1
|
Parolini C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem Pharmacol 2025; 235:116844. [PMID: 40044049 DOI: 10.1016/j.bcp.2025.116844] [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: 11/28/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Osteoporosis is the most common skeletal disease worldwide, characterized by low bone mineral density, resulting in weaker bones, and an increased risk of fragility fractures. The maintenance of bone mass relies on the precise balance between bone synthesis and resorption. The close relationship between the immune and skeletal systems, called "osteoimmunology", was coined to identify these overlapping "scientific worlds", and its function resides in the evaluation of the mutual effects of the skeletal and immune systems at the molecular and cellular levels, in both physiological and pathological states. Lipids play an essential role in skeletal metabolism and bone health. Indeed, bone marrow and its skeletal components demand a dramatic amount of daily energy to control hematopoietic turnover, acquire and maintain bone mass, and actively being involved in whole-body metabolism. Statins, the main therapeutic agents in lowering plasma cholesterol levels, are able to promote osteoblastogenesis and inhibit osteoclastogenesis. This review is meant to provide an updated overview of the pathophysiology of bone remodelling cycle, focusing on the interplay between bone, immune system and lipids. Novel therapeutic strategies for the management of osteoporosis are also discussed.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti', via Balzaretti 9 - Università degli Studi di Milano 20133 Milano, Italy.
| |
Collapse
|
2
|
Shirley S, Ichise H, Di Natale V, Jin J, Wu C, Zou R, Zhang W, Fang Y, Zhang Y, Chen M, Peng S, Basu U, Que J, Huang Y. A vasculature-resident innate lymphoid cell population in mouse lungs. Nat Commun 2025; 16:3718. [PMID: 40253407 PMCID: PMC12009297 DOI: 10.1038/s41467-025-58982-1] [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: 09/29/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Tissue-resident immune cells such as innate lymphoid cells (ILC) are known to reside in the parenchymal compartments of tissues and modulate local immune protection. Here we use intravascular cell labeling, parabiosis and multiplex 3D imaging to identify a population of group 3 ILCs in mice that are present within the intravascular space of lung blood vessels (vILC3). vILC3s are distributed broadly in alveolar capillary beds from which inhaled pathogens enter the lung parenchyma. By contrast, conventional ILC3s in tissue parenchyma are enriched in lymphoid clusters in proximity to large veins. In a mouse model of pneumonia, Pseudomonas aeruginosa infection results in rapid vILC3 expansion and production of chemokines including CCL4. Blocking CCL4 in vivo attenuates neutrophil recruitment to the lung at the early stage of infection, resulting in prolonged inflammation and delayed bacterial clearance. Our findings thus define the intravascular space as a site of ILC residence in mice, and reveal a unique immune cell population that interfaces with tissue alarmins and the circulating immune system for timely host defense.
Collapse
Affiliation(s)
- Simon Shirley
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Vincenzo Di Natale
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jiacheng Jin
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Christine Wu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Raymond Zou
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Wanwei Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yinshan Fang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA
| | - Yingyu Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miao Chen
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Sophia Peng
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Uttiya Basu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA.
| | - Yuefeng Huang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
3
|
Nouari W, Aribi M. Innate lymphoid cells, immune functional dynamics, epithelial parallels, and therapeutic frontiers in infections. Int Rev Immunol 2025:1-28. [PMID: 40242974 DOI: 10.1080/08830185.2025.2490233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Innate lymphoid cells (ILCs) have emerged as pivotal players in the field of immunology, expanding our understanding of innate immunity beyond conventional paradigms. This comprehensive review delves into the multifaceted world of ILCs, beginning with their serendipitous discovery and traversing their ontogeny and heterogeneity. We explore the distinct subsets of ILCs unraveling their intriguing plasticity, which adds a layer of complexity to their functional repertoire. As we journey through the functional activities of ILCs, we address their role in immune responses against various infections, categorizing their interactions with helminthic parasites, bacterial pathogens, fungal infections, and viral invaders. Notably, this review offers a detailed examination of ILCs in the context of specific infections, such as Mycobacterium tuberculosis, Citrobacter rodentium, Clostridium difficile, Salmonella typhimurium, Helicobacter pylori, Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Influenza virus, Cytomegalovirus, Herpes simplex virus, and severe acute respiratory syndrome coronavirus 2. This selection aimed for a comprehensive exploration of ILCs in various infectious contexts, opting for microorganisms based on extensive research findings rather than considerations of virulence or emergence. Furthermore, we raise intriguing questions about the potential immune functional resemblances between ILCs and epithelial cells, shedding light on their interconnectedness within the mucosal microenvironment. The review culminates in a critical assessment of the therapeutic prospects of targeting ILCs during infection, emphasizing their promise as novel immunotherapeutic targets. Nevertheless, due to their recent discovery and evolving understanding, effectively manipulating ILCs is challenging. Ensuring specificity and safety while evaluating long-term effects in clinical settings will be crucial.
Collapse
Affiliation(s)
- Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
4
|
Saini A, Hopkins LS, Serna VA, McCullen MVD, Selner NG, Bhattarai B, Fachi JL, Glynn R, Hayer KE, Bassing CH, Colonna M, Oltz EM. Cell type-specific enhancers regulate IL-22 expression in innate and adaptive lymphoid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646834. [PMID: 40291691 PMCID: PMC12026504 DOI: 10.1101/2025.04.02.646834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
IL-22, a signature cytokine of type 3 lymphoid cells, mediates epithelial homeostasis and protective pathogen responses in barrier tissues, while its deregulated expression drives chronic inflammation associated with colitis and psoriasis. Despite its therapeutic value, little is known about regulatory elements for IL-22 expression. We identify two conserved enhancers, E22-1 and E22-2, which differentially regulate Il22 in type 3 lymphoid subsets. These enhancers are required for steady-state expression of gut antimicrobial peptides, protection from C. rodentium infection, and development of IL-22-mediated psoriasis. E22-1 resembles many known enhancers, functioning in both Th-ILC counterparts. However, E22-2 is only required for IL-22 expression in ILC3s. Its ILC3 restriction relies on multiple Runx3 sites, combined with the lack of a functional RORγt motif, which is present in E22-1. Thus, although responding to similar stimuli, type 3 lymphoid cells use distinct cis-elements for IL-22 expression, with E22-2 likely serving as a homeostatic enhancer in barrier tissues.
Collapse
|
5
|
Fukasawa N, Tsunoda J, Sunaga S, Kiyohara H, Nakamoto N, Teratani T, Mikami Y, Kanai T. The gut-organ axis: Clinical aspects and immune mechanisms. Allergol Int 2025; 74:197-209. [PMID: 39979198 DOI: 10.1016/j.alit.2025.01.004] [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: 12/19/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 02/22/2025] Open
Abstract
The gut-brain axis exemplifies the bidirectional connection between the intestines and the brain, as evidenced by the impact of severe stress on gastrointestinal symptoms including abdominal pain and diarrhea, and conversely, the influence of abdominal discomfort on mood. Clinical observations support the notion of the gut-brain connection, including an increased prevalence of inflammatory bowel disease (IBD) in patients with depression and anxiety, as well as the association of changes in the gut microbiota with neurological disorders such as multiple sclerosis, Parkinson's disease, stroke and Alzheimer's disease. The gut and brain communicate via complex mechanisms involving inflammatory cytokines, immune cells, autonomic nerves, and gut microbiota, which contribute to the pathogenesis in certain gut and brain diseases. Two primary pathways mediate the bidirectional information exchange between the intestinal tract and the brain: signal transduction through bloodstream factors, such as bacterial metabolites and inflammatory cytokines, and neural pathways, such as neurotransmitters and inflammatory cytokines within the autonomic nervous system through the interaction between the nerve cells and beyond. In recent years, the basic mechanisms of the pathophysiology of the gut-brain axis have been gradually elucidated. Beyond the gut-brain interaction, emerging evidence suggests the influence of the gut extends to other organs, such as the liver and lungs, through intricate inter-organ communication pathways. An increasing number of reports on this clinical and basic cross-organ interactions underscore the potential for better understanding and novel therapeutic strategies targeting inter-organs networks. Further clarification of interactions between multiorgans premises transformative insights into cross-organ therapeutic strategies.
Collapse
Affiliation(s)
- Naoto Fukasawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Junya Tsunoda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shogo Sunaga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Kiyohara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
6
|
Lin YH, Wang YH, Peng YJ, Liu FC, Sytwu HK, Cheng CP. Interleukin 26 attenuates osteoblast differentiation in osteoarthritis patients by activating COX2 and NF-κB pathways. Int J Med Sci 2025; 22:1504-1515. [PMID: 40093804 PMCID: PMC11905269 DOI: 10.7150/ijms.102967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Aims: Osteoarthritis (OA) represents the prevailing form of degenerative joint pathology. Recent investigations have revealed a heightened expression of interleukin 26 (IL-26) in various inflammatory arthritic conditions, including OA. However, the specific impacts and functions of IL-26 on osteoblasts (OBs) within the context of OA remain inadequately elucidated. This study aims to clarify the effects and underlying mechanisms of IL-26 by examining its influence on osteoblasts isolated from OA patients and a murine osteoblast cell line. Methods: Human primary osteoblasts and mouse pre-osteoblast cells were subjected to treatment with β-glycerophosphate or concurrent treatment with IL-26 to observe the effects on osteoblast differentiation. The differentiation of osteoblasts was assessed through the expression of relevant genes using reverse transcription-polymerase chain reaction (RT-PCR). Key molecular mechanisms of downstream signaling pathways were examined through immunoblotting assays. Results: Our results reveal that IL-26 mitigates osteoblast differentiation and reduces the expression of the marker alkaline phosphatase. Furthermore, the NF-κB downstream OB proliferated marker iNOS and inhibition OB differentiated marker LCN2 messenger RNA are up-regulated in IL-26 treated group. Also, phosphorylation and nuclear translocation of NF-κB p65 occur following IL-26 stimulation. Additionally, IL-26 enhances the downstream transcription factor cyclooxygenase-2 (COX2), a major player associated with iNOS. STAT1, the canonical receptor signaling pathway of IL-26 is activated. Conclusion: In summary, our findings substantiate the role of IL-26 in osteoarthritis and identify it as a potential therapeutic target for intervention in osteoarthritic pathology.
