1
|
Lv J, Ibrahim YS, Yumashev A, Hjazi A, Faraz A, Alnajar MJ, Qasim MT, Ghildiyal P, Hussein Zwamel A, Fakri Mustafa Y. A comprehensive immunobiology review of IBD: With a specific glance to Th22 lymphocytes development, biology, function, and role in IBD. Int Immunopharmacol 2024; 137:112486. [PMID: 38901239 DOI: 10.1016/j.intimp.2024.112486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
The two primary forms of inflammatory disorders of the small intestine andcolon that make up inflammatory bowel disease (IBD) are ulcerative colitis (UC) and Crohn's disease (CD). While ulcerative colitis primarily affects the colon and the rectum, CD affects the small and large intestines, as well as the esophagus,mouth, anus, andstomach. Although the etiology of IBD is not completely clear, and there are many unknowns about it, the development, progression, and recurrence of IBD are significantly influenced by the activity of immune system cells, particularly lymphocytes, given that the disease is primarily caused by the immune system stimulation and activation against gastrointestinal (GI) tract components due to the inflammation caused by environmental factors such as viral or bacterial infections, etc. in genetically predisposed individuals. Maintaining homeostasis and the integrity of the mucosal barrier are critical in stopping the development of IBD. Specific immune system cells and the quantity of secretory mucus and microbiome are vital in maintaining this stability. Th22 cells are helper T lymphocyte subtypes that are particularly important for maintaining the integrity and equilibrium of the mucosal barrier. This review discusses the most recent research on these cells' biology, function, and evolution and their involvement in IBD.
Collapse
Affiliation(s)
- Jing Lv
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, PR China
| | - Yousif Saleh Ibrahim
- Department of Chemistry and Biochemistry, College of Medicine, University of Fallujah, Fallujah, Iraq
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Ali Faraz
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Majmaah 11952, Saudi Arabia.
| | | | - Maytham T Qasim
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar 64001, Iraq
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Medical Laboratory Technique College, The Islamic University of Aldiwaniyah, Aldiwaniyah, Iraq; Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
2
|
Toriseva M, Björkgren I, Junnila A, Mehmood A, Mattsson J, Raimoranta I, Kim B, Laiho A, Nees M, Elo L, Poutanen M, Breton S, Sipilä P. RUNX transcription factors are essential in maintaining epididymal epithelial differentiation. Cell Mol Life Sci 2024; 81:183. [PMID: 38630262 PMCID: PMC11023966 DOI: 10.1007/s00018-024-05211-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/06/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
Apart from the androgen receptor, transcription factors (TFs) that are required for the development and formation of the different segments of the epididymis have remained unknown. We identified TF families expressed in the developing epididymides, of which many showed segment specificity. From these TFs, down-regulation of runt related transcription factors (RUNXs) 1 and 2 expression coincides with epithelial regression in Dicer1 cKO mice. Concomitant deletion of both Runx1 and Runx2 in a mouse epididymal epithelial cell line affected cell morphology, adhesion and mobility in vitro. Furthermore, lack of functional RUNXs severely disturbed the formation of 3D epididymal organoid-like structures. Transcriptomic analysis of the epididymal cell organoid-like structures indicated that RUNX1 and RUNX2 are involved in the regulation of MAPK signaling, NOTCH pathway activity, and EMT-related gene expression. This suggests that RUNXs are master regulators of several essential signaling pathways, and necessary for the maintenance of proper differentiation of the epididymal epithelium.
Collapse
Affiliation(s)
- Mervi Toriseva
- Institute of Biomedicine, Cancer Research Unit and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Ida Björkgren
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Arttu Junnila
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Arfa Mehmood
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jesse Mattsson
- Institute of Biomedicine, Cancer Research Unit and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Inka Raimoranta
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Bongki Kim
- Program in Membrane Biology/Division of Nephrology, Massachusetts General Hospital, Simches Research Center, Boston, MA, 02114, USA
- Department of Animal Resources Science, Kongju National University, Chungcheongnam-do, Yesan, 32439, Republic of Korea
| | - Asta Laiho
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Matthias Nees
- Institute of Biomedicine, Cancer Research Unit and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Laura Elo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Institute of Medicine, The Sahlgrenska Academy, Gothenburg University, Göteborg, Sweden
| | - Sylvie Breton
- Program in Membrane Biology/Division of Nephrology, Massachusetts General Hospital, Simches Research Center, Boston, MA, 02114, USA
- Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Research Center-CHU de Québec, Université Laval, Québec, QC, Canada
| | - Petra Sipilä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
3
|
Wu Z, Lin X, Yuan G, Li N, Xu R. Innate lymphoid cells: New players in osteoimmunology. Eur J Immunol 2024; 54:e2350381. [PMID: 38234001 DOI: 10.1002/eji.202350381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Innate lymphoid cells (ILCs) are the most recently identified immune cell types existing in lymphoid and nonlymphoid organs. Albeit they lack the expression of antigen receptors, ILCs play vital roles in innate immune responses by producing multiple effector cytokines. The ILC family includes conventional natural killer cells and cytokine-producing ILCs, which are divided into group 1, group 2, and group 3 ILCs based on their effector cytokines and developmental requirements. Emerging evidence has indicated that ILCs are essential immune regulators of bone homeostasis, playing a critical role in osteoimmunology. In this mini-review, we discuss recent advances in the understanding of ILC functions in bone homeostasis under physiological and pathological conditions, with an emphasis on the communication between ILCs and bone cells including osteoclasts and osteoblasts, as well as the underlying immunoregulatory networks involving ILC-derived cytokines and growth factors. This review also discusses future research directions and the potential of targeting ILCs for the treatment of inflammation-associated bone disorders.
Collapse
Affiliation(s)
- Zuoxing Wu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Na Li
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Taggenbrock RLRE, van Gisbergen KPJM. ILC1: Development, maturation, and transcriptional regulation. Eur J Immunol 2023; 53:e2149435. [PMID: 36408791 PMCID: PMC10099236 DOI: 10.1002/eji.202149435] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
Type 1 Innate Lymphoid cells (ILC1s) are tissue-resident cells that partake in the regulation of inflammation and homeostasis. A major feature of ILC1s is their ability to rapidly respond after infections. The effector repertoire of ILC1s includes the pro-inflammatory cytokines IFN-γ and TNF-α and cytotoxic mediators such as granzymes, which enable ILC1s to establish immune responses and to directly kill target cells. Recent advances in the characterization of ILC1s have considerably furthered our understanding of ILC1 development and maintenance in tissues. In particular, it has become clear how ILC1s operate independently from conventional natural killer cells, with which they share many characteristics. In this review, we discuss recent developments with regards to the differentiation, polarization, and effector maturation of ILC1s. These processes may underlie the observed heterogeneity in ILC1 populations within and between different tissues. Next, we highlight transcriptional programs that control each of the separate steps in the differentiation of ILC1s. These transcriptional programs are shared with other tissue-resident type-1 lymphocytes, such as tissue-resident memory T cells (TRM ) and invariant natural killer T cells (iNKT), highlighting that ILC1s utilize networks of transcriptional regulation that are conserved between lymphocyte lineages to respond effectively to tissue-invading pathogens.
