. GPR183 functions intrinsically in ILC3s, and GPR183-deficient mice are more susceptible to enteric bacterial infection. Together, thes1e results reveal a role for the GPR183-7α,25-OHC pathway in regulating the accumulation, distribution, and anti-microbial and tissue-protective functions of ILC3s and define a critical role for this pathway in promoting innate immunity to enteric bacterial infection. Chu et al. demonstrate that GPR183 and its ligand 7α,25-OHC regulate the accumulation, distribution, and antimicrobial and tissue-protective functions of group 3 innate lymphoid cells, thus revealing a critical role for this pathway in promoting innate immunity against enteric bacterial infection.
Collapse
Affiliation(s)
- Coco Chu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Saya Moriyama
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Zhi Li
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Lei Zhou
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Anne-Laure Flamar
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Christoph S N Klose
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Jesper B Moeller
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Molecular Medicine, University of Southern Denmark, Odense 5000, Denmark
| | - Gregory G Putzel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.
| |
Collapse
|
3
|
Taborda NA, Correa LA, Feria MG, Rugeles MT. The Spontaneous Control of HIV Replication is Characterized by Decreased Pathological Changes in the Gut-associated Lymphoid Tissue. Curr HIV Res 2019; 16:338-344. [PMID: 30706820 PMCID: PMC6446516 DOI: 10.2174/1570162x17666190130115113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/22/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND HIV infection induces alterations in the gut-associated lymphoid tissue (GALT) that constitutes the most important site for viral replication due to the extensive presence of effector memory T-cells. In the case of HIV-controllers, several studies have reported fewer peripheral alterations and conserved immune responses that correlate with viral control; however, the histopathological characterization of GALT in those patients is still missing. In this study, we evaluated pathological alterations in GALT, trying to associate them with clinical parameters of HIV infected patients with or without evidence of viral control. METHODS This study included eight HIV-controllers (antiretroviral treatment-naïve patients, with viral loads below 2.000 copies/mL for at least 1 year); 14 Noncontrollers (antiretroviral treatmentnaïve patients, with viral loads > 2.000 copies/mL and CD4+ T cells count > 250 cells/μL), and 12 uninfected donors. Biopsy fragments were obtained by rectosigmoidoscopy and stained with hematoxylin and eosin, silver methenamine, Ziehl Neelsen, and modified Ziehl Neelsen. RESULTS Histopathological findings in HIV-controllers were similar to those observed in the uninfected group. In contrast, noncontrollers exhibited several alterations including condyloma acuminate, squamous metaplasia and acute colitis. These alterations were associated with disease progression. CONCLUSION HIV-controllers exhibit lower pathological alterations in the gut tissue, associated with higher CD4 T cell count, and lower viral load.
Collapse
Affiliation(s)
- Natalia A Taborda
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia.,Grupo de Investigaciones Biomedicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporacion Universitaria Remington, Medellin, Colombia
| | - Luis A Correa
- Seccion de Dermatologia, Departamento de Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia.,Laboratorio de Patología, Laboratorio Clínico VID, Obra de la Congregación Mariana, Medellín, Colombia
| | - Manuel Geronimo Feria
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | - María T Rugeles
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| |
Collapse
|
4
|
|
5
|
Vojkovics D, Kellermayer Z, Gábris F, Schippers A, Wagner N, Berta G, Farkas K, Balogh P. Differential Effects of the Absence of Nkx2-3 and MAdCAM-1 on the Distribution of Intestinal Type 3 Innate Lymphoid Cells and Postnatal SILT Formation in Mice. Front Immunol 2019; 10:366. [PMID: 30891037 PMCID: PMC6413488 DOI: 10.3389/fimmu.2019.00366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/13/2019] [Indexed: 01/08/2023] Open
Abstract
Seeding of leukocytes to developing lymphoid tissues in embryonic and early postnatal age and to the mucosa throughout adulthood depends on the interaction between endothelial MAdCAM-1 addressin and its cognate ligand α4β7 integrin. Nkx2-3 as a transcriptional regulator of MAdCAM-1 controls vascular patterning in visceral lymphoid tissues in mice, and has been identified as a susceptibility factor for inflammatory bowel diseases in humans, associated with lymphoid neogenesis in the inflamed intestines. The role of Nkx2-3 in the organogenesis of the solitary intestinal lymphoid tissues (SILTs) involving type 3 innate lymphoid cells (ILC3) is still unknown. Here we investigated the effect of Nkx2-3 on the postnatal distribution of intestinal ILC3s and the development of SILTs, comparing these to mice lacking MAdCAM-1, but preserving Nkx2-3. At 1 week of age small intestines (SI) contained significantly higher number of ILC3s relative to the colon, with a substantial reduction in MAdCAM-1−/− mice compared to C57BL/6 controls. One week later SI ILC3 number decreased in all genotypes, the number of colonic ILC3 of both Nkx2-3-deficient and Nkx2-3-heterozygous mice significantly increased. On the fourth postnatal week a further reduction of SI ILC3s was observed in both Nkx2-3-deficient and Nkx2-3-heterozygous mice, while in the colon the number of ILC3s showed a significant reduction in all genotypes. At 1 week of age only sporadic SILT components were present in all genotypes. By the second week mice deficient for either Nkx2-3 or MAdCAM-1 showed absence of SILT maturation compared to their relevant controls, lacking mature isolated lymphoid follicles (ILF). By the fourth week both Nkx2-3-deficient and Nkx2-3-heterozygous mice showed a similar distribution of ILFs relative to cryptopatches (CP), whereas in MAdCAM-1−/− mice CPs and immature ILFs were present, mature ILFs were scarce. Our data demonstrate that the complete absence of MAdCAM-1 partially impairs intestinal seeding of ILC3s and causes partial blockade of SILT maturation, without affecting peripheral lymph node development. In contrast, the inactivation of Nkx2-3 permits postnatal seeding, and its blocking effect on SILT maturation prevails at later stage, thus other adhesion molecules may compensate for the intestinal homing of ILC3s in the absence of MAdCAM-1.