Collapse
Affiliation(s)
- Yi-Hsuan Lin
- Department and Graduate institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hsun Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Cheng Liu
- Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Pi Cheng
- Department and Graduate institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Rødahl IE, Ivarsson MA, Loh L, Mold JE, Westgren M, Friberg D, Mjösberg J, Björkström NK, Marquardt N, Nixon DF, Michaëlsson J. Distinct Tissue-Dependent Composition and Gene Expression of Human Fetal Innate Lymphoid Cells. Eur J Immunol 2025; 55:e202451150. [PMID: 39676343 PMCID: PMC11830385 DOI: 10.1002/eji.202451150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
The human fetal immune system starts to develop in the first trimester and likely plays a crucial role in fetal development and maternal-fetal tolerance. Innate lymphoid cells (ILCs) are the earliest lymphoid cells to arise in the human fetus. ILCs consist of natural killer (NK) cells, ILC1s, ILC2s, and ILC3s that all share a common lymphoid origin. Here, we studied fetal ILC subsets, mainly NK cells and ILC3s and their potential progenitors, across human fetal tissues. Our results show that fetal ILC subsets have distinct distribution, developmental kinetics, and gene expression profiles across human fetal tissues. Furthermore, we identify CD34+RORγt+Eomes- and CD34+RORγt+Eomes+ cells in the fetal intestine, indicating that tissue-specific ILC progenitors exist already during fetal development.
Collapse
Affiliation(s)
- Inga E. Rødahl
- Center for Infectious MedicineDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Martin A. Ivarsson
- Center for Infectious MedicineDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Liyen Loh
- Division of Experimental MedicineDepartment of MedicineUniversity of California San FranciscoSan FranciscoUSA
| | - Jeff E. Mold
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| | - Magnus Westgren
- Center for Fetal MedicineDepartment of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
| | | | - Jenny Mjösberg
- Center for Infectious MedicineDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
- Clinical Lung‐ and Allergy Research UnitMedical Unit for Lung and Allergy DiseasesKarolinska University Hospital HuddingeStockholmSweden
| | - Niklas K. Björkström
- Center for Infectious MedicineDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Nicole Marquardt
- Center for Hematology and Regenerative MedicineDepartment of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Douglas F. Nixon
- Division of Experimental MedicineDepartment of MedicineUniversity of California San FranciscoSan FranciscoUSA
| | - Jakob Michaëlsson
- Center for Infectious MedicineDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| |
Collapse
|
8
|
Mai C, Fukui A, Saeki S, Takeyama R, Yamaya A, Shibahara H. Expression of NKp46 and other activating inhibitory receptors on uterine endometrial NK cells in females with various reproductive failures: A review. Reprod Med Biol 2025; 24:e12610. [PMID: 39807425 PMCID: PMC11725765 DOI: 10.1002/rmb2.12610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 01/16/2025] Open
Abstract
Background Uterine endometrial natural killer (uNK) cells represent major leukocytes in the mid-secretory phase of the cell cycle, and their number is further increased during early pregnancy. The activating and inhibitory receptors expressed on their surface mediate various functions of uNK cells, such as cytotoxicity, cytokine production, spiral artery remodeling, and self-recognition. Methods This study reviewed the most recent information (PubMed database, 175 articles included) regarding the activating and inhibitory receptors on uNK cells in human females with healthy pregnancies and the evidence indicating their significance in various reproductive failures. Main Findings Numerous studies have indicated that the natural cytotoxic receptors, killer cell immunoglobulin-like receptors, and C-type lectin receptors, particularly those expressed on uNK cells, play crucial roles in successful pregnancy. Conclusion As studies on human uNK cells are limited owing to the low availability of fertile samples, and the extrapolation of animal models has certain limitations, the in vivo role of uNK cells has not yet been fully elucidated. However, immunotherapies focusing on modulating uNK cell function have been controversial in terms of pregnancy outcomes. Further research is required to elucidate the role of uNK cells in reproduction.
Collapse
Affiliation(s)
- Chuxian Mai
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
- Reproductive Medicine Centre, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological DiseasesFirst Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Atsushi Fukui
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
| | - Shinichiro Saeki
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
| | - Ryu Takeyama
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
| | - Ayano Yamaya
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
| | - Hiroaki Shibahara
- Department of Obstetrics and GynecologySchool of Medicine, Hyogo Medical UniversityNishinomiyaHyogoJapan
| |
Collapse
|
9
|
Huang N, Ye L, Li H, Peng J, Wei H. Developmental patterns of intestinal group 3 innate lymphoid cells in piglets and their response to enterotoxigenic Escherichia coli infection. Vet Res 2024; 55:159. [PMID: 39695888 DOI: 10.1186/s13567-024-01418-3] [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: 10/06/2023] [Accepted: 09/22/2024] [Indexed: 12/20/2024] Open
Abstract
Diarrhoea and preweaning mortality in piglets are crucial factors impacting the economic sustainability of the swine industry. Pathogenic infections are among the main causes of diarrhea and mortality. Group 3 innate lymphoid cells (ILC3s) are crucial for safeguarding against pathogenic infections. However, knowledge regarding the development and function of ILC3s in suckling piglets is currently limited. Our findings demonstrate that the development of ILC3s in suckling piglets gradually progresses from day 1 to day 21, with a notable increase observed on day 28. Additionally, the development of NKp46+ILC3s and the production of interleukin (IL)-17A by ILC3s displayed consistent patterns with the changes observed in ILC3s. Notably, interferon (IFN)-γ levels significantly increased on day 14. Moreover, the production of IFN-γ by NKp46+ILC3s was greater than that by NKp46-ILC3s. Importantly, when piglets were subjected to a 4-h challenge with enterotoxigenic Escherichia coli, both the percentages of ILC3s significantly increased, accompanied by increased IL-22 production, highlighting their importance in maintaining intestinal health. The outcomes of this study provide valuable insights for future related research.
Collapse
Affiliation(s)
- Ningning Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Center of Cellular and Genetic Sciences, Henan Academy of Sciences, Zhengzhou, 450000, China
| | - Ling Ye
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hao Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.
| |
Collapse
|
10
|
Wang J, Lou W, Li Y, Jiang Y, Jiang X, Yang L. Progress in targeted therapy for ankylosing spondylitis: A review. Medicine (Baltimore) 2024; 103:e40742. [PMID: 39612456 PMCID: PMC11608699 DOI: 10.1097/md.0000000000040742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024] Open
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease characterized by axial osteoarticular inflammation and tendon enthesitis with unclear pathogenesis. Nonsteroidal anti-inflammatory drugs and antirheumatic drugs used in the traditional treatment of AS have some problems such as drug intolerance and inadequate treatment response. Since the introduction of biological agents in the treatment of AS, they have completely changed the treatment concept of AS, and because of their safety and good tolerance, they have become the main choice for clinical AS patients. This article systematically summarizes the current status of targeted therapy for AS worldwide, analyzes the advantages and disadvantages of different types of biological agents in the treatment of AS, and provides an objective evaluation of clinical targeted therapy for AS, in order to provide a new perspective for clinical standardized treatment.
Collapse
Affiliation(s)
- Jiapeng Wang
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| | - Wang Lou
- Department of Anesthesiology, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| | - Yingnan Li
- Burn the Brotherhood of Plastic Surgery, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| | - Yang Jiang
- Department of Medical Laboratory, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| | - Xue Jiang
- Department of Rehabilitation, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| | - Lin Yang
- Department of Anesthesiology, Jilin Province FAW General Hospital, Changchun City, Jilin Province, China
| |
Collapse
|
11
|
Li J, Wu Z, Wu Y, Hu X, Yang J, Zhu D, Wu M, Li X, Bentum-Ennin L, Wanglai H. IL-22, a vital cytokine in autoimmune diseases. Clin Exp Immunol 2024; 218:242-263. [PMID: 38651179 PMCID: PMC11557150 DOI: 10.1093/cei/uxae035] [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: 02/01/2024] [Revised: 03/05/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
Interleukin-22 (IL-22) is a vital cytokine that is dysregulated in various autoimmune conditions including rheumatoid arthritis (RA), multiple sclerosis (MS), and Alzheimer's disease (AD). As the starting point for the activation of numerous signaling pathways, IL-22 plays an important role in the initiation and development of autoimmune diseases. Specifically, imbalances in IL-22 signaling can interfere with other signaling pathways, causing cross-regulation of target genes which ultimately leads to the development of immune disorders. This review delineates the various connections between the IL-22 signaling pathway and autoimmune disease, focusing on the latest understanding of the cellular sources of IL-22 and its effects on various cell types. We further explore progress with pharmacological interventions related to targeting IL-22, describing how such therapeutic strategies promise to usher in a new era in the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Jiajin Li
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Zhen Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Yuxin Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - XinYu Hu
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Jun Yang
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Dacheng Zhu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Mingyue Wu
- The School of pharmacy, Anhui Medical University, Hefei, China
| | - Xin Li
- The School of pharmacy, Anhui Medical University, Hefei, China
| | | | - Hu Wanglai
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Fachi JL, de Oliveira S, Gilfillan S, Antonova AU, Hou J, Vinolo MAR, Colonna M. NKp46 + ILC3s promote early neutrophil defense against Clostridioides difficile infection through GM-CSF secretion. Proc Natl Acad Sci U S A 2024; 121:e2416182121. [PMID: 39475653 PMCID: PMC11551360 DOI: 10.1073/pnas.2416182121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a common cause of antibiotic-associated colitis. C. difficile proliferates and produces toxins that damage the colonic epithelium, leading to symptoms ranging from mild diarrhea to severe pseudomembranous colitis. The host's innate response to CDI occurs in two phases: an early phase in which neutrophils reduce the bacterial load and a late phase involving repair mechanisms to restore epithelial integrity. Group 3 innate lymphoid cells (ILC3s) are crucial in protecting the gut from CDI. Previous studies have shown that ILC3-derived IL-22 is essential in the late phase of CDI for epithelial repair and maintaining an intestinal microbiota that competes with C. difficile, preventing its expansion. Our study finds that ILC3s also protect during the early stages of CDI by sustaining neutrophils through GM-CSF. Less neutrophil production, accumulation, and activation was evident in ILC3-deficient mice than in wild-type (WT) mice, which led to exacerbated symptoms, impaired pathogen clearance, a compromised epithelial barrier, and increased mortality. The adoptive transfer of ILC3s into ILC3-deficient mice restored neutrophil responses and improved disease outcomes. Both in vitro and in vivo experiments revealed that GM-CSF production by ILC3s is crucial for neutrophil production and effective resistance during CDI. Using mice lacking NKp46+ ILC3s, we found that this subset significantly contributes to GM-CSF production in CDI. These findings highlight the critical role of the ILC3-neutrophil connection in early innate responses to CDI. Enhancing ILC3 production of GM-CSF could be a promising strategy for improving host defense against CDI and other enteric infections.