Collapse
Affiliation(s)
- Renske L R E Taggenbrock
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Nagel S. The Role of IRX Homeobox Genes in Hematopoietic Progenitors and Leukemia. Genes (Basel) 2023; 14:genes14020297. [PMID: 36833225 PMCID: PMC9957183 DOI: 10.3390/genes14020297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
IRX genes are members of the TALE homeobox gene class and encode six related transcription factors (IRX1-IRX6) controlling development and cell differentiation of several tissues in humans. Classification of TALE homeobox gene expression patterns for the hematopoietic compartment, termed TALE-code, has revealed exclusive IRX1 activity in pro-B-cells and megakaryocyte erythroid progenitors (MEPs), highlighting its specific contribution to developmental processes at these early stages of hematopoietic lineage differentiation. Moreover, aberrant expression of IRX homeobox genes IRX1, IRX2, IRX3 and IRX5 has been detected in hematopoietic malignancies, including B-cell precursor acute lymphoblastic leukemia (BCP-ALL), T-cell ALL, and some subtypes of acute myeloid leukemia (AML). Expression analyses of patient samples and experimental studies using cell lines and mouse models have revealed oncogenic functions in cell differentiation arrest and upstream and downstream genes, thus, revealing normal and aberrant regulatory networks. These studies have shown how IRX genes play key roles in the development of both normal blood and immune cells, and hematopoietic malignancies. Understanding their biology serves to illuminate developmental gene regulation in the hematopoietic compartment, and may improve diagnostic classification of leukemias in the clinic and reveal new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Cultures, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| |
Collapse
|
6
|
Zeiser R, Warnatz K, Rosshart S, Sagar, Tanriver Y. GVHD, IBD and primary immunodeficiencies: The gut as a target of immunopathology resulting from impaired immunity. Eur J Immunol 2022; 52:1406-1418. [PMID: 35339113 DOI: 10.1002/eji.202149530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 01/21/2022] [Indexed: 11/11/2022]
Abstract
The intestinal tract is the largest immunological organ in the body and has a central function of regulating local immune responses, as the intestinal epithelial barrier is a location where the immune system interacts with the gut microbiome including bacteria, fungi and viruses. Impaired immunity in the intestinal tract can lead to immunopathology, which manifests in different diseases such as inflammatory bowel disease (IBD) or intestinal graft-versus-host disease (GVHD). A disturbed communication between epithelial cells, immune cells and microbiome will shape pathogenic immune responses to antigens, which need to be counterbalanced by tolerogenic mechanisms and repair mechanisms. Here, we review how impaired intestinal immune function leads to immunopathology with a specific focus on innate immune cells, the role of the microbiome and the resulting clinical manifestations including intestinal GVHD, IBD and enteropathy in primary immunodeficiency. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology - Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Rosshart
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yakup Tanriver
- Department of Medicine IV (Nephrology and Primary Care), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Microbiology and Hygiene, Institute for Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Zinina VV, Ruehle F, Winkler P, Rebmann L, Lukas H, Möckel S, Diefenbach A, Mendez‐Lago M, Soshnikova N. ID2 controls differentiation of enteroendocrine cells in mouse small intestine. Acta Physiol (Oxf) 2022; 234:e13773. [PMID: 34985199 DOI: 10.1111/apha.13773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/15/2021] [Accepted: 01/01/2022] [Indexed: 12/17/2022]
Abstract
AIMS The mammalian gut is the largest endocrine organ. Dozens of hormones secreted by enteroendocrine cells regulate a variety of physiological functions of the gut but also of the pancreas and brain. Here, we examined the role of the helix-loop-helix transcription factor ID2 during the differentiation of intestinal stem cells along the enteroendocrine lineage. METHODS To assess the functions of ID2 in the adult mouse small intestine, we used single-cell RNA sequencing, genetically modified mice, and organoid assays. RESULTS We found that in the adult intestinal epithelium Id2 is predominantly expressed in enterochromaffin and peptidergic enteroendocrine cells. Consistently, the loss of Id2 leads to the reduction of Chromogranin A-positive enteroendocrine cells. In contrast, the numbers of tuft cells are increased in Id2 mutant small intestine. Moreover, ablation of Id2 elevates the numbers of Serotonin+ enterochromaffin cells and Ghrelin+ X-cells in the posterior part of the small intestine. Finally, ID2 acts downstream of BMP signalling during the differentiation of Glucagon-like peptide-1+ L-cells and Cholecystokinin+ I-cells towards Neurotensin+ PYY+ N-cells. CONCLUSION ID2 plays an important role in cell fate decisions in the adult small intestine. First, ID2 is essential for establishing a differentiation gradient for enterochromaffin and X-cells along the anterior-posterior axis of the gut. Next, ID2 is necessary for the differentiation of N-cells thus ensuring a differentiation gradient along the crypt-villi axis. Finally, ID2 suppresses the commitment of secretory intestinal epithelial progenitors towards tuft cell lineage and thus controls host immune response to commensal and parasitic microbiota.
Collapse
Affiliation(s)
- Valeriya V. Zinina
- Institute for Molecular Medicine University Medical Center of the Johannes Gutenberg‐University Mainz Germany
| | - Frank Ruehle
- Institute of Molecular Biology gGmbH Mainz Germany
| | - Patricia Winkler
- MSc Programme in Biomedicine Institute for Molecular Medicine University Medical Center of the Johannes Gutenberg‐University Mainz Germany
| | - Lisa Rebmann
- Institute for Molecular Medicine University Medical Center of the Johannes Gutenberg‐University Mainz Germany
- Faculty Medical and Life Sciences Hochschule Furtwangen University Furtwangen Germany
| | - Hanna Lukas
- Institute of Molecular Biology gGmbH Mainz Germany
| | | | - Andreas Diefenbach
- Laboratory of Innate Immunity Department of Microbiology, Infectious Diseases and Immunology Charité‐Universitätsmedizin Berlin Berlin Germany
- Berlin Institute of Health (BIH) Berlin Germany
- Mucosal and Developmental Immunology Deutsches Rheuma‐Forschungszentrum (DRFZ) Berlin Germany
| | | | - Natalia Soshnikova
- Institute for Molecular Medicine University Medical Center of the Johannes Gutenberg‐University Mainz Germany
| |
Collapse
|
8
|
Felipe Pereira R, Willian Lattari Tessarin G, Yamamoto Chiba F, Sara de Lima Coutinho Mattera M, Gomes Pereira A, Verônica Saori Tsosura T, Gustavo Balera Brito V, Akira Fujii de Oliveira R, Ervolino E, Helena Penha de Oliveira S, Tavares Angelo Cintra L, Hissako Matsushita D. Apical periodontitis promotes insulin resistance and alters adaptive immunity markers in rats. Saudi Dent J 2021; 33:979-986. [PMID: 34916766 PMCID: PMC8665232 DOI: 10.1016/j.sdentj.2021.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 05/25/2021] [Accepted: 07/04/2021] [Indexed: 11/17/2022] Open
Abstract
Periapical lesions promotes higher T-bet expression and lower GATA3 and FOXP3 expression. Periapical lesion promotes increased GATA3 and decreased FOXP3 expression in the group with 4 periapical lesions (4PL) The adaptive immune system, with a predominance of the Th1 polarization, may be involved in the development of insulin resistance in rats with Periapical lesions.