Collapse
Affiliation(s)
- Dóra Vojkovics
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Angela Schippers
- Department of Pediatrics, University Hospital RWTH, Aachen, Germany
| | - Norbert Wagner
- Department of Pediatrics, University Hospital RWTH, Aachen, Germany
| | - Gergely Berta
- Central Electron Microscope Laboratory, Department of Medical Biology, Medical School, University of Pécs, Pécs, Hungary
| | - Kornélia Farkas
- Department of Bioanalytics, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Li S, Bostick JW, Zhou L. Regulation of Innate Lymphoid Cells by Aryl Hydrocarbon Receptor. Front Immunol 2018; 8:1909. [PMID: 29354125 PMCID: PMC5760495 DOI: 10.3389/fimmu.2017.01909] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
With striking similarity to their adaptive T helper cell counterparts, innate lymphoid cells (ILCs) represent an emerging family of cell types that express signature transcription factors, including T-bet+ Eomes+ natural killer cells, T-bet+ Eomes- group 1 ILCs, GATA3+ group 2 ILCs, RORγt+ group 3 ILCs, and newly identified Id3+ regulatory ILC. ILCs are abundantly present in barrier tissues of the host (e.g., the lung, gut, and skin) at the interface of host-environment interactions. Active research has been conducted to elucidate molecular mechanisms underlying the development and function of ILCs. The aryl hydrocarbon receptor (Ahr) is a ligand-dependent transcription factor, best known to mediate the effects of xenobiotic environmental toxins and endogenous microbial and dietary metabolites. Here, we review recent progresses regarding Ahr function in ILCs. We focus on the Ahr-mediated cross talk between ILCs and other immune/non-immune cells in host tissues especially in the gut. We discuss the molecular mechanisms of the action of Ahr expression and activity in regulation of ILCs in immunity and inflammation, and the interaction between Ahr and other pathways/transcription factors in ILC development and function with their implication in disease.
Collapse
Affiliation(s)
- Shiyang Li
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - John W. Bostick
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
Tang H, Zhu M, Qiao J, Fu YX. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol 2017; 14:809-818. [PMID: 28413217 PMCID: PMC5649108 DOI: 10.1038/cmi.2017.13] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
Tertiary lymphoid structures (TLS) often develop at sites of persistent inflammation, including cancers and autoimmune diseases. In most cases, the presence of TLS correlates with active immune responses. Because of their proximity to pathological loci, TLS are an intriguing target for the manipulation of immune responses. For several years, it has become clear that lymphotoxin (LT) signalling plays critical roles in lymphoid tissue organogenesis and maintenance. In the current review, we will discuss the role of LT signalling in the development of TLS. With a focus on cancers and autoimmune diseases, we will highlight the correlations between TLS and disease progression. We will also discuss the current efforts and potential directions for manipulating TLS for immunotherapies.
Collapse
Affiliation(s)
- Haidong Tang
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mingzhao Zhu
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Qiao
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
8
|
Li S, Heller JJ, Bostick JW, Lee A, Schjerven H, Kastner P, Chan S, Chen ZE, Zhou L. Ikaros Inhibits Group 3 Innate Lymphoid Cell Development and Function by Suppressing the Aryl Hydrocarbon Receptor Pathway. Immunity 2017; 45:185-97. [PMID: 27438771 DOI: 10.1016/j.immuni.2016.06.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/27/2016] [Accepted: 05/10/2016] [Indexed: 02/08/2023]
Abstract
Group 3 innate lymphoid cells (ILC3s) expressing the transcription factor (TF) RORγt are important for the defense and homeostasis of host intestinal tissues. The zinc finger TF Ikaros, encoded by Ikzf1, is essential for the development of RORγt(+) fetal lymphoid tissue inducer (LTi) cells and lymphoid organogenesis, but its role in postnatal ILC3s is unknown. Here, we show that small-intestinal ILC3s had lower Ikaros expression than ILC precursors and other ILC subsets. Ikaros inhibited ILC3s in a cell-intrinsic manner through zinc-finger-dependent inhibition of transcriptional activity of the aryl hydrocarbon receptor, a key regulator of ILC3 maintenance and function. Ablation of Ikzf1 in RORγt(+) ILC3s resulted in increased expansion and cytokine production of intestinal ILC3s and protection against infection and colitis. Therefore, in contrast to being required for LTi development, Ikaros inhibits postnatal ILC3 development and function to regulate gut immune responses at steady state and in disease.
Collapse
Affiliation(s)
- Shiyang Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Jennifer J Heller
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John W Bostick
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Aileen Lee
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hilde Schjerven
- Department of Laboratory Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Philippe Kastner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Zongming E Chen
- Department of Laboratory Medicine in Geisinger Health System, 100 N. Academy Avenue, MC 19-20, Danville, PA 17822, USA
| | - Liang Zhou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
9
|
Kim M, Kim CH. Colonization and effector functions of innate lymphoid cells in mucosal tissues. Microbes Infect 2016; 18:604-614. [PMID: 27365193 PMCID: PMC5050099 DOI: 10.1016/j.micinf.2016.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/11/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
Innate lymphoid cells (ILCs) protect mucosal barrier tissues to fight infection and maintain tissue integrity. ILCs and their progenitors are developmentally programmed to migrate, differentiate and populate various mucosal tissues and associated lymphoid tissues. Functionally mature ILC subsets respond to diverse pathogens such as bacteria, viruses, fungi and parasites in subset-specific manners. In this review, we will discuss how ILCs populate mucosal tissues and regulate immune responses to distinct pathogens to protect the host and maintain tissue integrity.
Collapse
Affiliation(s)
- Myunghoo Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Weldon School of Biomedical Engineering, Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Chang H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Weldon School of Biomedical Engineering, Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
10
|
Abstract
The human fetus lives in a germ-free intrauterine environment and enters the outside world containing microorganisms from several sources, resulting in gut colonization. Full-term, vaginally born infants are completely colonized with a diverse array of bacterial families in clusters (Phyla) and species (>1000) by the first year of life. Colonizing bacteria communicating with the gut epithelium and underlying lymphoid tissues ('bacterial-epithelial crosstalk') result in a functional immune phenotype and no expression of disease (immune homeostasis). Appropriate colonization is influenced by the prebiotic effect of breast milk oligosaccharides. Adequate colonization results in an innate and adaptive mucosal immune phenotype via communication between molecular patterns on colonizing bacteria and pattern-recognition receptors (e.g., toll-like receptors) on epithelial and lymphoid cells. This ontogeny affects the immune system's capacity to develop oral tolerance to innocuous bacteria and benign antigens. Inadequate intestinal colonization with premature delivery, delivery by Cesarean section and excessive use of perinatal antibiotics results in the absence of adequate bacterial-epithelial crosstalk and an increased incidence of immune-mediated diseases [e.g., asthma, allergy in general and necrotizing enterocolitis (NEC)]. Fortunately, infants with inadequate intestinal colonization can be restored to a bacterial balance with the intake of probiotics. This has been shown to prevent debilitating diseases such as NEC. Thus, understanding the role of gut microbiota in programming of the immune phenotype may be important in preventing disease expression in later childhood and adulthood.