Collapse
Affiliation(s)
- José L. Fachi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - Sarah de Oliveira
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP13083-862, Brazil
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| | - JinChao Hou
- Department of Anesthesiology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou310052, China
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP13083-862, Brazil
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO63110
| |
Collapse
|
14
|
Lo JW, Schroeder JH, Roberts LB, Mohamed R, Cozzetto D, Beattie G, Omer OS, Ross EM, Heuts F, Jowett GM, Read E, Madgwick M, Neves JF, Korcsmaros T, Jenner RG, Walker LSK, Powell N, Lord GM. CTLA-4 expressing innate lymphoid cells modulate mucosal homeostasis in a microbiota dependent manner. Nat Commun 2024; 15:9520. [PMID: 39496592 PMCID: PMC11535242 DOI: 10.1038/s41467-024-51719-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/10/2024] [Indexed: 11/06/2024] Open
Abstract
The maintenance of intestinal homeostasis is a fundamental process critical for organismal integrity. Sitting at the interface of the gut microbiome and mucosal immunity, adaptive and innate lymphoid populations regulate the balance between commensal micro-organisms and pathogens. Checkpoint inhibitors, particularly those targeting the CTLA-4 pathway, disrupt this fine balance and can lead to inflammatory bowel disease and immune checkpoint colitis. Here, we show that CTLA-4 is expressed by innate lymphoid cells and that its expression is regulated by ILC subset-specific cytokine cues in a microbiota-dependent manner. Genetic deletion or antibody blockade of CTLA-4 in multiple in vivo models of colitis demonstrates that this pathway plays a key role in intestinal homeostasis. Lastly, we have found that this observation is conserved in human IBD. We propose that this population of CTLA-4-positive ILC may serve as an important target for the treatment of idiopathic and iatrogenic intestinal inflammation.
Collapse
Affiliation(s)
- Jonathan W Lo
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
| | | | - Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rami Mohamed
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Domenico Cozzetto
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
| | - Gordon Beattie
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
- Genomics Translational Technology Platform, UCL Cancer Institute, University College London, London, UK
| | - Omer S Omer
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Ellen M Ross
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Frank Heuts
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Geraldine M Jowett
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Emily Read
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
| | - Matthew Madgwick
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Joana F Neves
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
| | - Tamas Korcsmaros
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Richard G Jenner
- UCL Cancer Institute and CRUK City of London Centre, University College London, London, UK
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Nick Powell
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK.
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK.
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
15
|
Rahman MA, Silva de Castro I, Schifanella L, Bissa M, Franchini G. Vaccine induced mucosal and systemic memory NK/ILCs elicit decreased risk of SIV/SHIV acquisition. Front Immunol 2024; 15:1441793. [PMID: 39301032 PMCID: PMC11410642 DOI: 10.3389/fimmu.2024.1441793] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/09/2024] [Indexed: 09/22/2024] Open
Abstract
SIV and HIV-based envelope V1-deleted (ΔV1) vaccines, delivered systemically by the DNA/ALVAC/gp120 platform, decrease the risk of mucosal SIV or SHIV acquisition more effectively than V1-replete vaccines. Here we investigated the induction of mucosal and systemic memory-like NK cells as well as antigen-reactive ILC response by DNA/ALVAC/gp120-based vaccination and their role against SIV/SHIV infection. ΔV1 HIV vaccination elicited a higher level of mucosal TNF-α+ and CD107+ memory-like NK cells than V1-replete vaccination, suggesting immunogen dependence. Mucosal memory-like NK cells, systemic granzyme B+ memory NK cells, and vaccine-induced mucosal envelope antigen-reactive IL-17+ NKp44+ ILCs, IL-17+ ILC3s, and IL-13+ ILC2 subsets were linked to a lower risk of virus acquisition. Additionally, mucosal memory-like NK cells and mucosal env-reactive IFN-γ+ ILC1s and env- reactive IL-13+ ILC2 subsets correlated with viral load control. We further observed a positive correlation between post-vaccination systemic and mucosal memory-like NK cells, suggesting vaccination enhances the presence of these cells in both compartments. Mucosal and systemic memory-like NK cells positively correlated with V2-specific ADCC responses, a reproducible correlate of reduced risk of SIV/HIV infection. In contrast, an increased risk was associated with the level of mucosal PMA/Ionomycin-induced IFN-γ+ and CD107+ NKG2A-NKp44- ILCs. Plasma proteomic analyses demonstrated that suppression of mucosal memory-like NK cells was linked to the level of CCL-19, LT-α, TNFSF-12, and IL-15, suppression of systemic env-reactive granzyme B+ memory-like NK cells was associated with the level of OLR1, CCL-3, and OSM, and suppression of IL-17+ ILCs immunity was correlated with the level of IL-6 and CXCL-9. In contrast, FLT3 ligand was associated with promotion of protective mucosal env-reactive IL-17+ responses. These findings emphasize the importance of mucosal memory-like NK cell and envelope- reactive ILC responses for protection against mucosal SIV/SHIV acquisition.
Collapse
Affiliation(s)
- Mohammad Arif Rahman
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Isabela Silva de Castro
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Luca Schifanella
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Massimiliano Bissa
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
16
|
Villageliu DN, Cunningham KC, Smith DR, Knoell DL, Mandolfo M, Wyatt TA, Samuelson DR. Natural killer cell effector function is critical for host defense against alcohol-associated bacterial pneumonia. NPJ Biofilms Microbiomes 2024; 10:79. [PMID: 39227647 PMCID: PMC11372167 DOI: 10.1038/s41522-024-00558-w] [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: 09/05/2023] [Accepted: 08/25/2024] [Indexed: 09/05/2024] Open
Abstract
Alcohol use is an independent risk factor for the development of bacterial pneumonia due, in part, to impaired mucus-facilitated clearance, macrophage phagocytosis, and recruitment of neutrophils. Alcohol consumption is also known to reduce peripheral natural killer (NK) cell numbers and compromise NK cell cytolytic activity, especially NK cells with a mature phenotype. However, the role of innate lymphocytes, such as NK cells during host defense against alcohol-associated bacterial pneumonia is essentially unknown. We have previously shown that indole supplementation mitigates increases in pulmonary bacterial burden and improves pulmonary NK cell recruitment in alcohol-fed mice, which were dependent on aryl hydrocarbon receptor (AhR) signaling. Employing a binge-on-chronic alcohol-feeding model we sought to define the role and interaction of indole and NK cells during pulmonary host defense against alcohol-associated pneumonia. We demonstrate that alcohol dysregulates NK cell effector function and pulmonary recruitment via alterations in two key signaling pathways. We found that alcohol increases transforming growth factor beta (TGF-β) signaling while suppressing AhR signaling. We further demonstrated that NK cells isolated from alcohol-fed mice have a reduced ability to kill Klebsiella pneumoniae. NK cell migratory capacity to chemokines was also significantly altered by alcohol, as NK cells isolated from alcohol-fed mice exhibited preferential migration in response to CXCR3 chemokines but exhibited reduced migration in response to CCR2, CXCR4, and CX3CR1 chemokines. Together this data suggests that alcohol disrupts NK cell-specific TGF-β and AhR signaling pathways leading to decreased pulmonary recruitment and cytolytic activity thereby increasing susceptibility to alcohol-associated bacterial pneumonia.
Collapse
Affiliation(s)
- Daniel N Villageliu
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelly C Cunningham
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Deandra R Smith
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daren L Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mason Mandolfo
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Todd A Wyatt
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Veterans Affairs Nebraska, University of Nebraska Medical Center, Western Iowa Health Care System, Omaha, NE, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care, & Sleep, University of Nebraska Medical Center, Omaha, NE, USA.
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
17
|
Wang W, Li N, Guo X. The crosstalk between ILC3s and adaptive immunity in diseases. FEBS J 2024; 291:3965-3977. [PMID: 37994218 DOI: 10.1111/febs.17014] [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: 06/23/2023] [Revised: 09/26/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
RORγt+ group 3 innate lymphoid cells (ILC3s), the innate counterpart of Th17 cells, are enriched in the mucosal area and lymphoid tissues. ILC3s interact with a variety of cells through their effector molecules and play an important role in the host defense against a spectrum of infections. Recent studies suggest that the extensive crosstalk between ILC3s and adaptive immune cells, especially T cells, is essential for maintaining tissue homeostasis. Here we discuss recent advances in the crosstalk between ILC3s and adaptive immune responses in multiple tissues and diseases. Understanding how ILC3s engage with adaptive immune cells will enhance our comprehension of diseases and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Wenyan Wang
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Na Li
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
18
|
Piersma SJ. Tissue-specific features of innate lymphoid cells in antiviral defense. Cell Mol Immunol 2024; 21:1036-1050. [PMID: 38684766 PMCID: PMC11364677 DOI: 10.1038/s41423-024-01161-x] [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: 12/25/2023] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Innate lymphocytes (ILCs) rapidly respond to and protect against invading pathogens and cancer. ILCs include natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer (LTi) cells and include type I, type II, and type III immune cells. While NK cells have been well recognized for their role in antiviral immunity, other ILC subtypes are emerging as players in antiviral defense. Each ILC subset has specialized functions that uniquely impact the antiviral immunity and health of the host depending on the tissue microenvironment. This review focuses on the specialized functions of each ILC subtype and their roles in antiviral immune responses across tissues. Several viruses within infection-prone tissues will be highlighted to provide an overview of the extent of the ILC immunity within tissues and emphasize common versus virus-specific responses.
Collapse
Affiliation(s)
- Sytse J Piersma
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
19
|
Bennstein SB, Uhrberg M. Circulating innate lymphoid cells (cILCs): Unconventional lymphocytes with hidden talents. J Allergy Clin Immunol 2024; 154:523-536. [PMID: 39046403 DOI: 10.1016/j.jaci.2024.06.016] [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: 03/18/2024] [Revised: 05/16/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Innate lymphoid cells (ILCs) are a group of lymphocytes that are devoid of antigen-specific receptors and are mainly found in tissues. The subtypes ILC1, 2, and 3 mirror T-cell functionality in terms of cytokine production and expression of key transcription factors. Although the majority of ILCs are found in tissue (tILCs), they have also been described within the circulation (cILCs). As a result of their better accessibility and putative prognostic value, human cILCs are getting more and more attention in clinical research. However, cILCs are in many aspects functionally distinct from their tILC counterparts. In fact, from the 3 ILC subsets found within the circulation, only for cILC2s could a clear functional correspondence to their tissue counterparts be established. Indeed, cILC2s are emerging as a major driver of allergic reactions with a particular role in asthma. In contrast, recent studies revealed that cILC1s and cILC3s are predominantly in an immature state and constitute progenitors for natural killer cells and ILCs, respectively. We provide an overview about the phenotype and function of the different cILC subtypes compared to tILCs in health and disease, including transcriptomic signatures, frequency dynamics, and potential clinical value. Furthermore, we will highlight the dynamics of the NKp44+ ILC3 subset, which emerges as prognostic marker in peripheral blood for inflammatory bowel disease and leukemia.