Objective Apical periodontitis (AP) is a chronic or acute inflammatory disease usually developed from endodontic infections, predominantly due to gram-negative anaerobic bacteria invading the dental pulp. This study aimed to evaluate lymphocyte markers to assess the involvement of adaptive immunity in insulin resistance (IR) in a rat model of AP. Design. Forty-five male Wistar albino rats were divided into 3 groups (control, 1AP and 4AP). AP was induced in the upper right first molar (1AP), and in the first and second upper and lower right molars (4AP). The spleen was collected to evaluate the expression of transcription factors involved in lymphocyte polarization, including T-bet (Th1), GATA3 (Th2), and FOXP3 (Treg). Blood samples were assessed for serum cytokine levels transcribed by the respective lymphocyte polarizations, INF-γ (Th1), IL-4 (Th2) and TGF-β (Treg). In addition, glucose and insulin levels were measured to evaluate IR by the HOMA-IR method. Results The results showed higher T-bet expression on AP groups, along with lower GATA3 and FOXP3 expression in the 1AP, in addition to increased GATA3 and decreased FOXP3 expression in the 4AP group compared to the CN group. There was no difference in the INF-γ levels, while IL-4 was decreased in the AP groups. Taken together, these results suggest that the adaptive immune system, with a predominance of the Th1 polarization, may be involved in the development of IR in rats with AP. Conclusions AP promotes increase in the expression of T-bet (4AP) and decrease of FOXP3 expressions and IL-4 levels (1AP and 4AP). However, depending on the number of lesions (1 or 4 lesions), the expression of GATA3 appears differently. Thus, innate immunity and adaptive immunity may contribute to the IR observed in rats with AP.
Collapse
Key Words
- 1AP, group with 1 apical periodontitis
- 4AP, group with 4 apical periodontitis (4AP)
- AP, Apical periodontitis
- Adaptive immunity
- Apical periodontitis
- Endodontics
- FOXP3, forkhead box P3
- GATA3, Binding protein 3
- IL-4, interleukin-4
- INF-γ, Interferon-gamma
- IR, Insulin resistance
- Insulin resistance
- T-bet, T-box transcription factor TBX21
- TGF-β, Transforming growth factor beta
- Th1, T helper 1
- Th2, T helper 2
- Treg, Regulatory T cell
Collapse
Affiliation(s)
- Renato Felipe Pereira
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil
| | - Gestter Willian Lattari Tessarin
- Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil.,University Center North Paulista (UNORP), São José do Rio Preto, SP, Brazil
| | - Fernando Yamamoto Chiba
- Department of Preventive and Restorative Dentistry, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Maria Sara de Lima Coutinho Mattera
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil
| | - Amanda Gomes Pereira
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Thais Verônica Saori Tsosura
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil
| | - Victor Gustavo Balera Brito
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil
| | - Renan Akira Fujii de Oliveira
- Department of Preventive and Restorative Dentistry, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil.,Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Sandra Helena Penha de Oliveira
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil
| | - Luciano Tavares Angelo Cintra
- Department of Preventive and Restorative Dentistry, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| | - Doris Hissako Matsushita
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, PPGMCF, SBFis, São Paulo State University (UNESP), School of Dentistry, Araçatuba, São Paulo, Brazil.,Department of Basic Sciences, São Paulo State University (UNESP), School of Dentistry, Araçatuba, SP, Brazil
| |
Collapse
|
9
|
Kim J, Ryu S, Kim HY. Innate Lymphoid Cells in Tissue Homeostasis and Disease Pathogenesis. Mol Cells 2021; 44:301-309. [PMID: 33972473 PMCID: PMC8175152 DOI: 10.14348/molcells.2021.0053] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently discovered family of innate immune cells. ILCs can be categorized into three groups on the basis of the transcription factors that direct their functions and the cytokines they produce. Notably, these functions parallel the effector functions of T lymphocytes. ILCs play a frontline role in host defense and tissue homeostasis by responding rapidly to environmental factors, conducting effector responses in a tissue-specific manner, and interacting with hematopoietic and non-hematopoietic cells throughout the body. Moreover, recent studies reveal that ILCs are involved in development of various inflammatory diseases, such as respiratory diseases, autoimmune diseases, or cancer. In this review, we discuss the recent findings regarding the biology of ILCs in health and inflammatory diseases.
Collapse
Affiliation(s)
- Jihyun Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Seungwon Ryu
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
10
|
NKL-Code in Normal and Aberrant Hematopoiesis. Cancers (Basel) 2021; 13:cancers13081961. [PMID: 33921702 PMCID: PMC8073162 DOI: 10.3390/cancers13081961] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Gene codes represent expression patterns of closely related genes in particular tissues, organs or body parts. The NKL-code describes the activity of NKL homeobox genes in the hematopoietic system. NKL homeobox genes encode transcription factors controlling basic developmental processes. Therefore, aberrations of this code may contribute to deregulated hematopoiesis including leukemia and lymphoma. Normal and abnormal activities of NKL homeobox genes are described and mechanisms of (de)regulation, function, and diseases exemplified. Abstract We have recently described physiological expression patterns of NKL homeobox genes in early hematopoiesis and in subsequent lymphopoiesis and myelopoiesis, including terminally differentiated blood cells. We thereby systematized differential expression patterns of eleven such genes which form the so-called NKL-code. Due to the developmental impact of NKL homeobox genes, these data suggest a key role for their activity in normal hematopoietic differentiation processes. On the other hand, the aberrant overexpression of NKL-code-members or the ectopical activation of non-code members have been frequently reported in lymphoid and myeloid leukemia/lymphoma, revealing the oncogenic potential of these genes in the hematopoietic compartment. Here, I provide an overview of the NKL-code in normal hematopoiesis and instance mechanisms of deregulation and oncogenic functions of selected NKL genes in hematologic cancers. As well as published clinical studies, our conclusions are based on experimental work using hematopoietic cell lines which represent useful models to characterize the role of NKL homeobox genes in specific tumor types.
Collapse
|
11
|
Eomes cannot replace its paralog T-bet during expansion and differentiation of CD8 effector T cells. PLoS Pathog 2020; 16:e1008870. [PMID: 32991634 PMCID: PMC7546498 DOI: 10.1371/journal.ppat.1008870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/09/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022] Open
Abstract
The two T-box transcription factors T-bet and Eomesodermin (Eomes) are important regulators of cytotoxic lymphocytes (CTLs), such as activated CD8 T cells, which are essential in the fight against intracellular pathogens and tumors. Both transcription factors share a great degree of homology based on sequence analysis and as a result exert partial functional redundancy during viral infection. However, the actual degree of redundancy between T-bet and Eomes remains a matter of debate and is further confounded by their distinct spatiotemporal expression pattern in activated CD8 T cells. To directly investigate the functional overlap of these transcription factors, we generated a new mouse model in which Eomes expression is under the transcriptional control of the endogenous Tbx21 (encoding for T-bet) locus. Applying this model, we demonstrate that the induction of Eomes in lieu of T-bet cannot rescue T-bet deficiency in CD8 T cells during acute lymphocytic choriomeningitis virus (LCMV) infection. We found that the expression of Eomes instead of T-bet was not sufficient for early cell expansion or effector cell differentiation. Finally, we show that imposed expression of Eomes after acute viral infection promotes some features of exhaustion but must act in concert with other factors during chronic viral infection to establish all hallmarks of exhaustion. In summary, our results clearly underline the importance of T-bet in guiding canonical CTL development during acute viral infections. According to the World Health Organization infectious diseases kill over 17 million people per year. At the same time highly infectious viral diseases, such as Ebola and COVID-19 that are lacking specific treatments, are emerging to pose additional threats. It is therefore pivotal to precisely understand how our immune system responds towards pathogens to develop new treatment options. Here we have investigated the role of two related molecules, named T-bet and Eomes, that guide the development and function of lymphocytes in their fight against intracellular pathogens. We specifically focused on cytotoxic lymphocytes as these cells dominate the early phase of viral containment. We show that T-bet is essential for the expansion of cytotoxic lymphocytes and equip lymphocytes with the ability to efficiently eliminate virus-infected cells. Hence, our study provides new insights into the importance and specific actions of T-bet during acute viral infections and how this might be harnessed for future therapeutic interventions.