Collapse
|
11
|
Apoptosis of antigen-specific CTLs contributes to low immune response in gut-associated lymphoid tissue post vaccination. Vaccine 2014; 32:5198-205. [PMID: 25066739 DOI: 10.1016/j.vaccine.2014.07.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/26/2014] [Accepted: 07/15/2014] [Indexed: 01/15/2023]
Abstract
The gut-associated lymphoid tissue (GALT) represents a major reservoir of HIV in infected individuals. Vaccines can induce strong systemic immune responses but these have less impact on CD4 T cells activity and numbers in GALT. In this study, we vaccinated mice with an adenovirus vector that expressed the envelope gene from HIV and observed immune responses in the peripheral blood, spleen, liver, mesenteric lymph nodes, and Peyer's patches. We found that (1) the number of HIV-specific CD8 T cells was dramatically lower in GALT than in other tissues; (2) the programmed cell death protein-1 (PD-1) was expressed at high levels in HIV-specific CD8 T cells including memory T cells in GALT; and (3) high levels of HIV-specific CD8 T cell apoptosis were occurring in GALT. These results suggest that contributing to GALT becoming an HIV reservoir during infection is a combination of exhaustion and/or dysfunction of HIV-specific CTLs at that site. These results emphasize the importance of developing of an effective mucosal vaccine against HIV.
Collapse
|
12
|
Kobayashi T, Steinbach EC, Russo SM, Matsuoka K, Nochi T, Maharshak N, Borst LB, Hostager B, Garcia-Martinez JV, Rothman PB, Kashiwada M, Sheikh SZ, Murray PJ, Plevy SE. NFIL3-deficient mice develop microbiota-dependent, IL-12/23-driven spontaneous colitis. THE JOURNAL OF IMMUNOLOGY 2014; 192:1918-27. [PMID: 24442434 DOI: 10.4049/jimmunol.1301819] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
NFIL3 is a transcription factor that regulates multiple immunologic functions. In myeloid cells, NFIL3 is IL-10 inducible and has a key role as a repressor of IL-12p40 transcription. NFIL3 is a susceptibility gene for the human inflammatory bowel diseases. In this article, we describe spontaneous colitis in Nfil3(-/-) mice. Mice lacking both Nfil3 and Il10 had severe early-onset colitis, suggesting that NFIL3 and IL-10 independently regulate mucosal homeostasis. Lymphocytes were necessary for colitis, because Nfil3/Rag1 double-knockout mice were protected from disease. However, Nfil3/Rag1 double-knockout mice adoptively transferred with wild-type CD4(+) T cells developed severe colitis compared with Rag1(-/-) recipients, suggesting that colitis was linked to defects in innate immune cells. Colitis was abrogated in Nfil3/Il12b double-deficient mice, identifying Il12b dysregulation as a central pathogenic event. Finally, germ-free Nfil3(-/-) mice do not develop colonic inflammation. Thus, NFIL3 is a microbiota-dependent, IL-10-independent regulator of mucosal homeostasis via IL-12p40.
Collapse
Affiliation(s)
- Taku Kobayashi
- Center for Gastrointestinal Biology and Diseases, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Innate lymphoid cells in homeostasis, infection, chronic inflammation and tumors of the gastrointestinal tract. Curr Opin Gastroenterol 2013; 29:581-7. [PMID: 24100718 DOI: 10.1097/mog.0b013e328365d339] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW To highlight the functions of a recently discovered group of immune cells known as innate lymphoid cells (ILCs) during homeostasis and infections of the gastrointestinal tract. RECENT FINDINGS ILCs are lymphocytes that lack specific antigen receptors. They are found in the mucosae and mucosal-associated lymphoid tissues, where they promptly initiate cytokine responses to pathogens upon initial exposure. ILCs have been classified into distinct groups based on their cytokine secretion: ILC1 produce IFN-γ, ILC2 secrete IL-5 and IL-13, and ILC3 produce IL-22 and IL-17. Recent studies have discovered the heterogeneity of ILC1 and ILC3 in the gastrointestinal tract. ILC1 subsets may contribute to the inflammatory bowel disease. ILC3 subsets may be beneficial in the defense against gastrointestinal infections, but their sustained activation may lead to cancer. SUMMARY ILCs may provide a target for new avenues of therapeutic intervention in inflammatory bowel disease and gastrointestinal cancer.
Collapse
|
14
|
|
15
|
Nochi T, Denton PW, Wahl A, Garcia JV. Cryptopatches are essential for the development of human GALT. Cell Rep 2013; 3:1874-84. [PMID: 23791525 DOI: 10.1016/j.celrep.2013.05.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/14/2013] [Accepted: 05/22/2013] [Indexed: 12/19/2022] Open
Abstract
Abnormal gut-associated lymphoid tissue (GALT) in humans is associated with infectious and autoimmune diseases, which cause dysfunction of the gastrointestinal (GI) tract immune system. To aid in investigating GALT pathologies in vivo, we bioengineered a human-mouse chimeric model characterized by the development of human GALT structures originating in mouse cryptopatches. This observation expands our mechanistic understanding of the role of cryptopatches in human GALT genesis and emphasizes the evolutionary conservation of this developmental process. Immunoglobulin class switching to IgA occurs in these GALT structures, leading to numerous human IgA-producing plasma cells throughout the intestinal lamina propria. CD4+ T cell depletion within GALT structures results from HIV infection, as it does in humans. This human-mouse chimeric model represents the most comprehensive experimental platform currently available for the study and for the preclinical testing of therapeutics designed to repair disease-damaged GALT.
Collapse
Affiliation(s)
- Tomonori Nochi
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
16
|
Nishimori JH, Newman TN, Oppong GO, Rapsinski GJ, Yen JH, Biesecker SG, Wilson RP, Butler BP, Winter MG, Tsolis RM, Ganea D, Tükel Ç. Microbial amyloids induce interleukin 17A (IL-17A) and IL-22 responses via Toll-like receptor 2 activation in the intestinal mucosa. Infect Immun 2012; 80:4398-408. [PMID: 23027540 PMCID: PMC3497426 DOI: 10.1128/iai.00911-12] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/25/2012] [Indexed: 12/16/2022] Open
Abstract
The Toll-like receptor 2 (TLR2)/TLR1 receptor complex responds to amyloid fibrils, a common component of biofilm material produced by members of the phyla Firmicutes, Bacteroidetes, and Proteobacteria. To determine whether this TLR2/TLR1 ligand stimulates inflammatory responses when bacteria enter intestinal tissue, we investigated whether expression of curli amyloid fibrils by the invasive enteric pathogen Salmonella enterica serotype Typhimurium contributes to T helper 1 and T helper 17 responses by measuring cytokine production in the mouse colitis model. A csgBA mutant, deficient in curli production, elicited decreased expression of interleukin 17A (IL-17A) and IL-22 in the cecal mucosa compared to the S. Typhimurium wild type. In TLR2-deficient mice, IL-17A and IL-22 expression was blunted during S. Typhimurium infection, suggesting that activation of the TLR2 signaling pathway contributes to the expression of these cytokines. T cells incubated with supernatants from bone marrow-derived dendritic cells (BMDCs) treated with curli fibrils released IL-17A in a TLR2-dependent manner in vitro. Lower levels of IL-6 and IL-23 production were detected in the supernatants of the TLR2-deficient BMDCs treated with curli fibrils. Consistent with this, three distinct T-cell populations-CD4(+) T helper cells, cytotoxic CD8(+) T cells, and γδ T cells-produced IL-17A in response to curli fibrils in the intestinal mucosa during S. Typhimurium infection. Notably, decreased IL-6 expression by the dendritic cells and decreased IL-23 expression by the dendritic cells and macrophages were observed in the cecal mucosa of mice infected with the curli mutant. We conclude that TLR2 recognition of bacterial amyloid fibrils in the intestinal mucosa represents a novel mechanism of immunoregulation, which contributes to the generation of inflammatory responses, including production of IL-17A and IL-22, in response to bacterial entry into the intestinal mucosa.