Collapse
Affiliation(s)
- Sabrina B Bennstein
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
20
|
Wang W, Ma L, Liu B, Ouyang L. The role of trained immunity in sepsis. Front Immunol 2024; 15:1449986. [PMID: 39221248 PMCID: PMC11363069 DOI: 10.3389/fimmu.2024.1449986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection, characterized by a systemic inflammatory response to infection. The use of antibiotics, fluid resuscitation, and organ support therapy has limited prognostic benefit in patients with sepsis, and its incidence is not diminishing, which is attracting increased attention in medicine. Sepsis remains one of the most debilitating and expensive illnesses. One of the main reasons of septic mortality is now understood to be disruption of immune homeostasis. Immunotherapy is revolutionizing the treatment of illnesses in which dysregulated immune responses play a significant role. This "trained immunity", which is a potent defense against infection regardless of the type of bacteria, fungus, or virus, is attributed to the discovery that the innate immune cells possess immune memory via metabolic and epigenetic reprogramming. Here we reviewed the immunotherapy of innate immune cells in sepsis, the features of trained immunity, and the relationship between trained immunity and sepsis.
Collapse
Affiliation(s)
| | | | | | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
21
|
Belghith M, Maghrebi O, Ben Laamari R, Hanachi M, Hrir S, Saied Z, Belal S, Driss A, Ben Sassi S, Boussoffara T, Barbouche MR. Increased IL-22 in cerebrospinal fluid of neuro-behçet's disease patients. Cytokine 2024; 179:156617. [PMID: 38631183 DOI: 10.1016/j.cyto.2024.156617] [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: 09/10/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Remitting-Relapsing Multiple Sclerosis (RRMS) and Neuro-Behçet Disease (NBD) are two chronic neuro-inflammatory disorders leading to brain damage and disability in young adults. Herein, we investigated in these patients the cytokine response by beads-based multiplex assays during the early stages of these disorders. Cytokine investigations were carried out on treatment-naive patients suffering from RRMS and NBD recruited at the first episode of clinical relapse. Our findings demonstrate that Cerebrospinal Fluid (CSF) cells from NBD patients, but not RRMS, secrete significant high levels of IL-22 which is associated with elevated IL-22 mRNA expression. We also observed an increase in IL-22 levels in the definite NBD subgroup as compared to the probable NBD one, indicating a clear relationship between elevated IL-22 levels and diagnostic certainty. Interestingly, we found no correlation of IL-22 secretion between CSF and serum arguing about intrathecal release of IL-22 in the CNS of NBD patients. Moreover, we showed by correlogram analysis that this cytokine doesn't correlate with IL-17A, IL-17F and IL-21 suggesting that this cytokine is secreted by Th22 cells and not by Th17 cells in the CSF of NBD patients. Finally, we found elevated levels of IL-6 and a positive correlation between IL and 6 and IL-22 in the CSF of NBD. In conclusion, these results suggest that IL-6 contributes to the production of IL-22 by T cells leading to the exacerbation of inflammation and damage within the CNS of NBD patients.
Collapse
Affiliation(s)
- Meriam Belghith
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Olfa Maghrebi
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Faculty of Medicine of Tunis, 1006, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Rafika Ben Laamari
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Mariem Hanachi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics-LR16IPT09, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Sana Hrir
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia
| | - Zakaria Saied
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Samir Belal
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Adel Driss
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA.
| | - Samia Ben Sassi
- Faculty of Medicine of Tunis, 1006, Tunisia; Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia
| | - Thouraya Boussoffara
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, Tunis, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Mohamed-Ridha Barbouche
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Science, Arabian Gulf University 26671, Manama, Bahrain.
| |
Collapse
|
22
|
Vo DN, Yuan O, Kanaya M, Telliam-Dushime G, Li H, Kotova O, Caglar E, Honnens de Lichtenberg K, Rahman SH, Soneji S, Scheding S, Bryder D, Malmberg KJ, Sitnicka E. A temporal developmental map separates human NK cells from noncytotoxic ILCs through clonal and single-cell analysis. Blood Adv 2024; 8:2933-2951. [PMID: 38484189 PMCID: PMC11176970 DOI: 10.1182/bloodadvances.2023011909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/05/2024] [Indexed: 06/04/2024] Open
Abstract
ABSTRACT Natural killer (NK) cells represent the cytotoxic member within the innate lymphoid cell (ILC) family that are important against viral infections and cancer. Although the NK cell emergence from hematopoietic stem and progenitor cells through multiple intermediate stages and the underlying regulatory gene network has been extensively studied in mice, this process is not well characterized in humans. Here, using a temporal in vitro model to reconstruct the developmental trajectory of NK lineage, we identified an ILC-restricted oligopotent stage 3a CD34-CD117+CD161+CD45RA+CD56- progenitor population, that exclusively gave rise to CD56-expressing ILCs in vitro. We also further investigated a previously nonappreciated heterogeneity within the CD56+CD94-NKp44+ subset, phenotypically equivalent to stage 3b population containing both group-1 ILC and RORγt+ ILC3 cells, that could be further separated based on their differential expression of DNAM-1 and CD161 receptors. We confirmed that DNAM-1hi S3b and CD161hiCD117hi ILC3 populations distinctively differed in their expression of effector molecules, cytokine secretion, and cytotoxic activity. Furthermore, analysis of lineage output using DNA-barcode tracing across these stages supported a close developmental relationship between S3b-NK and S4-NK (CD56+CD94+) cells, whereas distant to the ILC3 subset. Cross-referencing gene signatures of culture-derived NK cells and other noncytotoxic ILCs with publicly available data sets validated that these in vitro stages highly resemble transcriptional profiles of respective in vivo ILC counterparts. Finally, by integrating RNA velocity and gene network analysis through single-cell regulatory network inference and clustering we unravel a network of coordinated and highly dynamic regulons driving the cytotoxic NK cell program, as a guide map for future studies on NK cell regulation.
Collapse
Affiliation(s)
- Dang Nghiem Vo
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ouyang Yuan
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Minoru Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Gladys Telliam-Dushime
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Hongzhe Li
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Olga Kotova
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Emel Caglar
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Cell Therapy Research, Novo Nordisk A/S, Måløv, Copenhagen, Denmark
| | | | | | - Shamit Soneji
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Stefan Scheding
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Hematology, Skåne University Hospital, Lund, Sweden
| | - David Bryder
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ewa Sitnicka
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Horn V, Sonnenberg GF. Group 3 innate lymphoid cells in intestinal health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:428-443. [PMID: 38467885 PMCID: PMC11144103 DOI: 10.1038/s41575-024-00906-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/13/2024]
Abstract
The gastrointestinal tract is an immunologically rich organ, containing complex cell networks and dense lymphoid structures that safeguard this large absorptive barrier from pathogens, contribute to tissue physiology and support mucosal healing. Simultaneously, the immune system must remain tolerant to innocuous dietary antigens and trillions of normally beneficial microorganisms colonizing the intestine. Indeed, a dysfunctional immune response in the intestine underlies the pathogenesis of numerous local and systemic diseases, including inflammatory bowel disease, food allergy, chronic enteric infections or cancers. Here, we discuss group 3 innate lymphoid cells (ILC3s), which have emerged as orchestrators of tissue physiology, immunity, inflammation, tolerance and malignancy in the gastrointestinal tract. ILC3s are abundant in the developing and healthy intestine but their numbers or function are altered during chronic disease and cancer. The latest studies provide new insights into the mechanisms by which ILC3s fundamentally shape intestinal homeostasis or disease pathophysiology, and often this functional dichotomy depends on context and complex interactions with other cell types or microorganisms. Finally, we consider how this knowledge could be harnessed to improve current treatments or provoke new opportunities for therapeutic intervention to promote gut health.
Collapse
Affiliation(s)
- Veronika Horn
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
24
|
Wang L, Villafuerte Gálvez JA, Lee C, Wu S, Kelly CP, Chen X, Cao Y. Understanding host immune responses in Clostridioides difficile infection: Implications for pathogenesis and immunotherapy. IMETA 2024; 3:e200. [PMID: 38898983 PMCID: PMC11183162 DOI: 10.1002/imt2.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
Clostridioides difficile (C. difficile) is the predominant causative agent of nosocomial diarrhea worldwide. Infection with C. difficile occurs due to the secretion of large glycosylating toxin proteins, which can lead to toxic megacolon or mortality in susceptible hosts. A critical aspect of C. difficile's biology is its ability to persist asymptomatically within the human host. Individuals harboring asymptomatic colonization or experiencing a single episode of C. difficile infection (CDI) without recurrence exhibit heightened immune responses compared to symptomatic counterparts. The significance of these immune responses cannot be overstated, as they play critical roles in the development, progression, prognosis, and outcomes of CDI. Nonetheless, our current comprehension of the immune responses implicated in CDI remains limited. Therefore, further investigation is imperative to elucidate their underlying mechanisms. This review explores recent advancements in comprehending CDI pathogenesis and how the host immune system response influences disease progression and severity, aiming to enhance our capacity to develop immunotherapy-based treatments for CDI.
Collapse
Affiliation(s)
- Lamei Wang
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Javier A. Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Christina Lee
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Shengru Wu
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ciaran P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Yangchun Cao
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
25
|
Gilliet M, Modlin RL. Immunobiology of IL-26. J Invest Dermatol 2024; 144:1217-1222. [PMID: 38206272 DOI: 10.1016/j.jid.2023.10.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/22/2023] [Accepted: 10/22/2023] [Indexed: 01/12/2024]
Abstract
T helper 17 (Th17) cells produce a set of cytokines that include IL-17 family members, IL-21, IL-22, and IL-26. These cytokines all contribute to the classic function of Th17 cells in combatting extracellular infection and promoting inflammation in autoimmune diseases. However, of the Th17 cytokines, only IL-26 has direct antimicrobial activity against microbes and can activate a broad range of immune cells through its ability to bind DNA and trigger pattern recognition receptors. It is noteworthy that IL-26 is produced by mammalian cells, including human Th17 cells, but is absent in rodents. As such, IL-26 is a potential therapeutic target to augment host immune responses against microbial pathogens but also to prevent inflammation and tissue damage in a variety of autoimmune diseases.
Collapse
Affiliation(s)
- Michel Gilliet
- Department of Dermatology, CHUV University Hospital and University of Lausanne (UNIL), Lausanne, Switzerland.