Collapse
|
12
|
Krylatov AV, Tsibulnikov SY, Mukhomedzyanov AV, Boshchenko AA, Goldberg VE, Jaggi AS, Erben RG, Maslov LN. The Role of Natriuretic Peptides in the Regulation of Cardiac Tolerance to Ischemia/Reperfusion and Postinfarction Heart Remodeling. J Cardiovasc Pharmacol Ther 2020; 26:131-148. [PMID: 32840121 DOI: 10.1177/1074248420952243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past 10 years, mortality from acute myocardial infarction has not decreased despite the widespread introduction of percutaneous coronary intervention. The reason for this situation is the absence in clinical practice of drugs capable of preventing reperfusion injury of the heart with high efficiency. In this regard, noteworthy natriuretic peptides (NPs) which have the infarct-limiting effect, prevent reperfusion cardiac injury, prevent adverse post-infarction remodeling of the heart. Atrial natriuretic peptide does not have the infarct-reducing effect in rats with alloxan-induced diabetes mellitus. NPs have the anti-apoptotic and anti-inflammatory effects. There is indirect evidence that NPs inhibit pyroptosis and autophagy. Published data indicate that NPs inhibit reactive oxygen species production in cardiomyocytes, aorta, heart, kidney and the endothelial cells. NPs can suppress aldosterone, angiotensin II, endothelin-1 synthesize and secretion. NPs inhibit the effects aldosterone, angiotensin II on the post-receptor level through intracellular signaling events. NPs activate guanylyl cyclase, protein kinase G and protein kinase A, and reduce phosphodiesterase 3 activity. NO-synthase and soluble guanylyl cyclase are involved in the cardioprotective effect of NPs. The cardioprotective effect of natriuretic peptides is mediated via activation of kinases (AMPK, PKC, PI3 K, ERK1/2, p70s6 k, Akt) and inhibition of glycogen synthase kinase 3β. The cardioprotective effect of NPs is mediated via sarcolemmal KATP channel and mitochondrial KATP channel opening. The cardioprotective effect of brain natriuretic peptide is mediated via MPT pore closing. The anti-fibrotic effect of NPs may be mediated through inhibition TGF-β1 expression. Natriuretic peptides can inhibit NF-κB activity and activate GATA. Hemeoxygenase-1 and peroxisome proliferator-activated receptor γ may be involved in the infarct-reducing effect of NPs. NPs exhibit the infarct-limiting effect in patients with acute myocardial infarction. NPs prevent post-infarction remodeling of the heart. To finally resolve the question of the feasibility of using NPs in AMI, a multicenter, randomized, blind, placebo-controlled study is needed to assess the effect of NPs on the mortality of patients after AMI.
Collapse
Affiliation(s)
- Andrey V Krylatov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Sergey Y Tsibulnikov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | | | - Alla A Boshchenko
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Victor E Goldberg
- Cancer Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Amteshwar S Jaggi
- 429174Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Reinhold G Erben
- Department of Biomedical Research, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Leonid N Maslov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| |
Collapse
|
13
|
Stokic-Trtica V, Diefenbach A, Klose CSN. NK Cell Development in Times of Innate Lymphoid Cell Diversity. Front Immunol 2020; 11:813. [PMID: 32733432 PMCID: PMC7360798 DOI: 10.3389/fimmu.2020.00813] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
After being described in the 1970s as cytotoxic cells that do not require MHC-dependent pre-activation, natural killer (NK) cells remained the sole member of innate lymphocytes for decades until lymphoid tissue-inducer cells in the 1990s and helper-like innate lymphoid lineages from 2008 onward completed the picture of innate lymphoid cell (ILC) diversity. Since some of the ILC members, such as ILC1s and CCR6- ILC3s, share specific markers previously used to identify NK cells, these findings provoked the question of how to delineate the development of NK cell and helper-like ILCs and how to properly identify and genetically interfere with NK cells. The description of eomesodermin (EOMES) as a lineage-specifying transcription factor of NK cells provided a candidate that may serve as a selective marker for the genetic targeting and identification of NK cells. Unlike helper-like ILCs, NK cell activation is, to a large degree, regulated by the engagement of activating and inhibitory surface receptors. NK cell research has revealed some elegant mechanisms of immunosurveillance, coined "missing-self" and "induced-self" recognition, thus complementing "non-self recognition", which is predominantly utilized by adaptive lymphocytes and myeloid cells. Notably, the balance of activating and inhibitory signals perceived by surface receptors can be therapeutically harnessed for anti-tumor immunity mediated by NK cells. This review aims to summarize the similarities and the differences in development, function, localization, and phenotype of NK cells and helper-like ILCs, with the purpose to highlight the unique feature of NK cell development and regulation.
Collapse
Affiliation(s)
- Vladislava Stokic-Trtica
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Max-Planck Institute for Infection Biology, Berlin, Germany
| | - Andreas Diefenbach
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
[Expression and clinical significance of runt-related transcription factor 3 in children with bronchiolitis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21. [PMID: 31642435 PMCID: PMC7389732 DOI: 10.7499/j.issn.1008-8830.2019.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To study the mRNA level of runt-related transcription factor 3 (RUNX3) in children with bronchiolitis and its clinical significance in bronchiolitis. METHODS A total of 54 young children with bronchiolitis were enrolled as the bronchiolitis group, among whom 28 with atopic constitution were enrolled in the atopic bronchiolitis group and 26 with non-atopic constitution were enrolled in the non-atopic bronchiolitis group. A total of 48 healthy young children were enrolled as the healthy control group, among whom 24 with atopic constitution were enrolled in the atopic healthy control group and 24 with non-atopic constitution were enrolled in the non-atopic healthy control group. Quantitative real-time PCR was used to measure the mRNA level of RUNX3 in peripheral blood mononuclear cells. ELISA was used to measure the serum levels of interleukin-4 (IL-4) and interferon gamma (IFN-γ). RESULTS The bronchiolitis group had a significantly lower mRNA level of RUNX3 than the healthy control group, and the atopic bronchiolitis group had a significantly lower mRNA level of RUNX3 than the non-atopic bronchiolitis, atopic healthy control, and non-atopic healthy control groups (P<0.05). The bronchiolitis group had a significantly higher serum level of IL-4 than the healthy control group, and the atopic bronchiolitis group had a significantly higher serum level of IL-4 than the non-atopic healthy control group (P<0.05). The bronchiolitis group had a significantly lower serum level of IFN-γ than the healthy control group, and the atopic bronchiolitis group had a significantly lower serum level of IFN-γ than the non-atopic bronchiolitis, atopic healthy control, and non-atopic healthy control groups (P<0.05). The correlation analysis showed that the mRNA level of RUNX3 was negatively correlated with the serum level of IL-4 and was positively correlated with the serum level of IFN-γ (P<0.05). CONCLUSIONS Measurement of RUNX3 gene expression in peripheral blood mononuclear cells has a certain value in identifying children with atopic constitution at high risk of asthma among children with bronchiolitis.