Collapse
Affiliation(s)
- Jessalyn H. Nishimori
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Tiffanny N. Newman
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Gertrude O. Oppong
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Glenn J. Rapsinski
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Jui-Hung Yen
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Steven G. Biesecker
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - R. Paul Wilson
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Brian P. Butler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Maria G. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Renee M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, USA
| | - Doina Ganea
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Çagla Tükel
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
IL-2 receptor γ-chain molecule is critical for intestinal T-cell reconstitution in humanized mice. Mucosal Immunol 2012; 5:555-66. [PMID: 22569301 PMCID: PMC3697924 DOI: 10.1038/mi.2012.31] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Intestinal immune cells are important in host defense, yet the determinants for human lymphoid homeostasis in the intestines are poorly understood. In contrast, lymphoid homeostasis has been studied extensively in mice, where the requirement for a functional common γ-chain molecule has been established. We hypothesized that humanized mice could offer insights into human intestinal lymphoid homeostasis if generated in a strain with an intact mouse common γ-chain molecule. To address this hypothesis, we used three mouse strains (non-obese diabetic (NOD)/severe-combined immunodeficient (SCID) (N/S); NOD/SCID γ-chain(-/-) (NSG); and Rag2(-/-) γ-chain(-/-) (DKO)) and two humanization techniques (bone marrow liver thymus (BLT) and human CD34(+) cell bone marrow transplant of newborn mice (hu)) to generate four common types of humanized mice: N/S-BLT, NSG-BLT, NSG-hu, and DKO-hu mice. The highest levels of intestinal human T cells throughout the small and large intestines were observed in N/S-BLT mice, which have an intact common γ-chain molecule. Furthermore, the small intestine lamina propria T-cell populations of N/S-BLT mice exhibit a human intestine-specific surface phenotype. Thus, the extensive intestinal immune reconstitution of N/S-BLT mice was both quantitatively and qualitatively better when compared with the other models tested such that N/S-BLT mice are well suited for the analysis of human intestinal lymphocyte trafficking and human-specific diseases affecting the intestines.
Collapse
|
18
|
Russell SE, Walsh PT. Sterile inflammation - do innate lymphoid cell subsets play a role? Front Immunol 2012; 3:246. [PMID: 22891068 PMCID: PMC3413015 DOI: 10.3389/fimmu.2012.00246] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/24/2012] [Indexed: 01/13/2023] Open
Abstract
The recent identification of several novel innate lymphoid cell (iLC) subsets has increased our understanding of the mechanisms which link the innate and adaptive immune systems. While the contribution of these subsets toward the pathogenesis of human disease remains largely to be determined, it seems likely that they will play a particularly important role in sterile inflammatory settings where the innate response is seen as a critical mediator of inflammation. Several recent studies have highlighted the role of endogenous damage-associated molecular patterns such as IL-33, IL-1α, and IL-1β in promoting lymphoid cell responses. This review discusses the influence of such endogenous danger signals on novel iLCs such as lymphoid tissue-inducer cells, innate type 2 helper cells, and γδ T cells and explores how these responses may contribute to the development of an inflammatory response in a sterile setting.
Collapse
Affiliation(s)
- Shane E Russell
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
19
|
Fitzsimmons RL, Lau P, Muscat GEO. Retinoid-related orphan receptor alpha and the regulation of lipid homeostasis. J Steroid Biochem Mol Biol 2012; 130:159-68. [PMID: 21723946 DOI: 10.1016/j.jsbmb.2011.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/15/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
Many nuclear hormone receptors (NRs) control lipid, glucose and energy homeostasis in an organ specific manner. Concordantly, dysfunctional NR signalling results in metabolic disease. The Retinoic acid receptor-related orphan receptor alpha (RORα), a member of the NR1F subgroup, is expressed in metabolic tissues. Previous studies identified the role of this NR in dyslipidemia, apo-lipoprotein metabolism and atherosclerosis. Recent data is underscoring the significant role of this orphan NR in the regulation of phase I/II metabolism (bile acids, xenobiotics, steroids etc.), adiposity, insulin signalling, and glucose tolerance. Moreover, oxygenated sterols, have been demonstrated to function as native ligands and inverse agonists. This review focuses on the rapidly emerging and evolving role of RORα in the control of lipid and glucose homeostasis in major mass metabolic tissues. Article from the special issue orphan receptors.
Collapse
Affiliation(s)
- Rebecca L Fitzsimmons
- Obesity Research Centre, Institute for Molecular Bioscience, University of Queensland, Services Rd St. Lucia, Queensland, 4072 Australia
| | | | | |
Collapse
|
20
|
Zhu M, Fu YX. The role of core TNF/LIGHT family members in lymph node homeostasis and remodeling. Immunol Rev 2012; 244:75-84. [PMID: 22017432 DOI: 10.1111/j.1600-065x.2011.01061.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lymph nodes (LNs) maintain active homeostasis at steady state. However, in response to changes in the local environment, such as local infection, cancer, vaccination, and autoimmune disease, dramatic remodeling of LN occurs. This remodeling includes changes in size, lymph and blood flow, immune cell trafficking and cellularity, lymphatic and blood vessel growth and activation, as well as microarchitecture. Therefore, inflammatory conditions often lead to enlarged nodes; after local inflammation resolves, LNs actively regress in size and return to steady state. Remodeling of lymphatic vessels (LVs) and blood vessels (BVs) during both the expansion and regression phases are key steps in controlling LN size as well as function. The cells, membrane-associated molecules, and soluble cytokines that are essential for LV and BV homeostasis as well as dynamic changes in the expansion and regression phases have not been well defined. Understanding the underlying cellular and molecular mechanisms behind LN remodeling would help us to better control undesired immune responses (e.g. inflammation and autoimmune diseases) or promote desired responses (e.g. antitumor immunity and vaccination). In this review, we focus on how the closely related tumor necrosis factor (TNF) members: LIGHT (TNFSF14), lymphotoxin-αβ, and TNF-α contribute to the remodeling of LNs at various stages of inflammation.