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
26
|
Jones R, Robinson AT, Beach LB, Lindsey ML, Kirabo A, Hinton A, Erlandson KM, Jenkins ND. Exercise to Prevent Accelerated Vascular Aging in People Living With HIV. Circ Res 2024; 134:1607-1635. [PMID: 38781293 PMCID: PMC11126195 DOI: 10.1161/circresaha.124.323975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Given advances in antiretroviral therapy, the mortality rate for HIV infection has dropped considerably over recent decades. However, people living with HIV (PLWH) experience longer life spans coupled with persistent immune activation despite viral suppression and potential toxicity from long-term antiretroviral therapy use. Consequently, PLWH face a cardiovascular disease (CVD) risk more than twice that of the general population, making it the leading cause of death among this group. Here, we briefly review the epidemiology of CVD in PLWH highlighting disparities at the intersections of sex and gender, age, race/ethnicity, and the contributions of social determinants of health and psychosocial stress to increased CVD risk among individuals with marginalized identities. We then overview the pathophysiology of HIV and discuss the primary factors implicated as contributors to CVD risk among PLWH on antiretroviral therapy. Subsequently, we highlight the functional evidence of premature vascular dysfunction as an early pathophysiological determinant of CVD risk among PLWH, discuss several mechanisms underlying premature vascular dysfunction in PLWH, and synthesize current research on the pathophysiological mechanisms underlying accelerated vascular aging in PLWH, focusing on immune activation, chronic inflammation, and oxidative stress. We consider understudied aspects such as HIV-related changes to the gut microbiome and psychosocial stress, which may serve as mechanisms through which exercise can abrogate accelerated vascular aging. Emphasizing the significance of exercise, we review various modalities and their impacts on vascular health, proposing a holistic approach to managing CVD risks in PLWH. The discussion extends to critical future study areas related to vascular aging, CVD, and the efficacy of exercise interventions, with a call for more inclusive research that considers the diversity of the PLWH population.
Collapse
Affiliation(s)
- Raymond Jones
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Lauren B. Beach
- Department of Medical Social Sciences, Northwestern, Chicago, IL
- Department of Preventive Medicine, Northwestern, Chicago, IL
| | - Merry L. Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, TN
- Research Service, Nashville VA Medical Center, Nashville, TN
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Nashville, TN
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN
- Vanderbilt Institute for Global Health, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | | | - Nathaniel D.M. Jenkins
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| |
Collapse
|
27
|
Hammitzsch A, Ossadnik A, Bachmann Q, Merwald-Fraenk H, Lorenz G, Witt M, Wiesent F, Mühlhofer H, Simone D, Bowness P, Heemann U, Arbogast M, Moog P, Schmaderer C. Increased interleukin-26 in the peripheral joints of patients with axial spondyloarthritis and psoriatic arthritis, co-localizing with CD68-positive synoviocytes. Front Immunol 2024; 15:1355824. [PMID: 38799447 PMCID: PMC11127564 DOI: 10.3389/fimmu.2024.1355824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/01/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives IL26 levels are elevated in the blood and synovial fluid of patients with inflammatory arthritis. IL26 can be produced by Th17 cells and locally within joints by tissue-resident cells. IL26 induces osteoblast mineralization in vitro. As osteoproliferation and Th17 cells are important factors in the pathogenesis of axial spondyloarthritis (axSpA), we aimed to clarify the cellular sources of IL26 in spondyloarthritis. Methods Serum, peripheral blood mononuclear cells (n = 15-35) and synovial tissue (n = 3-9) of adult patients with axSpA, psoriatic arthritis (PsA) and rheumatoid arthritis (RA) and healthy controls (HCs, n = 5) were evaluated by ELISA, flow cytometry including PrimeFlow assay, immunohistochemistry and immunofluorescence and quantitative PCR. Results Synovial tissue of axSpA patients shows significantly more IL26-positive cells than that of HCs (p < 0.01), but numbers are also elevated in PsA and RA patients. Immunofluorescence shows co-localization of IL26 with CD68, but not with CD3, SMA, CD163, cadherin-11, or CD90. IL26 is elevated in the serum of RA and PsA (but not axSpA) patients compared with HCs (p < 0.001 and p < 0.01). However, peripheral blood CD4+ T cells from axSpA and PsA patients show higher positivity for IL26 in the PrimeFlow assay compared with HCs. CD4+ memory T cells from axSpA patients produce more IL26 under Th17-favoring conditions (IL-1β and IL-23) than cells from PsA and RA patients or HCs. Conclusion IL26 production is increased in the synovial tissue of SpA and can be localized to CD68+ macrophage-like synoviocytes, whereas circulating IL26+ Th17 cells are only modestly enriched. Considering the osteoproliferative properties of IL26, this offers new therapeutic options independent of Th17 pathways.
Collapse
Affiliation(s)
- Ariane Hammitzsch
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Andreas Ossadnik
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Quirin Bachmann
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helga Merwald-Fraenk
- Amedes Holding AG, Ambulatory Healthcare Center (MVZ) Endokrinologikum München, Munich, Germany
| | - Georg Lorenz
- Department of Nephrology and Rheumatology, Klinik Augustinum München, Munich, Germany
| | | | - Franziska Wiesent
- Amedes Holding AG, Ambulatory Healthcare Center (MVZ) Endokrinologikum München, Munich, Germany
| | - Heinrich Mühlhofer
- Clinic and Policlinic of Orthopaedics and Sports’ Orthopaedics, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Davide Simone
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Paul Bowness
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Uwe Heemann
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Arbogast
- Department of Rheumatic Orthopedics and Hand Surgery, Klinik Oberammergau, Waldburg-Zeil Kliniken GmbH und Co KG, Oberammergau, Germany
| | - Philipp Moog
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
28
|
Rallón N, Jiménez-Carretero D, Restrepo C, Ligos JM, Valentín-Quiroga J, Mahillo I, Cabello A, López-Collazo E, Sánchez-Cabo F, Górgolas M, Estrada V, Benito JM. A specific natural killer cells phenotypic signature associated to long term elite control of HIV infection. J Med Virol 2024; 96:e29646. [PMID: 38699988 DOI: 10.1002/jmv.29646] [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: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Elite controllers (ECs) are an exceptional group of people living with HIV (PLWH) that control HIV replication without therapy. Among the mechanisms involved in this ability, natural killer (NK)-cells have recently gained much attention. We performed an in-deep phenotypic analysis of NK-cells to search for surrogate markers associated with the long term spontaneous control of HIV. Forty-seven PLWH (22 long-term EC [PLWH-long-term elite controllers (LTECs)], 15 noncontrollers receiving antiretroviral treatment [ART] [PLWH-onART], and 10 noncontrollers cART-naïve [PLWH-offART]), and 20 uninfected controls were included. NK-cells homeostasis was analyzed by spectral flow cytometry using a panel of 15 different markers. Data were analyzed using FCSExpress and R software for unsupervised multidimensional analysis. Six different subsets of NK-cells were defined on the basis of CD16 and CD56 expression, and the multidimensional analysis revealed the existence of 68 different NK-cells clusters based on the expression levels of the 15 different markers. PLWH-offART presented the highest disturbance of NK-cells homeostasis and this was not completely restored by long-term ART. Interestingly, long term spontaneous control of HIV (PLWH-LTEC group) was associated with a specific profile of NK-cells homeostasis disturbance, characterized by an increase of CD16dimCD56dim subset when compared to uninfected controls (UC) group and also to offART and onART groups (p < 0.0001 for the global comparison), an increase of clusters C16 and C26 when compared to UC and onART groups (adjusted p-value < 0.05 for both comparisons), and a decrease of clusters C10 and C20 when compared to all the other groups (adjusted p-value < 0.05 for all comparisons). These findings may provide clues to elucidate markers of innate immunity with a relevant role in the long-term control of HIV.
Collapse
Affiliation(s)
- Norma Rallón
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Daniel Jiménez-Carretero
- Unidad de Bioinformática, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Clara Restrepo
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | | | | | - Ignacio Mahillo
- Department of Statistics, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Alfonso Cabello
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Eduardo López-Collazo
- Grupo de respuesta inmune innata, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Unidad de Bioinformática, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Górgolas
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | - José M Benito
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| |
Collapse
|
29
|
Serafini N, Di Santo JP. Group 3 innate lymphoid cells: A trained Gutkeeper. Immunol Rev 2024; 323:126-137. [PMID: 38491842 DOI: 10.1111/imr.13322] [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] [Indexed: 03/18/2024]
Abstract
Group 3 innate lymphoid cells (ILC3s) are tissue-resident immune lymphocytes that critically regulate intestinal homeostasis, organogenesis, and immunity. ILC3s possess the capacity to "sense" the inflammatory environment within tissues, especially in the context of pathogen challenges that imprints durable non-antigen-specific changes in ILC3 function. As such, ILC3s become a new actor in the emerging field of trained innate immunity. Here, we summarize recent discoveries regarding ILC3 responses to bacterial challenges and the role these encounters play in triggering trained innate immunity. We further discuss how signaling events throughout ILC3 ontogeny potentially control the development and function of trained ILC3s. Finally, we highlight the open questions surrounding ILC3 "training" the answers to which may reveal new insights into innate immunity. Understanding the fundamental concepts behind trained innate immunity could potentially lead to the development of new strategies for improving immunity-based modulation therapies for inflammation, infectious diseases, and cancer.
Collapse
Affiliation(s)
- Nicolas Serafini
- Innate Immunity Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, Paris, France
| |
Collapse
|
30
|
Kühnel I, Vogler I, Spreu J, Bonig H, Döring C, Steinle A. The activating receptor NKp65 is selectively expressed by human ILC3 and demarcates ILC3 from mature NK cells. Eur J Immunol 2024; 54:e2250318. [PMID: 38072999 DOI: 10.1002/eji.202250318] [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: 12/06/2022] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 02/27/2024]
Abstract
Innate lymphocytes comprise cytotoxic natural killer (NK) cells and tissue-resident innate lymphoid cells (ILC) that are subgrouped according to their cytokine profiles into group 1 ILC (ILC1), ILC2, and ILC3. However, cell surface receptors unambiguously defining or specifically activating such ILC subsets are scarcely known. Here, we report on the physiologic expression of the human activating C-type lectin-like receptor (CTLR) NKp65, a high-affinity receptor for the CTLR keratinocyte-associated C-type lectin (KACL). Tracking rare NKp65 transcripts in human blood, we identify ILC3 to selectively express NKp65. NKp65 expression not only demarcates "bona fide" ILC3 from likewise RORγt-expressing ILC precursors and lymphoid tissue inducer cells but also from mature NK cells which acquire the NKp65-relative NKp80 during a Notch-dependent differentiation from NKp65+ precursor cells. Hence, ILC3 and NK cells mutually exclusively and interdependently express the genetically coupled sibling receptors NKp65 and NKp80. Much alike NKp80, NKp65 promotes cytotoxicity by innate lymphocytes which may become relevant during pathophysiological reprogramming of ILC3. Altogether, we report the selective expression of the activating immunoreceptor NKp65 by ILC3 demarcating ILC3 from mature NK cells and endowing ILC3 with a dedicated immunosensor for the epidermal immune barrier.