Collapse
|
15
|
Liu W, Yue S, Zheng X, Hu M, Cao J, Zheng Y. aFARP-ChIP-seq, a convenient and reliable method for genome profiling in as few as 100 cells with a capability for multiplexing ChIP-seq. Epigenetics 2019; 14:877-893. [PMID: 31169445 PMCID: PMC6691993 DOI: 10.1080/15592294.2019.1621139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/04/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022] Open
Abstract
Much effort has been devoted to understand how chromatin modification regulates development and disease. Despite recent progress, however, it remains difficult to obtain high-quality epigenomic maps using chromatin-immunoprecipitation-coupled deep sequencing (ChIP-seq) in samples with low-cell numbers. Here, we present an Atlantis dsDNase-based technology, aFARP-ChIP-seq, that provides accurate profiling of genome-wide histone modifications in as few as 100 cells. By mapping histone lysine trimethylation (H3K4me3) and acetylation (H3K27Ac) in group I innate lymphoid cells (ILC1) sorted from different tissues in parallel, aFARP-ChIP-seq uncovers putative active promoter and enhancer landscapes of several tissue-specific Natural Killer cells (NK) and ILC1. aFARP-ChIP-seq is also highly effective in mapping transcription factor binding sites in small number of cells. Thus, aFARP-ChIP-seq offers multiplexing mapping of both epigenome and transcription factor binding sites using a small number of cells.
Collapse
Affiliation(s)
- Wenbin Liu
- Department of Embryology, Carnegie Institution for Science Baltimore, Baltimore, MD, USA
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Sibiao Yue
- Department of Embryology, Carnegie Institution for Science Baltimore, Baltimore, MD, USA
| | - Xiaobin Zheng
- Department of Embryology, Carnegie Institution for Science Baltimore, Baltimore, MD, USA
| | - Minjie Hu
- Department of Embryology, Carnegie Institution for Science Baltimore, Baltimore, MD, USA
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, PR China
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science Baltimore, Baltimore, MD, USA
| |
Collapse
|
16
|
Human IL-23R Cytokine-Binding Homology Region-Fc Fusion Protein Ameliorates Psoriasis via the Decrease of Systemic Th17 and ILC3 Cell Responses. Int J Mol Sci 2019; 20:ijms20174170. [PMID: 31454926 PMCID: PMC6747249 DOI: 10.3390/ijms20174170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 11/21/2022] Open
Abstract
Interleukin (IL)-23 is considered an effective therapeutic target for the treatment of psoriasis because of the crucial role of the IL-23/IL-17 axis in the pathogenesis of psoriasis, and it has recently been reported to be involved in ILC3 cell differentiation. In this study, we report that eukaryotically expressed rhIL23R-CHR/Fc, as an endogenous extracellular receptor analogue, could be a natural antagonist in an imiquimod (IMQ)-induced psoriasis-like mouse model, including the antagonizing effect of suppressed inflammation in the skin lesion, decreased production of pro-inflammatory cells, and reduced the expression of pro-inflammatory factors. The rhIL23R-CHR/Fc fusion protein inhibits both innate immune and adaptive immune-mediated inflammatory responses. These findings shed light on rhIL23R-CHR/Fc as a promising candidate therapy for the treatment of psoriasis.
Collapse
|
17
|
Jetten AM, Cook DN. (Inverse) Agonists of Retinoic Acid-Related Orphan Receptor γ: Regulation of Immune Responses, Inflammation, and Autoimmune Disease. Annu Rev Pharmacol Toxicol 2019; 60:371-390. [PMID: 31386594 DOI: 10.1146/annurev-pharmtox-010919-023711] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Retinoic acid-related orphan receptor γt (RORγt) functions as a ligand-dependent transcription factor that regulates multiple proinflammatory genes and plays a critical role in several inflammatory and autoimmune diseases. Various endogenous and synthetic RORγ (inverse) agonists have been identified that regulate RORγ transcriptional activity, including many cholesterol intermediates and oxysterols. Changes in cholesterol biosynthesis and metabolism can therefore have a significant impact on the generation of oxysterol RORγ ligands and, consequently, can control RORγt activity and inflammation. These observations contribute to a growing literature that connects cholesterol metabolism to the regulation of immune responses and autoimmune disease. Loss of RORγ function in knockout mice and in mice treated with RORγ inverse agonists results in reduced production of proinflammatory cytokines, such as IL-17A/F, and increased resistance to autoimmune disease in several experimental rodent models. Thus, RORγt inverse agonists might provide an attractive therapeutic approach to treat a variety of autoimmune diseases.
Collapse
Affiliation(s)
- Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA;
| | - Donald N Cook
- Immunogenetics Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
18
|
Branzk N, Gronke K, Diefenbach A. Innate lymphoid cells, mediators of tissue homeostasis, adaptation and disease tolerance. Immunol Rev 2019; 286:86-101. [PMID: 30294961 DOI: 10.1111/imr.12718] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/05/2018] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILC) are a recently identified group of tissue-resident innate lymphocytes. Available data support the view that ILC or their progenitors are deposited and retained in tissues early during ontogeny. Thereby, ILC become an integral cellular component of tissues and organs. Here, we will review the intriguing relationships between ILC and basic developmental and homeostatic processes within tissues. Studying ILC has already led to the appreciation of the integral roles of immune cells in tissue homeostasis, morphogenesis, metabolism, regeneration, and growth. This area of immunology has not yet been studied in-depth but is likely to reveal important networks contributing to disease tolerance and may be harnessed for future therapeutic approaches.
Collapse
Affiliation(s)
- Nora Branzk
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Konrad Gronke
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
19
|
Martinez-Gonzalez I, Ghaedi M, Steer CA, Mathä L, Vivier E, Takei F. ILC2 memory: Recollection of previous activation. Immunol Rev 2019; 283:41-53. [PMID: 29664572 DOI: 10.1111/imr.12643] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immunological memory, traditionally thought to belong to T and B cells, has now been extended to innate lymphocytes, including NK cells and ILC2s, myeloid cells such as macrophages, also termed "trained immunity" and more recently to epithelial stem cells. In this review, we discuss the mechanisms underlying memory generation on ILC2s and speculate about their potential role in human allergic diseases, such as asthma. Moreover, we examine the relevance of the spontaneous ILC2 activation in the lung during the neonatal period in order to efficiently respond to stimuli later in life. These "training" of neonatal ILC2s may have an impact on the generation of memory ILC2s in the adulthood.