Collapse
Affiliation(s)
- Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
21
|
Abstract
The mammalian alimentary tract harbors hundreds of species of commensal microorganisms (microbiota) that intimately interact with the host and provide it with genetic, metabolic, and immunological attributes. Recent reports have indicated that the microbiota composition and its collective genomes (microbiome) are major factors in predetermining the type and robustness of mucosal immune responses. In this review, we discuss the recent advances in our understanding of host-microbiota interactions and their effect on the health and disease susceptibility of the host.
Collapse
Affiliation(s)
- Kenya Honda
- Department of Immunology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| | | |
Collapse
|
22
|
Fuchs A, Colonna M. Natural killer (NK) and NK-like cells at mucosal epithelia: Mediators of anti-microbial defense and maintenance of tissue integrity. Eur J Microbiol Immunol (Bp) 2011; 1:257-66. [PMID: 24516732 DOI: 10.1556/eujmi.1.2011.4.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 10/11/2011] [Indexed: 12/30/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that play important roles in the defense against microbial pathogens through secretion of IFN-γ and recognition and lysis of virally or bacterially infected host cells. A recently identified population of NK-like cells that shares characteristics of both NK cells and lymphoid tissue inducer (LTi) cells promotes innate immune responses in epithelial tissue through the secretion of IL-22. In contrast to classical NK cells, NK-like cells are localized preferentially at mucosal sites, such as the intestinal mucosa. In this review, we consider the function of NK and NK-like cells in anti-microbial defense as well as the maintenance of tissue integrity in the mucosal epithelium of the intestine, lung, and female reproductive tract. Current experimental evidence supports an important protective role for IL-22-producing NK-like cells during intestinal infections, whereas classical NK cells are crucial in the early defense against many pathogens in the respiratory tract. NK cells isolated from the pregnant uterus differ significantly in phenotype and function from those at other tissue locations. Uterine NK cells clearly contribute to the tissue remodeling that takes place during placentation, but their role in anti-microbial defense remains largely undefined.
Collapse
Affiliation(s)
- A Fuchs
- Department of Pathology and Immunology, Washington University School of Medicine St. Louis, Missouri 63110 USA
| | - M Colonna
- Department of Pathology and Immunology, Washington University School of Medicine St. Louis, Missouri 63110 USA
| |
Collapse
|
23
|
Reynders A, Yessaad N, Vu Manh TP, Dalod M, Fenis A, Aubry C, Nikitas G, Escalière B, Renauld JC, Dussurget O, Cossart P, Lecuit M, Vivier E, Tomasello E. Identity, regulation and in vivo function of gut NKp46+RORγt+ and NKp46+RORγt- lymphoid cells. EMBO J 2011; 30:2934-47. [PMID: 21685873 PMCID: PMC3160256 DOI: 10.1038/emboj.2011.201] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/26/2011] [Indexed: 12/14/2022] Open
Abstract
The gut is a major barrier against microbes and encloses various innate lymphoid cells (ILCs), including two subsets expressing the natural cytotoxicity receptor NKp46. A subset of NKp46(+) cells expresses retinoic acid receptor-related orphan receptor γt (RORγt) and produces IL-22, like lymphoid tissue inducer (LTi) cells. Other NKp46(+) cells lack RORγt and produce IFN-γ, like conventional Natural Killer (cNK) cells. The identity, the regulation and the in vivo functions of gut NKp46(+) ILCs largely remain to be unravelled. Using pan-genomic profiling, we showed here that small intestine (SI) NKp46(+)RORγt(-) ILCs correspond to SI NK cells. Conversely, we identified a transcriptional programme conserved in fetal LTi cells and adult SI NKp46(+)RORγt(+) and NKp46(-)RORγt(+) ILCs. We also demonstrated that the IL-1β/IL-1R1/MyD88 pathway, but not the commensal flora, drove IL-22 production by NKp46(+)RORγt(+) ILCs. Finally, oral Listeria monocytogenes infection induced IFN-γ production in SI NK and IL-22 production in NKp46(+)RORγt(+) ILCs, but only IFN-γ contributed to control bacteria dissemination. NKp46(+) ILC heterogeneity is thus associated with subset-specific transcriptional programmes and effector functions that govern their implication in gut innate immunity.
Collapse
Affiliation(s)
- Ana Reynders
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Nadia Yessaad
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Thien-Phong Vu Manh
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Aurore Fenis
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Camille Aubry
- Unité des Interactions Bactéries-Cellules, Department of Cellular Biology and Infection, Institut Pasteur, Paris, France
- Inserm U604, Paris, France
- INRA USC2020, Paris, France
| | - Georgios Nikitas
- Inserm U604, Paris, France
- Microbes and Host Barriers Group, Department of Infection and Epidemiology, Institut Pasteur, Paris, France
| | - Bertrand Escalière
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| | - Jean Christophe Renauld
- Ludwig Institute for Cancer Research Ltd, Experimental Medicine Unit, Universite Catholique de Louvain, Brussels, Belgium
| | - Olivier Dussurget
- Unité des Interactions Bactéries-Cellules, Department of Cellular Biology and Infection, Institut Pasteur, Paris, France
- Inserm U604, Paris, France
- INRA USC2020, Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Department of Cellular Biology and Infection, Institut Pasteur, Paris, France
- Inserm U604, Paris, France
- INRA USC2020, Paris, France
| | - Marc Lecuit
- Inserm U604, Paris, France
- Microbes and Host Barriers Group, Department of Infection and Epidemiology, Institut Pasteur, Paris, France
- Université Paris Descartes, Centre d'Infectiologie Necker-Pasteur, Service des Maladies Infectieuses et Tropicales, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Conception, Marseille, France
| | - Elena Tomasello
- Centre d'Immunologie de Marseille-Luminy, Université de la Mediterannée, Campus du Luminy, Marseille, France
- Institut National de la Santé et de la Recherche Medicale U631, Marseille, France
- Centre National de la Recherche Scientifique, Unite Mixte de Recherche 6102, Marseille, France
| |
Collapse
|
24
|
Dumoutier L, de Heusch M, Orabona C, Satoh-Takayama N, Eberl G, Sirard JC, Di Santo JP, Renauld JC. IL-22 is produced by γC-independent CD25+ CCR6+ innate murine spleen cells upon inflammatory stimuli and contributes to LPS-induced lethality. Eur J Immunol 2011; 41:1075-85. [PMID: 21400499 DOI: 10.1002/eji.201040878] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 11/25/2010] [Accepted: 01/11/2011] [Indexed: 11/07/2022]
Abstract
IL-22 is a Th17 cytokine that plays a key role in immune responses against extracellular bacteria. In mucosal lymphoid tissues, IL-22 production is mainly due to an IL-23-responsive NK-like cell subset that shares some markers with lymphoid tissue inducer (LTi) cells. Here, we identified a new spleen cell population responsible for IL-22 production upon either in vitro stimulation by anti-CD3 antibodies or in vivo stimulation by lipopolysaccharide (LPS) via IL-2- and an IL-23-dependent mechanisms, respectively. These cells represent 1% of spleen cells from recombination activating gene (Rag2)-deficient mice, and correspond to a discrete innate lymphoid cell population expressing CD25, CCR6 and IL-7R. This population comprises 60-70% CD4(+) cells, which produce IL-22, and are still present in common γ chain-deficient mice; the CD4(-) subset coexpresses IL-22 and IL-17, and is common γ chain-dependent. The importance of IL-22 production for the LPS-triggered response is highlighted by the fact that IL-22-deficient mice are more resistant to LPS-induced mortality.