Collapse
Affiliation(s)
- Ines Kühnel
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Isabel Vogler
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Jessica Spreu
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Frankfurt am Main, Germany
| | - Claudia Döring
- Dr. Senckenbergisches Institute of Pathology, Goethe University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
31
|
Ma Z, An P, Hao S, Huang Z, Yin A, Li Y, Tian J. Single-cell sequencing analysis and multiple machine-learning models revealed the cellular crosstalk of dendritic cells and identified FABP5 and KLRB1 as novel biomarkers for psoriasis. Front Immunol 2024; 15:1374763. [PMID: 38596682 PMCID: PMC11002082 DOI: 10.3389/fimmu.2024.1374763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
Background Psoriasis is an immune-mediated disorder influenced by environmental factors on a genetic basis. Despite advancements, challenges persist, including the diminishing efficacy of biologics and small-molecule targeted agents, alongside managing recurrence and psoriasis-related comorbidities. Unraveling the underlying pathogenesis and identifying valuable biomarkers remain pivotal for diagnosing and treating psoriasis. Methods We employed a series of bioinformatics (including single-cell sequencing data analysis and machine learning techniques) and statistical methods to integrate and analyze multi-level data. We observed the cellular changes in psoriatic skin tissues, screened the key genes Fatty acid binding protein 5 (FABP5) and The killer cell lectin-like receptor B1 (KLRB1), evaluated the efficacy of six widely prescribed drugs on psoriasis treatment in modulating the dendritic cell-associated pathway, and assessed their overall efficacy. Finally, RT-qPCR, immunohistochemistry, and immunofluorescence assays were used to validate. Results The regulatory influence of dendritic cells (DCs) on T cells through the CD70/CD27 signaling pathway may emerge as a significant facet of the inflammatory response in psoriasis. Notably, FABP5 and KLRB1 exhibited up-regulation and co-localization in psoriatic skin tissues and M5-induced HaCaT cells, serving as potential biomarkers influencing psoriasis development. Conclusion Our study analyzed the impact of DC-T cell crosstalk in psoriasis, elucidated the characterization of two biomarkers, FABP5 and KLRB1, in psoriasis, and highlighted the promise and value of tofacitinib in psoriasis therapy targeting DCs.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Pingyu An
- Basic Medical College, Harbin Medical University, Harbin, China
| | - Siyu Hao
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhangxin Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Anqi Yin
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuzhen Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiangtian Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| |
Collapse
|
32
|
Cheng A, Kashyap A, Salvator H, Rosen LB, Colby D, Ardeshir-Larijani F, Loehrer PJ, Ding L, Lugo Reyes SO, Riminton S, Ballman M, Rocco JM, Marciano BE, Freeman AF, Browne SK, Hsu AP, Zelazny A, Rajan A, Sereti I, Zerbe CS, Lionakis MS, Holland SM. Anti-Interleukin-23 Autoantibodies in Adult-Onset Immunodeficiency. N Engl J Med 2024; 390:1105-1117. [PMID: 38507753 DOI: 10.1056/nejmoa2210665] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
BACKGROUND Autoantibodies against interleukin-12 (anti-interleukin-12) are often identified in patients with thymoma, but opportunistic infections develop in only some of these patients. Interleukin-12 (with subunits p40 and p35) shares a common subunit with interleukin-23 (subunits p40 and p19). In a patient with disseminated Burkholderia gladioli infection, the identification of both anti-interleukin-23 and anti-interleukin-12 prompted further investigation. METHODS Among the patients (most of whom had thymoma) who were known to have anti-interleukin-12, we screened for autoantibodies against interleukin-23 (anti-interleukin-23). To validate the potential role of anti-interleukin-23 with respect to opportunistic infection, we tested a second cohort of patients with thymoma as well as patients without either thymoma or known anti-interleukin-12 who had unusual infections. RESULTS Among 30 patients with anti-interleukin-12 who had severe mycobacterial, bacterial, or fungal infections, 15 (50%) also had autoantibodies that neutralized interleukin-23. The potency of such neutralization was correlated with the severity of these infections. The neutralizing activity of anti-interleukin-12 alone was not associated with infection. In the validation cohort of 91 patients with thymoma, the presence of anti-interleukin-23 was associated with infection status in 74 patients (81%). Overall, neutralizing anti-interleukin-23 was detected in 30 of 116 patients (26%) with thymoma and in 30 of 36 patients (83%) with disseminated, cerebral, or pulmonary infections. Anti-interleukin-23 was present in 6 of 32 patients (19%) with severe intracellular infections and in 2 of 16 patients (12%) with unusual intracranial infections, including Cladophialophora bantiana and Mycobacterium avium complex. CONCLUSIONS Among patients with a variety of mycobacterial, bacterial, or fungal infections, the presence of neutralizing anti-interleukin-23 was associated with severe, persistent opportunistic infections. (Funded by the National Institute of Allergy and Infectious Diseases and others.).
Collapse
Affiliation(s)
- Aristine Cheng
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Anuj Kashyap
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Helene Salvator
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Lindsey B Rosen
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Devon Colby
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Fatemeh Ardeshir-Larijani
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Patrick J Loehrer
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Li Ding
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Saul O Lugo Reyes
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sean Riminton
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Madison Ballman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Joseph M Rocco
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Beatriz E Marciano
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Alexandra F Freeman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sarah K Browne
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Amy P Hsu
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Adrian Zelazny
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Arun Rajan
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Irini Sereti
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Christa S Zerbe
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Michail S Lionakis
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Steven M Holland
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| |
Collapse
|
33
|
Katirci E, Kendirci-Katirci R, Korgun ET. Are innate lymphoid cells friend or foe in human pregnancy? Am J Reprod Immunol 2024; 91:e13834. [PMID: 38500395 DOI: 10.1111/aji.13834] [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: 08/31/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Innate lymphoid cells (ILCs) are involved in the innate immune system because they lack specific antigen receptors and lineage markers. ILCs also display phenotypic and characteristic features of adaptive immune cells. Therefore, ILCs are functional in essential interactions between adaptive and innate immunity. ILCs are found in both lymphoid and nonlymphoid tissues and migrate to the area of inflammation during the inflammatory process. ILCs respond to pathogens by producing a variety of cytokines and are involved in the barrier defense of antigens and in many immunological processes such as allergic events. Recent research has shown that ILCs are functional during human pregnancy and have been suggested to be essential for the healthy progression of pregnancy. In this review, we focus on the role of ILCs in human pregnancy by discussing the relationship between ILCs and the pregnancy microenvironment, specifically summarizing the role of ILCs in physiological and pathological pregnancies.
Collapse
Affiliation(s)
- Ertan Katirci
- Department of Histology and Embryology, Faculty of Medicine, Ahi Evran University, Kirsehir, Turkey
| | - Remziye Kendirci-Katirci
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Emin Turkay Korgun
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
34
|
Stosik M, Tokarz-Deptuła B, Deptuła W. Innate lymphoid cells (ILCs) in teleosts against data on ILCs in humans. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109415. [PMID: 38296004 DOI: 10.1016/j.fsi.2024.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/13/2024]
Abstract
It is assumed that cells corresponding to innate lymphoid cells (ILCs) in humans, in addition to lymphoid tissue inducer cells (LTi), are also found in teleosts. In this systematic group of organisms, however, they are a poorly understood cell population. In contrast to the data on ILCs in humans, which also remain incomplete despite advanced research, in teleosts, these cells require much more attention. ILCs in teleosts have been presented as cells that may be evolutionary precursors of NK cells or ILCs identified in mammals, including humans. It is a highly heterogeneous group of cells in both humans and fish and their properties, as revealed by studies in humans, are most likely to remain strictly dependent on the location of these cells and the physiological state of the individual from which they originate. They form a bridge between innate and adaptive immunity. The premise of this paper is to review the current knowledge of ILCs in teleosts, taking into account data on similar cells in humans. A review of the knowledge concerning these particular cells, elements of innate immunity mechanisms as equivalent to, or perhaps dominant over, adaptive immunity mechanisms in teleosts, as presented, may inspire the need for further research.