Collapse
Affiliation(s)
- Itziar Martinez-Gonzalez
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Terry Fox Laboratory British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Maryam Ghaedi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Terry Fox Laboratory British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Catherine A Steer
- Terry Fox Laboratory British Columbia Cancer Agency, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Laura Mathä
- Terry Fox Laboratory British Columbia Cancer Agency, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Eric Vivier
- Centre d' Immunologie de Marseille-Luminy, Université d'Aix-Marseille, INSERM, CNRS, Marseille, France.,Innate Pharma Research Labs., Innate Pharma, Marseille, France
| | - Fumio Takei
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Terry Fox Laboratory British Columbia Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
20
|
Yu Y, Wang L, Gu G. The correlation between Runx3 and bronchial asthma. Clin Chim Acta 2018; 487:75-79. [PMID: 30218658 DOI: 10.1016/j.cca.2018.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
Abstract
Runx3, a member of the Runt-related transcription factor family, has attracted extensive attention due to its important role in the development of immune systems, especially in the differentiation of T cells. Accumulated evidence indicated that altered expression of Runx3 regulates a variety of target genes in different tissues/cells. Studies in animal models suggested that Runx3 may regulate the development of T cell lineage including those of innate lymphoid cells, Treg cells and dendritic cells, which may contribute to the development of hypersensitivity and asthma. Specifically, Runx3 modulates Th1/Th2 balance and hence, the production of interleukins, which induce inflammatory responses. Understanding the roles and mechanisms of Runx3 in the regulation of immune function provides a basis for the design of novel preventive and treatment models for bronchial asthma. This article reviews published data from cell lines, animal models, and patients, concerning the relationship between Runx3 expression alteration and asthma. Epigenetic regulation of Runx3 by DNA hypermethylation and microRNA, and the implication of these pathways in asthma are also discussed.
Collapse
Affiliation(s)
- Yanyan Yu
- The children's hospital affiliated of Suzhou University, Suzhou 215000, Jiangsu Province, China.
| | - Leilei Wang
- Children Asthma Department, Lianyungang Maternal and Child Hospital Jiangsu Province, Lianyungang 222006, Jiangsu Province, China
| | - Guixiong Gu
- The children's hospital affiliated of Suzhou University, Suzhou 215000, Jiangsu Province, China.
| |
Collapse
|
21
|
Dai Q, Li Y, Yu H, Wang X. Suppression of Th1 and Th17 Responses and Induction of Treg Responses by IL-18-Expressing Plasmid Gene Combined with IL-4 on Collagen-Induced Arthritis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5164715. [PMID: 29854762 PMCID: PMC5964485 DOI: 10.1155/2018/5164715] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/22/2018] [Accepted: 03/29/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES IL-18 is a proinflammatory cytokine with multiple immunoregulatory properties. We studied the effect of IL-18 gene therapy on the development of murine collagen-induced arthritis (CIA). METHODS Plasmid pCAGGS-IL-18 along or in combination with IL-10 or IL-4 was administered to CIA mice. The incidence and severity of arthritis of the paws were determined by a visual scale. Joint destruction was determined by histology. The levels of a panel of cytokines and transcription factors in the synovium were determined by reverse transcription polymerase chain reaction and quantitative RT-PCR. Quantitative RT-PCR was employed to detect the mRNA expression of TLRs and their pathway on the surface of DCs. RESULTS IL-18 gene therapy had no therapeutic effect on CIA mice. Additional coadministration with low dosage of recombinant IL-4 ameliorated the disease progression. Histopathological examination of the joints showed intact cartilage surface in IL-18 gene combined with IL-4-treated mice. The synovium of IL-18 gene combined with rIL4-treated mice had lower expression of TNF-α, IFN-γ, and IL-17 and higher expression of IL-10. The mechanism of this response appeared to involve modulation of transcription factors FoxP3 and GATA-3. The DCs in the spleen and lymph nodes of IL-18 gene combined with rIL4-treated mice had lower expression of TLR2, MyD88, and NF-kB. CONCLUSIONS Our findings indicate that pIL-18 gene combined with IL-4 ameliorates arthritis in the CIA mouse by suppression of Th1 and Th17 cytokines and increasing expression of FoxP3 and GATA-3. The plasmid backbone and multiple immunoregulatory properties of IL-18 appear to play a major role in the pIL-18 coadministration with rIL-4-mediated immunomodulation of arthritis through blocking the TLR2/MyD88/NF-kappa B signaling pathway.
Collapse
Affiliation(s)
- Qiaomei Dai
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pathology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yang Li
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haiyue Yu
- Department of Rheumatology, Qiqihar First Hospital, Qiqihar, China
| | - Xiaoyan Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
22
|
Sciumè G, Shih HY, Mikami Y, O'Shea JJ. Epigenomic Views of Innate Lymphoid Cells. Front Immunol 2017; 8:1579. [PMID: 29250060 PMCID: PMC5715337 DOI: 10.3389/fimmu.2017.01579] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/02/2017] [Indexed: 12/30/2022] Open
Abstract
The discovery of innate lymphoid cells (ILCs) with selective production of cytokines typically attributed to subsets of T helper cells forces immunologists to reassess the mechanisms by which selective effector functions arise. The parallelism between ILCs and T cells extends beyond these two cell types and comprises other innate-like T lymphocytes. Beyond the recognition of specialized effector functionalities in diverse lymphocytes, features typical of T cells, such as plasticity and memory, are also relevant for innate lymphocytes. Herein, we review what we have learned in terms of the molecular mechanisms underlying these shared functions, focusing on insights provided by next generation sequencing technologies. We review data on the role of lineage-defining- and signal-dependent transcription factors (TFs). ILC regulomes emerge developmentally whereas the much of the open chromatin regions of T cells are generated acutely, in an activation-dependent manner. And yet, these regions of open chromatin in T cells and ILCs have remarkable overlaps, suggesting that though accessibility is acquired by distinct modes, the end result is that convergent signaling pathways may be involved. Although much is left to be learned, substantial progress has been made in understanding how TFs and epigenomic status contribute to ILC biology in terms of differentiation, specification, and plasticity.