Collapse
Affiliation(s)
- Laure Dumoutier
- Ludwig Institute for Cancer Research, Brussels Branch, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Suzuki K, Kawamoto S, Maruya M, Fagarasan S. GALT: organization and dynamics leading to IgA synthesis. Adv Immunol 2011; 107:153-85. [PMID: 21034974 DOI: 10.1016/b978-0-12-381300-8.00006-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since its discovery more than four decades ago, immunoglobulin (Ig) A has been the subject of continuous and intensive studies. The major concepts derived were that the precursors of IgA plasma cells are generated in follicular organized structures with the help of T cells and the secreted IgAs provide protection against mucosal pathogens. However, only recently we began to appreciate that IgAs play key roles in regulation of bacterial communities in the intestine and that the repertoire of gut microbiota is closely linked to the proper functioning of the immune system. In this review, we highlight the complex and dynamic mutualistic relationships between bacteria and immune cells and discuss the sites and pathways leading to IgA synthesis in gut-associated lymphoid tissues (GALT).
Collapse
Affiliation(s)
- Keiichiro Suzuki
- Research Center for Allergy and Immunology, RIKEN Yokohama Tsurumi, Yokohama, Japan
| | | | | | | |
Collapse
|
26
|
Aliahmad P, de la Torre B, Kaye J. Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages. Nat Immunol 2010; 11:945-52. [PMID: 20818394 PMCID: PMC2943551 DOI: 10.1038/ni.1930] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 08/03/2010] [Indexed: 12/14/2022]
Abstract
TOX is a DNA-binding factor required for development of CD4(+) T cells, natural killer T cells and regulatory T cells. Here we document that both natural killer (NK) cell development and lymphoid tissue organogenesis were also inhibited in the absence of TOX. We found that the development of lymphoid tissue-inducer cells, a rare subset of specialized cells that has an integral role in lymphoid tissue organogenesis, required TOX. Tox was upregulated considerably in immature NK cells in the bone marrow, consistent with the loss of mature NK cells in the absence of this nuclear protein. Thus, many cell lineages of the immune system share a TOX-dependent step for development.
Collapse
Affiliation(s)
- Parinaz Aliahmad
- Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | |
Collapse
|
27
|
Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu Rev Immunol 2010; 28:243-73. [PMID: 20192805 DOI: 10.1146/annurev-immunol-030409-101314] [Citation(s) in RCA: 374] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In mammals, the gastrointestinal tract harbors an extraordinarily dense and complex community of microorganisms. The gut microbiota provide strong selective pressure to the host to evolve adaptive immune responses required for the maintenance of local and systemic homeostasis. The continuous antigenic presence in the gut imposes a dynamic remodeling of gut-associated lymphoid tissues (GALT) and the selection of multiple layered strategies for immunoglobulin (Ig) A production. The composite and dynamic gut environment also necessitates heterogeneous, versatile, and convertible T cells, capable of inhibiting (Foxp3(+) T cells) or helping (T(FH) cells) local immune responses. In this review, we describe recent advances in our understanding of dynamic pathways that lead to IgA synthesis, in gut follicular structures and in extrafollicular sites, by T cell-dependent and T cell-independent mechanisms. We discuss the finely tuned regulatory mechanisms for IgA production and emphasize the role of mucosal IgA in the selection and maintenance of the appropriate microbial composition that is necessary for immune homeostasis.
Collapse
|
28
|
Chong MMW, Simpson N, Ciofani M, Chen G, Collins A, Littman DR. Epigenetic propagation of CD4 expression is established by the Cd4 proximal enhancer in helper T cells. Genes Dev 2010; 24:659-69. [PMID: 20360383 DOI: 10.1101/gad.1901610] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The stability of a lineage program (cellular memory) is dependent on mechanisms that epigenetically maintain active or repressed states of gene expression (transcriptional memory). Although epigenetic silencing of genes has been clearly demonstrated from yeast to mammals, heritable maintenance of active transcription has been less clearly defined. To investigate the potential role of active transcriptional memory during lineage diversification, we employed targeted mutation of a positive-acting cis element in the Cd4 locus to determine the impact on CD4 expression and the differentiation of CD4(+) helper T cells in mice. We show that the proximal enhancer (E4(P)) of Cd4 is essential for CD4 expression in immature CD4(+)8(+) thymocytes. Furthermore, its loss resulted in reduced and unstable expression of CD4 in mature T cells. However, if the enhancer was deleted after cells had already committed to the helper T-cell lineage, CD4 expression remained high and was stable upon cell division. "Active" histone modifications, once initiated by E4(P), were also propagated independently of the enhancer. Thus, E4(P) is responsible for establishing an epigenetically inherited active Cd4 locus in the helper T-cell lineage. To our knowledge, this is the first genetic demonstration of active transcriptional memory in mammalian cells.
Collapse
Affiliation(s)
- Mark M W Chong
- The Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | | | |
Collapse
|
29
|
Finke D. Induction of intestinal lymphoid tissue formation by intrinsic and extrinsic signals. Semin Immunopathol 2009; 31:151-69. [PMID: 19506873 DOI: 10.1007/s00281-009-0163-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/20/2009] [Indexed: 12/20/2022]
Abstract
Since the discovery of inducer cells as a separate lineage for organogenesis of Peyer's patches in the small intestine of fetal mice, a lot of progress has been made in understanding the molecular pathways involved in the generation of lymphoid tissue and the maintenance of the lymphoid architecture. The findings that inducer cells also exist in adult mice and in humans, have a lineage relationship to natural killer cells, and can be stimulated during infections highlight their possible role in establishing innate and adaptive immune responses. Novel concepts in the development of intestinal lymphoid tissues have been made in the past few years suggesting that lymphoid organs are more plastic as previously thought and depend on antigenic stimulation. In addition, the generation of novel lymphoid organs in the gut under inflammatory conditions indicates a function in chronic diseases. The present review summarizes current knowledge on the basic framework of signals required for developing lymphoid tissue under normal and inflammatory conditions.