Collapse
Affiliation(s)
- Michał Stosik
- Institute of Biological Sciences, University of Zielona Góra, Poland
| | | | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
35
|
Barrow AD, Cella M, Edeling MA, Khan MAAK, Cervantes-Barragan L, Bugatti M, Schmedt C, Vermi W, Colonna M. Cutting Edge: PDGF-DD Binding to NKp44 Costimulates TLR9 Signaling and Proinflammatory Cytokine Secretion in Human Plasmacytoid Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:369-374. [PMID: 38117750 DOI: 10.4049/jimmunol.2200496] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/22/2023] [Indexed: 12/22/2023]
Abstract
NKp44 is a human receptor originally found on activated NK cells, group 1 and group 3 innate lymphoid cells that binds dimers of platelet-derived growth factor D (PDGF-DD). NKp44 is also expressed on tissue plasmacytoid dendritic cells (PDCs), but NKp44-PDGF-DD interaction on PDCs remains unstudied. Engagement of NKp44 with PDGF-DD in vitro enhanced PDC secretion of IFN-α, TNF, and IL-6 in response to the TLR9 ligand CpG-ODN, but not TLR7/8 ligands. In tissues, PDCs were found in close contact with PDGF-DD-expressing cells in the high endothelial venules and epithelium of tonsils, melanomas, and skin lesions infected with Molluscum contagiosum. Recombinant PDGF-DD enhanced the serum IFN-α response to systemic HSV-1 infection in a humanized mouse model. We conclude that NKp44 integrates with TLR9 signaling to enhance PDC cytokine production. These findings may have bearings for immune responses to TLR9-based adjuvants, therapy for tumors expressing PDGF-DD, and infections with DNA viruses that induce PDGF-DD expression to enhance viral spread.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Melissa Anne Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Md Abdullah-Al-Kamran Khan
- Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Luisa Cervantes-Barragan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Microbiology and Immunology, Emory University, School of Medicine, Atlanta, GA
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, School of Medicine, University of Brescia, Brescia, Italy
| | | | - William Vermi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular and Translational Medicine, Section of Pathology, School of Medicine, University of Brescia, Brescia, Italy
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
36
|
Bourinet M, Anty R, Gual P, Luci C. Roles of innate lymphoid cells in metabolic and alcohol-associated liver diseases. JHEP Rep 2024; 6:100962. [PMID: 38304237 PMCID: PMC10831956 DOI: 10.1016/j.jhepr.2023.100962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/09/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024] Open
Abstract
Innate lymphoid cells (ILCs) have been identified as potent regulators of inflammation, cell death and wound healing, which are the main biological processes involved in the progression of chronic liver disease. Obesity and chronic alcohol consumption are the leading contributors to chronic liver diseases in developed countries, due to inappropriate lifestyles. In particular, inflammation is a key factor in these liver abnormalities and promotes the development of more severe lesions such as fibrosis, cirrhosis and hepatocellular carcinoma. Opposite roles of ILC subsets have been described in the development of chronic liver disease, depending on the stage and aetiology of the disease. The heterogeneous family of ILCs encompasses cytotoxic natural killer cells, the cytokine-producing type 1, 2 and 3 ILCs and lymphoid tissue inducer cells. Dysfunction of these immune cells provokes uncontrolled inflammation and tissue damage, which are the basis for tumour development. In this review, we provide an overview of the recent and putative roles of ILC subsets in obesity and alcohol-associated liver diseases, which are currently the major contributors to end-stage liver complications such as fibrosis/cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Manon Bourinet
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Rodolphe Anty
- Université Côte d’Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Philippe Gual
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Carmelo Luci
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| |
Collapse
|
37
|
Van der Meer JMR, Bulder I, Kuijk C, Kleijer M, Verheij MW, Omar SZ, Haverkate NJE, Dolstra H, Blom B, Hazenberg MD, Voermans C. Generation of human ILC3 from allogeneic and autologous CD34 + hematopoietic progenitors toward adoptive transfer. Cytotherapy 2024; 26:136-144. [PMID: 38149947 DOI: 10.1016/j.jcyt.2023.11.011] [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: 07/23/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023]
Abstract
Type 3 innate lymphoid cells (ILC3) are important in tissue homeostasis. In the gut, ILC3 repair damaged epithelium and suppress inflammation. In allogeneic hematopoietic cell transplantation (HCT), ILC3 protect against graft-versus-host disease (GvHD), most likely by restoring tissue damage and preventing inflammation. We hypothesize that supplementing HCT grafts with interleukin-22 (IL-22)-producing ILC3 may prevent acute GvHD. We therefore explored ex vivo generation of human IL-22-producing ILC3 from hematopoietic stem and progenitor cells (HSPC) obtained from adult, neonatal and fetal sources. We established a stroma-free system culturing human cord blood-derived CD34+ HSPC with successive cytokine mixes for 5 weeks. We analyzed the presence of phenotypically defined ILC, their viability, proliferation and IL-22 production (after stimulation) by flow cytometry and enzyme-linked immunosorbent assay (ELISA). We found that the addition of recombinant human IL-15 and the enhancer of zeste homolog 1/2 inhibitor UNC1999 promoted ILC3 generation. Similar results were demonstrated when UNC1999 was added to CD34+ HSPC derived from healthy adult granulocyte colony-stimulating factor mobilized peripheral blood and bone marrow, but not fetal liver. UNC1999 did not negatively impact IL-22 production in any of the HSPC sources. Finally, we observed that autologous HSPC mobilized from the blood of adults with hematological malignancies also developed into ILC3, albeit with a significantly lower capacity. Together, we developed a stroma-free protocol to generate large quantities of IL-22-producing ILC3 from healthy adult human HSPC that can be applied for adoptive transfer to prevent GvHD after allogeneic HCT.
Collapse
Affiliation(s)
- Jolien M R Van der Meer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Ingrid Bulder
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Carlijn Kuijk
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Myrddin W Verheij
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Said Z Omar
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke J E Haverkate
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Mette D Hazenberg
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
38
|
Yamada K, Huang ZQ, Reily C, Green TJ, Suzuki H, Novak J, Suzuki Y. LIF/JAK2/STAT1 Signaling Enhances Production of Galactose-Deficient IgA1 by IgA1-Producing Cell Lines Derived From Tonsils of Patients With IgA Nephropathy. Kidney Int Rep 2024; 9:423-435. [PMID: 38344714 PMCID: PMC10851019 DOI: 10.1016/j.ekir.2023.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 02/28/2024] Open
Abstract
Introduction Galactose-deficient IgA1 (Gd-IgA1) plays a key role in the pathogenesis of IgA nephropathy (IgAN). Tonsillectomy has been beneficial to some patients with IgAN, possibly due to the removal of tonsillar cytokine-activated cells producing Gd-IgA1. To test this hypothesis, we used immortalized IgA1-producing cell lines derived from tonsils of patients with IgAN or obstructive sleep apnea (OSA) and assessed the effect of leukemia inhibitory factor (LIF) or oncostatin M (OSM) on Gd-IgA1 production. Methods Gd-IgA1 production was measured by lectin enzyme-linked immunosorbent assay; JAK-STAT signaling in cultured cells was assessed by immunoblotting of cell lysates; and validated by using small interfering RNA (siRNA) knock-down and small-molecule inhibitors. Results IgAN-derived cells produced more Gd-IgA1 than the cells from patients with OSA, and exhibited elevated Gd-IgA1 production in response to LIF, but not OSM. This effect was associated with dysregulated STAT1 phosphorylation, as confirmed by STAT1 siRNA knock-down. JAK2 inhibitor, AZD1480 exhibited a dose-dependent inhibition of the LIF-induced Gd-IgA1 overproduction. Unexpectedly, high concentrations of AZD1480, but only in the presence of LIF, reduced Gd-IgA1 production in the cells derived from patients with IgAN to that of the control cells from patients with OSA. Based on modeling LIF-LIFR-gp130-JAK2 receptor complex, we postulate that LIF binding to LIFR may sequester gp130 and/or JAK2 from other pathways; and when combined with JAK2 inhibition, enables full blockade of the aberrant O-glycosylation pathways in IgAN. Conclusion In summary, IgAN cells exhibit LIF-mediated overproduction of Gd-IgA1 due to abnormal signaling. JAK2 inhibitors can counter these LIF-induced effects and block Gd-IgA1 synthesis in IgAN.
Collapse
Affiliation(s)
- Koshi Yamada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Zhi-Qiang Huang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Todd J. Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Yamada K, Novak J, Suzuki Y. GWAS-follow-up Studies Identified a Connection between Abnormal LIF/JAK2/STAT1 Signaling and Overproduction of Galactose-Deficient IgA1 in the Tonsillar IgA1-Secreting Cells from Patients with IgA Nephropathy. JOURNAL OF CLINICAL RESEARCH & BIOETHICS 2024; 15:478. [PMID: 38440092 PMCID: PMC10911063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Affiliation(s)
- Koshi Yamada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Zecher BF, Ellinghaus D, Schloer S, Niehrs A, Padoan B, Baumdick ME, Yuki Y, Martin MP, Glow D, Schröder-Schwarz J, Niersch J, Brias S, Müller LM, Habermann R, Kretschmer P, Früh T, Dänekas J, Wehmeyer MH, Poch T, Sebode M, Ellinghaus E, Degenhardt F, Körner C, Hoelzemer A, Fehse B, Oldhafer KJ, Schumacher U, Sauter G, Carrington M, Franke A, Bunders MJ, Schramm C, Altfeld M. HLA-DPA1*02:01~B1*01:01 is a risk haplotype for primary sclerosing cholangitis mediating activation of NKp44+ NK cells. Gut 2024; 73:325-337. [PMID: 37788895 PMCID: PMC10850656 DOI: 10.1136/gutjnl-2023-329524] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/11/2023] [Indexed: 10/05/2023]
Abstract
OBJECTIVE Primary sclerosing cholangitis (PSC) is characterised by bile duct strictures and progressive liver disease, eventually requiring liver transplantation. Although the pathogenesis of PSC remains incompletely understood, strong associations with HLA-class II haplotypes have been described. As specific HLA-DP molecules can bind the activating NK-cell receptor NKp44, we investigated the role of HLA-DP/NKp44-interactions in PSC. DESIGN Liver tissue, intrahepatic and peripheral blood lymphocytes of individuals with PSC and control individuals were characterised using flow cytometry, immunohistochemical and immunofluorescence analyses. HLA-DPA1 and HLA-DPB1 imputation and association analyses were performed in 3408 individuals with PSC and 34 213 controls. NK cell activation on NKp44/HLA-DP interactions was assessed in vitro using plate-bound HLA-DP molecules and HLA-DPB wildtype versus knock-out human cholangiocyte organoids. RESULTS NKp44+NK cells were enriched in livers, and intrahepatic bile ducts of individuals with PSC showed higher expression of HLA-DP. HLA-DP haplotype analysis revealed a highly elevated PSC risk for HLA-DPA1*02:01~B1*01:01 (OR 1.99, p=6.7×10-50). Primary NKp44+NK cells exhibited significantly higher degranulation in response to plate-bound HLA-DPA1*02:01-DPB1*01:01 compared with control HLA-DP molecules, which were inhibited by anti-NKp44-blocking. Human cholangiocyte organoids expressing HLA-DPA1*02:01-DPB1*01:01 after IFN-γ-exposure demonstrated significantly increased binding to NKp44-Fc constructs compared with unstimulated controls. Importantly, HLA-DPA1*02:01-DPB1*01:01-expressing organoids increased degranulation of NKp44+NK cells compared with HLA-DPB1-KO organoids. CONCLUSION Our studies identify a novel PSC risk haplotype HLA-DP A1*02:01~DPB1*01:01 and provide clinical and functional data implicating NKp44+NK cells that recognise HLA-DPA1*02:01-DPB1*01:01 expressed on cholangiocytes in PSC pathogenesis.