Collapse
Affiliation(s)
- Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Han-Yu Shih
- Lymphocyte and Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, United States
| | - Yohei Mikami
- Lymphocyte and Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, United States
| | - John J O'Shea
- Lymphocyte and Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, United States
| |
Collapse
|
23
|
Mora-Velandia LM, Castro-Escamilla O, Méndez AG, Aguilar-Flores C, Velázquez-Avila M, Tussié-Luna MI, Téllez-Sosa J, Maldonado-García C, Jurado-Santacruz F, Ferat-Osorio E, Martínez-Barnetche J, Pelayo R, Bonifaz LC. A Human Lin - CD123 + CD127 low Population Endowed with ILC Features and Migratory Capabilities Contributes to Immunopathological Hallmarks of Psoriasis. Front Immunol 2017; 8:176. [PMID: 28303135 PMCID: PMC5332395 DOI: 10.3389/fimmu.2017.00176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Innate lymphoid cells (ILC) are members of a heterogeneous family with a lymphoid origin that mimics the T helper (Th) cytokine profile. ILC are involved in early effector cytokine-mediated responses during infections in peripheral tissues. ILC also play an important role in chronic skin inflammatory diseases, including psoriasis. Although classical ILC express CD127, it has been recently reported that the presence of non-classical CD127- ILC populations and an early ILC precursor (EILP) CD127low. ILC development has predominately been investigated in mouse models. However, in humans, different transcription factors have been described for ILC identification. NFIL3 (nuclear factor, IL-3 regulated) is crucial for ILC development in response to IL-7. CD123 (IL-3Rα) is usually used to exclude basophils during ILC identification, however, it is unknown if in response to IL-3, NFIL3 could be relevant to induce ILC features in Lin- CD123+ populations in addition, is also unknown whether peripheral blood (PB) population with ILC features may have skin-homing potential to participate in skin inflammatory chronic diseases. Here, we report a Lin- CD123+ CD127low CD7+ CLA+ population that share some phenotypic properties with basophils, but expresses several transcription factors for ILC commitment such as inhibitor of DNA binding 2 (Id2), NFIL3, promyelocytic leukemia zinc finger (PLZF), thymocyte selection-associated high-mobility group box protein (TOX), and T cell factor-1 (TCF-1). In addition, this population expresses different ILC markers: CD132, CD90, CD161, α4 integrin, c-Kit, CRTH2, AhR, and IL-23R. IL-3 prevents apoptosis and increases their NFIL3, TOX, and PLZF expression. In PB, the CD123+ CD127low population is predominantly a conspicuous population that expresses T-bet and RORγt. The Lin- CD123+ CD127low population in PB has a limited Th type cytokine expression and highly expresses IL-8. The Lin- CD123+ CD127low population expresses skin-homing receptors (cutaneous lymphocyte antigen and CXCR4) and transmigrates through endothelial cells in response to SDF-1. An equivalent Lin- CD123low population was identified in control skin, which shows a broader phenotypic diversity and cytokine production, including IL-22 and IL-17. Remarkably, the CD123low population in the lesion and non-lesion skin of psoriasis patients expresses IL-17 and IL-22. Our findings suggest the identification of an alternative Lin- CD123+ CD127low population with ILC features endowed with migratory capabilities that might contribute to immunopathological hallmarks of psoriasis.
Collapse
Affiliation(s)
- Luz María Mora-Velandia
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Octavio Castro-Escamilla
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Andrés González Méndez
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Cristina Aguilar-Flores
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Martha Velázquez-Avila
- Unidad de Investigación en Enfermedades Oncológicas, Hospital de Oncología, Centro Médico Nacional 'Siglo XXI' , Mexico City , Mexico
| | - María Isabel Tussié-Luna
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico; Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Juan Téllez-Sosa
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos , Mexico
| | - César Maldonado-García
- Centro Dermatológico "Dr. Ladislao de la Pascua", Secretaria de Salud de la Ciudad de México , Mexico City , Mexico
| | - Fermín Jurado-Santacruz
- Centro Dermatológico "Dr. Ladislao de la Pascua", Secretaria de Salud de la Ciudad de México , Mexico City , Mexico
| | - Eduardo Ferat-Osorio
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Jesus Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos , Mexico
| | - Rosana Pelayo
- Unidad de Investigación en Enfermedades Oncológicas, Hospital de Oncología, Centro Médico Nacional 'Siglo XXI' , Mexico City , Mexico
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| |
Collapse
|
24
|
Saez de Guinoa J, Jimeno R, Farhadi N, Jervis PJ, Cox LR, Besra GS, Barral P. CD1d-mediated activation of group 3 innate lymphoid cells drives IL-22 production. EMBO Rep 2017; 18:39-47. [PMID: 27799287 PMCID: PMC5210076 DOI: 10.15252/embr.201642412] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/07/2016] [Accepted: 10/04/2016] [Indexed: 12/21/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous family of immune cells that play a critical role in a variety of immune processes including host defence against infection, wound healing and tissue repair. Whether these cells are involved in lipid-dependent immunity remains unexplored. Here we show that murine ILCs from a variety of tissues express the lipid-presenting molecule CD1d, with group 3 ILCs (ILC3s) showing the highest level of expression. Within the ILC3 family, natural cytotoxicity triggering receptor (NCR)-CCR6+ cells displayed the highest levels of CD1d. Expression of CD1d on ILCs is functionally relevant as ILC3s can acquire lipids in vitro and in vivo and load lipids on CD1d to mediate presentation to the T-cell receptor of invariant natural killer T (iNKT) cells. Conversely, engagement of CD1d in vitro and administration of lipid antigen in vivo induce ILC3 activation and production of IL-22. Taken together, our data expose a previously unappreciated role for ILCs in CD1d-mediated immunity, which can modulate tissue homeostasis and inflammatory responses.
Collapse
Affiliation(s)
| | - Rebeca Jimeno
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Nazanin Farhadi
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Peter J Jervis
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Liam R Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, UK
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| |
Collapse
|
25
|
Yang D, Yang W, Tian Z, van Velkinburgh JC, Song J, Wu Y, Ni B. Innate lymphoid cells as novel regulators of obesity and its-associated metabolic dysfunction. Obes Rev 2016; 17:485-98. [PMID: 26948388 DOI: 10.1111/obr.12397] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/08/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
Abstract
The increased prevalence of obesity worldwide has been accompanied by increases in risk and rates of obesity-associated metabolic dysfunctions, such as insulin resistance. The chronic, low-grade inflammatory condition of obesity highlights the pathophysiological link between the immune system and the metabolic system, which has yet to be fully understood. Recent studies of obesity have started to uncover potential regulatory roles for the innate lymphoid cells (ILCs), which under normal conditions serve to regulate development of lymphoid tissue and function of the mucosal immune system. The ILCs are a newly identified immune cell population with complicated composition and subsequently diverse and dynamic functions. Studies to determine the distribution profile of the various ILCs in adipose tissue provide intriguing clues as to their regulatory capacity in obesity and its associated metabolic dysfunctions. Here, we review the recent findings supporting a role for ILCs as regulators of obesity or its associated insulin resistance, and discuss the potential underlying molecular mechanism as well as its promise as a therapeutic target for clinical applications. © 2016 World Obesity.
Collapse
Affiliation(s)
- D Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - W Yang
- Department of Dermatology, 181st Hospital of PLA, Guilin, China
| | - Z Tian
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - J C van Velkinburgh
- van Velkinburgh Initiative for Collaboratory BioMedical Research, Santa Fe, NM, USA
| | - J Song
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Y Wu
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - B Ni
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| |
Collapse
|
26
|
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: 1.6] [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
|
27
|
Allan DSJ, Kirkham CL, Aguilar OA, Qu LC, Chen P, Fine JH, Serra P, Awong G, Gommerman JL, Zúñiga-Pflücker JC, Carlyle JR. An in vitro model of innate lymphoid cell function and differentiation. Mucosal Immunol 2015; 8:340-51. [PMID: 25138665 DOI: 10.1038/mi.2014.71] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/28/2014] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILC) are RAG-independent lymphocytes with important roles in innate immunity, and include group-1 (natural killer (NK) cell, ILC1), group-2 (ILC2), and group-3 (lymphoid tissue inducer (LTi), NCR(+) ILC3) subsets. Group-3 ILC express Rorγt, produce interleukin (IL)-22, and are critically important in the normal function of mucosal tissues. Here, we describe a novel model cell line for the study of ILC function and differentiation. The parental MNK cell line, derived from NKR-P1B(+) fetal thymocytes, shows a capacity to differentiate in γc cytokines. One IL-7-responsive subline, designated MNK-3, expresses Rorγt and produces high levels of IL-22 in response to IL-23 and IL-1β stimulation. MNK-3 cells display surface markers and transcript expression characteristic of group-3 ILC, including IL-7Rα (CD127), c-kit (CD117), CCR6, Thy1 (CD90), RANK, RANKL, and lymphotoxin (LTα1β2). Using an in vitro assay of LTi cell activity, MNK-3 cells induce ICAM-1 and VCAM-1 expression on stromal cells in a manner dependent upon LTα1β2 expression. A second IL-2-responsive subline, MNK-1, expresses several NK cell receptors, perforin and granzymes, and shows some cytotoxic activity. Thus, MNK-1 cells serve as a model of ILC1/NK development and differentiation, whereas MNK-3 cells provide an attractive in vitro system to study the function of ILC3/LTi cells.