Collapse
Affiliation(s)
- Daniela Finke
- Department of Biomedicine, Developmental Immunology, University of Basel, Basel, Switzerland.
| |
Collapse
|
30
|
Louten J, Boniface K, de Waal Malefyt R. Development and function of TH17 cells in health and disease. J Allergy Clin Immunol 2009; 123:1004-11. [PMID: 19410689 DOI: 10.1016/j.jaci.2009.04.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 03/31/2009] [Accepted: 04/01/2009] [Indexed: 12/19/2022]
Abstract
T(H)17 cells are the newest member of the T(H) cell family and are characterized by their ability to produce specific cytokines such as IL-17, IL-22, IL-17F, and CCL20. In this review, conditions for the differentiation of T(H)17 cells are defined in both murine and human systems, with discussion of T(H)17-specific cytokines and transcription factors. Functionally, T(H)17 cells contribute to host defense as a new effector T(H) cell subset with a role in protection against extracellular bacteria through activities on immune and nonimmune cells. Their activities, however, are also pivotal in the development of autoimmune diseases under pathologic conditions. T(H)17 cells are also beginning to be associated with the development and pathophysiology of allergic diseases, such as allergic contact dermatitis, atopic dermatitis, and asthma. Lymphoid tissue inducer-like cells and natural killer-like cells, termed RORgammat(+)NKp46(+) or NK-22 cells, might also play a role in allergic diseases because of their propensity to produce IL-17 and IL-22.
Collapse
Affiliation(s)
- Jennifer Louten
- Department of Immunology, Schering-Plough Biopharma, Palo Alto, Calif, USA
| | | | | |
Collapse
|
31
|
Tomasello E, Reynders A, Vivier E. A novel mucosal RORγtNKp46 cell subset is a source of interleukin-22. F1000 BIOLOGY REPORTS 2009; 1:28. [PMID: 20948658 PMCID: PMC2924696 DOI: 10.3410/b1-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lymphoid tissue-inducer cells are hematopoietic cells essential for the organogenesis of several lymphoid structures during both fetal and adult life, whereas natural killer cells are key effector lymphocytes of the innate immune system. A series of recent reports has identified RORγt+NKp46+ interleukin-22-producing cells in gut and tonsils that share features with both lymphoid tissue-inducer cells and natural killer cells and that may be involved in mucosal immunity and homeostasis.
Collapse
Affiliation(s)
- Elena Tomasello
- Centre d Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM U631, CNRS UMR 6102, 163 Avenue du Luminy, Case 906, 13288 Marseille CEDEX 09, France
| | | | | |
Collapse
|
32
|
Kim MY, Kim KS, McConnell F, Lane P. Lymphoid tissue inducer cells: architects of CD4 immune responses in mice and men. Clin Exp Immunol 2009; 157:20-6. [PMID: 19659766 DOI: 10.1111/j.1365-2249.2009.03932.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In this review, we summarize the current understanding of the multiple functions of the mouse lymphoid tissue inducer (LTi) cells in: (i) the development of organized lymphoid tissue, (ii) the generation and maintenance of CD4-dependent immunity in adult lymphoid tissues; and (iii) the regulation of central tolerance in thymus. By contrast with mouse LTi cells, which have been well described, the human equivalent is only just beginning to be characterized. Human LTi-like cells expressing interleukin (IL)-22 have been identified recently and found to differentiate into natural killer (NK) cells. The relationship of LTi cells to NK cells is discussed in the light of several studies reporting a close relationship in the mouse between LTi cells and transcription factor retinoid-related orphan receptor gammat-dependent IL-22 producing NK cells in the gut. We also outline our data suggesting that these cells are present in adult human lymphoid tissues.
Collapse
Affiliation(s)
- M-Y Kim
- Department of Bioinformatics and Life Science, The College of Natural Science, Soongsil University, Seoul, Korea.
| | | | | | | |
Collapse
|
33
|
Vaughn LE, Holt PS, Gast RK. Cellular assessment of crop lymphoid tissue from specific-pathogen-free white leghorn chickens after Salmonella enteritidis challenge. Avian Dis 2009; 52:657-64. [PMID: 19166059 DOI: 10.1637/8369-052308-reg.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The crop may be an important site along the upper alimentary tract in which a humoral immune response against Salmonella Enteritidis (SE) is elicited locally. The mucosal immune response within the crop (ingluvies) of specific-pathogen-free (SPF) white leghorn (WL) chickens against SE was investigated. Three trials were conducted using SPF WL pullets at age 5-6 wk. Trial 1 consisted of 77 birds evaluated for 10 wk post-SE infection (pi), trial 2 was composed of 72 birds monitored through 8 wk pi, and trial 3 was made up of 30 birds assessed for 5 wk pi. Birds were challenged per os with 10(8) colony-forming units/ml SE phage type 13. Crop lavage samples, crop tissues, ceca, and/or liver-spleen were collected preinfection and then at weekly intervals post-SE infection. Bacteriologic examination of cecal contents and/or liver-spleen occurred weekly to monitor progression of SE infection. Crop lavages were analyzed for SE-lipopolysaccharide (LPS)-specific immunoglobulin A (IgA) by enzyme-linked immunosorbent assay to assess humoral immune response. General histologic staining (hematoxylin and eosin [H&E] and methyl green-pyronin [MGP]) and immunohistochemical (IHC) staining (monoclonal antibodies CD45 and Bu-1) were applied to serial sections of crop to evaluate lymphoid tissue via light microscopy, to grade isolated lymphoid follicles (ILFs) by using score 0 (minimal, < 50 microm in diameter) to score 5 (sizable, > 200 microm in diameter) scale, and to characterize the cellular population of ILFs. Results revealed that cecum samples and liver-spleen samples were 100% SE culture positive at 1 wk pi, and then the percentage of SE positives progressively declined over time. Markedly increased crop SE-LPS-specific IgA antibodies were detected in crop samples by 2-3 wk pi, and the humoral response remained elevated above week 0 baseline for the duration of each trial. Crop ILFs of score 3 to 5 were observed in H&E-stained tissues, with an increased proportion of ILFs in post-SE-infected crops vs. uninfected. MGP staining showed plasma cells scattered within and at the periphery of ILFs. IHC staining revealed CD45 (pan-leukocyte) and Bu-1 (B-lymphocyte)-positive cells within crop ILFs. The chicken crop seems to be an organ in which lymphoid tissue may arise in response to enteric SE infection, and a site in which a humoral response may be generated against the SE pathogen.