Collapse
Affiliation(s)
- D S J Allan
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - C L Kirkham
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - O A Aguilar
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - L C Qu
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - P Chen
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J H Fine
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - P Serra
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - G Awong
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J L Gommerman
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - J C Zúñiga-Pflücker
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J R Carlyle
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Park SY, Schuller-Levis G, Park E. A novel cysteine sulfinic Acid decarboxylase knock-out mouse: immune function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 803:89-98. [PMID: 25833490 DOI: 10.1007/978-3-319-15126-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Seung Yong Park
- School of Veterinary Medicine, Konkuk University, Seoul, Korea
| | | | | |
Collapse
|
29
|
Annunziato F, Romagnani C, Romagnani S. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol 2014; 135:626-35. [PMID: 25528359 DOI: 10.1016/j.jaci.2014.11.001] [Citation(s) in RCA: 536] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/27/2014] [Accepted: 11/04/2014] [Indexed: 02/07/2023]
Abstract
The immune system has tailored its effector functions to optimally respond to distinct species of microbes. Based on emerging knowledge on the different effector T-cell and innate lymphoid cell (ILC) lineages, it is clear that the innate and adaptive immune systems converge into 3 major kinds of cell-mediated effector immunity, which we propose to categorize as type 1, type 2, and type 3. Type 1 immunity consists of T-bet(+) IFN-γ-producing group 1 ILCs (ILC1 and natural killer cells), CD8(+) cytotoxic T cells (TC1), and CD4(+) TH1 cells, which protect against intracellular microbes through activation of mononuclear phagocytes. Type 2 immunity consists of GATA-3(+) ILC2s, TC2 cells, and TH2 cells producing IL-4, IL-5, and IL-13, which induce mast cell, basophil, and eosinophil activation, as well as IgE antibody production, thus protecting against helminthes and venoms. Type 3 immunity is mediated by retinoic acid-related orphan receptor γt(+) ILC3s, TC17 cells, and TH17 cells producing IL-17, IL-22, or both, which activate mononuclear phagocytes but also recruit neutrophils and induce epithelial antimicrobial responses, thus protecting against extracellular bacteria and fungi. On the other hand, type 1 and 3 immunity mediate autoimmune diseases, whereas type 2 responses can cause allergic diseases.
Collapse
Affiliation(s)
- Francesco Annunziato
- Department of Experimental and Clinical Medicine and the DENOTHE Center, University of Florence, Florence, Italy
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma Forschungszentrum, Berlin, Germany
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and the DENOTHE Center, University of Florence, Florence, Italy.
| |
Collapse
|
30
|
Diefenbach A, Colonna M, Koyasu S. Development, differentiation, and diversity of innate lymphoid cells. Immunity 2014; 41:354-365. [PMID: 25238093 DOI: 10.1016/j.immuni.2014.09.005] [Citation(s) in RCA: 440] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Indexed: 01/11/2023]
Abstract
Recent years have witnessed the discovery of an unprecedented complexity in innate lymphocyte lineages, now collectively referred to as innate lymphoid cells (ILCs). ILCs are preferentially located at barrier surfaces and are important for protection against pathogens and for the maintenance of organ homeostasis. Inappropriate activation of ILCs has been linked to the pathogenesis of inflammatory and autoimmune disorders. Recent evidence suggests that ILCs can be grouped into two separate lineages, cytotoxic ILCs represented by conventional natural killer (cNK) cells and cytokine-producing helper-like ILCs (i.e., ILC1s, ILC2s, ILC3s). We will focus here on current work in humans and mice that has identified core transcriptional circuitry required for the commitment of lymphoid progenitors to the ILC lineage. The striking similarities in transcriptional control of ILC and T cell lineages reveal important insights into the evolution of transcriptional programs required to protect multicellular organisms against infections and to fortify barrier surfaces.
Collapse
Affiliation(s)
- Andreas Diefenbach
- Research Centre for Immunology and Immunotherapy, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany; Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S. Euclid, St. Louis, MO 63110, USA
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Research Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
31
|
Yu X, Wang Y, Deng M, Li Y, Ruhn KA, Zhang CC, Hooper LV. The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor. eLife 2014; 3. [PMID: 25310240 PMCID: PMC4356142 DOI: 10.7554/elife.04406] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/10/2014] [Indexed: 12/21/2022] Open
Abstract
Innate lymphoid cells (ILCs) are recently identified lymphocytes that limit infection and promote tissue repair at mucosal surfaces. However, the pathways underlying ILC development remain unclear. Here we show that the transcription factor NFIL3 directs the development of a committed bone marrow precursor that differentiates into all known ILC lineages. NFIL3 was required in the common lymphoid progenitor (CLP), and was essential for the differentiation of αLP, a bone marrow cell population that gives rise to all known ILC lineages. Clonal differentiation studies revealed that CXCR6(+) cells within the αLP population differentiate into all ILC lineages but not T- and B-cells. We further show that NFIL3 governs ILC development by directly regulating expression of the transcription factor TOX. These findings establish that NFIL3 directs the differentiation of a committed ILC precursor that gives rise to all ILC lineages and provide insight into the defining role of NFIL3 in ILC development.
Collapse
Affiliation(s)
- Xiaofei Yu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yuhao Wang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Mi Deng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yun Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kelly A Ruhn
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
32
|
Abstract
Innate lymphoid cells (ILCs) are a recently appreciated immune cell population that is constitutively found in the healthy mammalian gastrointestinal (GI) tract and associated lymphoid tissues. Translational studies have revealed that alterations in ILC populations are associated with GI disease in patients, such as inflammatory bowel disease, HIV infection and colon cancer, suggesting a potential role for ILCs in either maintaining intestinal health or promoting intestinal disease. Mouse models identified that ILCs have context-dependent protective and pathologic functions either during the steady state, or following infection, inflammation or tissue damage. This review will discuss the associations of altered intestinal ILCs with human GI diseases, and the functional consequences of targeting ILCs in mouse models. Collectively, our current understanding of ILCs suggests that the development of novel therapeutic strategies to modulate ILC responses will be of significant clinical value to prevent or treat human GI diseases.
Collapse
Affiliation(s)
- Gregory F Sonnenberg
- Division of Gastroenterology, Department of Medicine, and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|