Collapse
Affiliation(s)
- Lara E Vaughn
- Egg Safety & Quality Research Unit, Russell Research Center, USDA, Agricultural Research Service, 950 College Station Road, Athens, GA 30605, USA
| | | | | |
Collapse
|
34
|
Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, Littman DR, O'Shea JJ. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med 2009; 206:35-41. [PMID: 19114665 PMCID: PMC2626689 DOI: 10.1084/jem.20072713] [Citation(s) in RCA: 596] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 12/02/2008] [Indexed: 02/06/2023] Open
Abstract
The interleukin (IL) 17 family of cytokines has emerged to be critical for host defense as well as the pathogenesis of autoimmune and autoinflammatory disorders, and serves to link adaptive and innate responses. Recent studies have identified a new subset of T cells that selectively produce IL-17 (Th17 cells; Bettelli, E., T. Korn, and V.K. Kuchroo. 2007. Curr. Opin. Immunol. 19:652-657; Kolls, J.K., and A. Linden. 2004. Immunity. 21:467-476), but the regulation of IL-17 production by innate immune cells is less well understood. We report that in vitro stimulation with IL-23 induced IL-17 production by recombination activating gene (Rag) 2(-/-) splenocytes but not Rag2(-/-) common gamma chain(-/-) splenocytes. We found that a major source of IL-17 was CD4(+)CD3(-)NK1.1(-)CD11b(-)Gr1(-)CD11c(-)B220(-) cells, a phenotype that corresponds to lymphoid tissue inducer-like cells (LTi-like cells), which constitutively expressed the IL-23 receptor, aryl hydrocarbon receptor, and CCR6. In vivo challenge with the yeast cell wall product zymosan rapidly induced IL-17 production in these cells. Genetic deletion of signal transducer and activator of transcription 3 reduced but did not abrogate IL-17 production in LTi-like cells. Thus, it appears that splenic LTi-like cells are a rapid source of IL-17 and IL-22, which might contribute to dynamic organization of secondary lymphoid organ structure or host defense.
Collapse
MESH Headings
- Animals
- Antigens, CD/analysis
- CD4 Antigens/analysis
- Cells, Cultured
- DNA-Binding Proteins/genetics
- Flow Cytometry
- Gene Expression/drug effects
- Immune System/cytology
- Immune System/metabolism
- Immunity, Innate/immunology
- Interleukin Receptor Common gamma Subunit/genetics
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-23/pharmacology
- Interleukins/genetics
- Interleukins/immunology
- Interleukins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Nuclear Receptor Subfamily 1, Group F, Member 3
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, CCR6/genetics
- Receptors, Interleukin/genetics
- Receptors, Retinoic Acid/genetics
- Receptors, Thyroid Hormone/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/genetics
- Spleen/cytology
- Spleen/metabolism
- Zymosan/pharmacology
- Interleukin-22
Collapse
Affiliation(s)
- Hiroaki Takatori
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Genetic perturbation of the putative cytoplasmic membrane-proximal salt bridge aberrantly activates alpha(4) integrins. Blood 2008; 112:5007-15. [PMID: 18809756 DOI: 10.1182/blood-2008-03-144543] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
alpha(4) integrins play a pivotal role in leukocyte migration and tissue-specific homing. The ability of integrins to bind ligand is dynamically regulated by activation-dependent conformational changes triggered in the cytoplasmic domain. An NMR solution structure defined a putative membrane-proximal salt bridge between the alpha(IIb)beta(3) integrin cytoplasmic tails, which restrains integrins in their low-affinity state. However, the physiological importance of this salt bridge in alpha(4) integrin regulation remains to be elucidated. To address this question, we disrupted the salt bridge in murine germ line by mutating the conserved cytoplasmic arginine R(GFFKR) in alpha(4) integrins. In lymphocytes from knock-in mice (alpha(4)-R/A(GFFKR)), alpha(4)beta(1) and alpha(4)beta(7) integrins exhibited constitutively up-regulated ligand binding. However, transmigration of these cells across VCAM-1 and MAdCAM-1 substrates, or across endothelial monolayers, was reduced. Perturbed detachment of the tail appeared to cause the reduced cell migration of alpha(4)-R/A(GFFKR) lymphocytes. In vivo, alpha(4)-R/A(GFFKR) cells exhibited increased firm adhesion to Peyer patch venules but reduced homing to the gut. Our results demonstrate that the membrane-proximal salt bridge plays a critical role in supporting proper alpha(4) integrin adhesive dynamics. Loss of this interaction destabilizes the nonadhesive conformation, and thereby perturbs the properly balanced cycles of adhesion and deadhesion required for efficient cell migration.
Collapse
|
36
|
Abstract
Secondary lymphoid organs develop during embryogenesis or in the first few weeks after birth according to a highly coordinated series of interactions between newly emerging hematopoietic cells and immature mesenchymal or stromal cells. These interactions are orchestrated by homeostatic chemokines, cytokines, and growth factors that attract hematopoietic cells to sites of future lymphoid organ development and promote their survival and differentiation. In turn, lymphotoxin-expressing hematopoietic cells trigger the differentiation of stromal and endothelial cells that make up the scaffolding of secondary lymphoid organs. Lymphotoxin signaling also maintains the expression of adhesion molecules and chemokines that govern the ultimate structure and function of secondary lymphoid organs. Here we describe the current paradigm of secondary lymphoid organ development and discuss the subtle differences in the timing, molecular interactions, and cell types involved in the development of each secondary lymphoid organ.
Collapse
|
37
|
Fagarasan S. Evolution, development, mechanism and function of IgA in the gut. Curr Opin Immunol 2008; 20:170-7. [PMID: 18456485 DOI: 10.1016/j.coi.2008.04.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 04/03/2008] [Accepted: 04/07/2008] [Indexed: 12/20/2022]
Abstract
Since its discovery as the most abundant Ig produced at mucosal surfaces, IgA has been the subject of continuous studies. The concepts emerged were that the precursors for IgA plasma cells are efficiently generated in follicular organized structures in the gut with the help of CD4 T cells and that secretory IgA provides protection against mucosal pathogens. Novel conceptual advances have been made in the past few years in describing new sites, mechanisms and functions of mucosal IgA synthesis.
Collapse
Affiliation(s)
- Sidonia Fagarasan
- Laboratory for Mucosal Immunity, RIKEN Research Center for Allergy and Immunology, 230-0045 Tsurumi, Yokohama, Japan.
| |
Collapse
|
38
|
Flores MV, Hall C, Jury A, Crosier K, Crosier P. The zebrafish retinoid-related orphan receptor (ror) gene family. Gene Expr Patterns 2007; 7:535-43. [PMID: 17374568 DOI: 10.1016/j.modgep.2007.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 02/04/2007] [Accepted: 02/05/2007] [Indexed: 10/23/2022]
Abstract
The retinoid-related orphan receptors Rora, b and c are highly conserved transcription factors belonging to the steroid hormone receptor superfamily. Mammalian ROR proteins perform key regulatory roles in a number of processes during embryonic development and in the adult including neurogenesis, bone metabolism and modulation of circadian rhythms. A more recent area of interest has been their roles in the development and function of the immune system. In particular, RORA has been implicated in the regulation of inflammatory cytokine production, and RORC has been shown to be essential in the development of the T lymphocyte repertoire and of secondary lymphoid organs. We cloned the zebrafish orthologs for the Ror gene family. Assignment of orthologies was supported by analysis of the phylogenetic relationships between zebrafish and other vertebrate Ror genes based on sequence similarities, and conserved syntenies with the human Ror gene loci.
Collapse
Affiliation(s)
- Maria Vega Flores
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | |
Collapse
|