Collapse
Affiliation(s)
- Britta F Zecher
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | | | | | | | | | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Maureen P Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Dawid Glow
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennifer Schröder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sébastien Brias
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
| | | | | | | | | | | | - Malte H Wehmeyer
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Poch
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marcial Sebode
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | | | - Angelique Hoelzemer
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Leibniz Institute of Virology, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karl J Oldhafer
- Department of General & Abdominal Surgery, Asklepios Hospital Barmbek, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Madeleine J Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Ist Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Martin Zeitz Center for Rare Diseases and Hamburg Centre for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Altfeld
- Leibniz Institute of Virology, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
41
|
Abramson J, Dobeš J, Lyu M, Sonnenberg GF. The emerging family of RORγt + antigen-presenting cells. Nat Rev Immunol 2024; 24:64-77. [PMID: 37479834 PMCID: PMC10844842 DOI: 10.1038/s41577-023-00906-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 07/23/2023]
Abstract
Antigen-presenting cells (APCs) are master regulators of the immune response by directly interacting with T cells to orchestrate distinct functional outcomes. Several types of professional APC exist, including conventional dendritic cells, B cells and macrophages, and numerous other cell types have non-classical roles in antigen presentation, such as thymic epithelial cells, endothelial cells and granulocytes. Accumulating evidence indicates the presence of a new family of APCs marked by the lineage-specifying transcription factor retinoic acid receptor-related orphan receptor-γt (RORγt) and demonstrates that these APCs have key roles in shaping immunity, inflammation and tolerance, particularly in the context of host-microorganism interactions. These RORγt+ APCs include subsets of group 3 innate lymphoid cells, extrathymic autoimmune regulator-expressing cells and, potentially, other emerging populations. Here, we summarize the major findings that led to the discovery of these RORγt+ APCs and their associated functions. We discuss discordance in recent reports and identify gaps in our knowledge in this burgeoning field, which has tremendous potential to advance our understanding of fundamental immune concepts.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Mengze Lyu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
42
|
Valle-Noguera A, Sancho-Temiño L, Castillo-González R, Villa-Gómez C, Gomez-Sánchez MJ, Ochoa-Ramos A, Yagüe-Fernández P, Soler Palacios B, Zorita V, Raposo-Ponce B, González-Granado JM, Aragonés J, Cruz-Adalia A. IL-18-induced HIF-1α in ILC3s ameliorates the inflammation of C. rodentium-induced colitis. Cell Rep 2023; 42:113508. [PMID: 38019650 DOI: 10.1016/j.celrep.2023.113508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are vital for defending tissue barriers from invading pathogens. Hypoxia influences the production of intestinal ILC3-derived cytokines by activating HIF. Yet, the mechanisms governing HIF-1α in ILC3s and other innate RORγt+ cells during in vivo infections are poorly understood. In our study, transgenic mice with specific Hif-1a gene inactivation in innate RORγt+ cells (RAG1KO HIF-1α▵Rorc) exhibit more severe colitis following Citrobacter rodentium infection, primarily due to the inability to upregulate IL-22. We find that HIF-1α▵Rorc mice have impaired IL-22 production in ILC3s, while non-ILC3 innate RORγt+ cells, also capable of producing IL-22, remain unaffected. Furthermore, we show that IL-18, induced by Toll-like receptor 2, selectively triggers IL-22 in ILC3s by transcriptionally upregulating HIF-1α, revealing an oxygen-independent regulatory pathway. Our results highlight that, during late-stage C. rodentium infection, IL-18 induction in the colon promotes IL-22 through HIF-1α in ILC3s, which is crucial for protection against this pathogen.
Collapse
Affiliation(s)
- Ana Valle-Noguera
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Lucía Sancho-Temiño
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Raquel Castillo-González
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cristina Villa-Gómez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - María José Gomez-Sánchez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Anne Ochoa-Ramos
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Blanca Soler Palacios
- Department of Immunology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Virginia Zorita
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - José María González-Granado
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Julián Aragonés
- Hospital Santa Cristina, Fundación de Investigación Hospital de la Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Aránzazu Cruz-Adalia
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.
| |
Collapse
|
43
|
Hartley VL, Qaqish AM, Wood MJ, Studnicka BT, Iwai K, Liu TC, MacDuff DA. HOIL1 Regulates Group 3 Innate Lymphoid Cells in the Colon and Protects against Systemic Dissemination, Colonic Ulceration, and Lethality from Citrobacter rodentium Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1823-1834. [PMID: 37902285 PMCID: PMC10841105 DOI: 10.4049/jimmunol.2300351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023]
Abstract
Heme-oxidized IRP2 ubiquitin ligase-1 (HOIL1)-deficient patients experience chronic intestinal inflammation and diarrhea as well as increased susceptibility to bacterial infections. HOIL1 is a component of the linear ubiquitin chain assembly complex that regulates immune signaling pathways, including NF-κB-activating pathways. We have shown previously that HOIL1 is essential for survival following Citrobacter rodentium gastrointestinal infection of mice, but the mechanism of protection by HOIL1 was not examined. C. rodentium is an important murine model for human attaching and effacing pathogens, enteropathogenic and enterohemorrhagic Escherichia coli that cause diarrhea and foodborne illnesses and lead to severe disease in children and immunocompromised individuals. In this study, we found that C. rodentium infection resulted in severe colitis and dissemination of C. rodentium to systemic organs in HOIL1-deficient mice. HOIL1 was important in the innate immune response to limit early replication and dissemination of C. rodentium. Using bone marrow chimeras and cell type-specific knockout mice, we found that HOIL1 functioned in radiation-resistant cells and partly in radiation-sensitive cells and in myeloid cells to limit disease, but it was dispensable in intestinal epithelial cells. HOIL1 deficiency significantly impaired the expansion of group 3 innate lymphoid cells and their production of IL-22 during C. rodentium infection. Understanding the role HOIL1 plays in type 3 inflammation and in limiting the pathogenesis of attaching and effacing lesion-forming bacteria will provide further insight into the innate immune response to gastrointestinal pathogens and inflammatory disorders.
Collapse
Affiliation(s)
- Victoria L. Hartley
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Arwa M. Qaqish
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Matthew J. Wood
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Brian T. Studnicka
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Donna A. MacDuff
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
44
|
Choi HS, Kuchroo VK. Expansion of the Innate Lymphocyte Family: Discovery of IL-22-Producing ILC3s. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1609-1611. [PMID: 37983522 DOI: 10.4049/jimmunol.2300390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
This Pillars of Immunology article is a commentary on “A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity,” a pivotal article written by M. Cella, A. Fuchs, W. Vermi, F. Facchetti, K. Otero, J. K. M. Lennerz, J. M. Doherty, J. C. Mills, and M. Colonna, and published in Nature, in 2009. https://www.nature.com/articles/nature07537.
Collapse
Affiliation(s)
- Hee Sun Choi
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA; and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA; and Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
45
|
Gupta SK, Vyavahare S, Duchesne Blanes IL, Berger F, Isales C, Fulzele S. Microbiota-derived tryptophan metabolism: Impacts on health, aging, and disease. Exp Gerontol 2023; 183:112319. [PMID: 37898179 DOI: 10.1016/j.exger.2023.112319] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
The intricate interplay between gut microbiota and the host is pivotal in maintaining homeostasis and health. Dietary tryptophan (TRP) metabolism initiates a cascade of essential endogenous metabolites, including kynurenine, kynurenic acid, serotonin, and melatonin, as well as microbiota-derived Trp metabolites like tryptamine, indole propionic acid (IPA), and other indole derivatives. Notably, tryptamine and IPA, among the indole metabolites, exert crucial roles in modulating immune, metabolic, and neuronal responses at both local and distant sites. Additionally, these metabolites demonstrate potent antioxidant and anti-inflammatory activities. The levels of microbiota-derived TRP metabolites are intricately linked to the gut microbiota's health, which, in turn, can be influenced by age-related changes. This review aims to comprehensively summarize the cellular and molecular impacts of tryptamine and IPA on health and aging-related complications. Furthermore, we explore the levels of tryptamine and IPA and their corresponding bacteria in select diseased conditions, shedding light on their potential significance as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ian L Duchesne Blanes
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ford Berger
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
46
|
He J, Zhao M, Ma X, Li D, Kong J, Yang F. The role and application of three IFN-related reactions in psoriasis. Biomed Pharmacother 2023; 167:115603. [PMID: 37776636 DOI: 10.1016/j.biopha.2023.115603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
The pathophysiology of psoriasis is a highly complicated one. Due to the disease's specificity, it not only affects the patient's skin negatively but also manifests systemic pathological changes. These clinical symptoms seriously harm the patient's physical and mental health. IFN, a common immunomodulatory factor, has been increasingly demonstrated to have a significant role in the development of psoriatic skin disease. Psoriasis is connected with a variety of immunological responses. New targets for the therapy of autoimmune skin diseases may emerge from further research on the mechanics of the associated IFN upstream and downstream pathways. Different forms of IFNs do not behave in the same manner in psoriasis, and understanding how different types of IFNs are involved in psoriasis may provide a better notion for future research. This review focuses on the involvement of three types of IFNs in psoriasis and related therapeutic investigations, briefly describing the three IFNs' production and signaling, as well as the dual effects of IFNs on the skin. It is intended that it would serve as a model for future research.
Collapse
Affiliation(s)
- Jiaming He
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Minghui Zhao
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoyu Ma
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Dilong Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyan Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Fan Yang
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
47
|
Hegewisch-Solloa E, Nalin AP, Freud AG, Mace EM. Deciphering the localization and trajectory of human natural killer cell development. J Leukoc Biol 2023; 114:487-506. [PMID: 36869821 DOI: 10.1093/jleuko/qiad027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 03/05/2023] Open
Abstract
Innate immune cells represent the first line of cellular immunity, comprised of both circulating and tissue-resident natural killer cells and innate lymphoid cells. These innate lymphocytes arise from a common CD34+ progenitor that differentiates into mature natural killer cells and innate lymphoid cells. The successive stages in natural killer cell maturation are characterized by increased lineage restriction and changes to phenotype and function. Mechanisms of human natural killer cell development have not been fully elucidated, especially the role of signals that drive the spatial localization and maturation of natural killer cells. Cytokines, extracellular matrix components, and chemokines provide maturation signals and influence the trafficking of natural killer cell progenitors to peripheral sites of differentiation. Here we present the latest advances in our understanding of natural killer and innate lymphoid cell development in peripheral sites, including secondary lymphoid tissues (i.e. tonsil). Recent work in the field has provided a model for the spatial distribution of natural killer cell and innate lymphoid cell developmental intermediates in tissue and generated further insights into the developmental niche. In support of this model, future studies using multifaceted approaches seek to fully map the developmental trajectory of human natural killer cells and innate lymphoid cells in secondary lymphoid tissues.
Collapse
Affiliation(s)
- Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| | - Ansel P Nalin
- Biomedical Sciences Graduate Program, Medical Scientist Training Program, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave. Columbus, OH 43210, USA
| | - Aharon G Freud
- Department of Pathology, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 12th Ave. Columbus, OH 43210, USA
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| |
Collapse
|
48
|
Samuelson D, Villageliu D, Cunningham K, Smith D, Knoell D, Mandolfo M, Wyatt T. Regulation of Natural Killer Cell TGF-β and AhR Signaling Pathways Via the Intestinal Microbiota is Critical for Host Defense Against Alcohol-Associated Bacterial Pneumonia. RESEARCH SQUARE 2023:rs.3.rs-3328953. [PMID: 37886455 PMCID: PMC10602187 DOI: 10.21203/rs.3.rs-3328953/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Alcohol use is an independent risk factor for the development of bacterial pneumonia due, in part, to impaired mucus-facilitated clearance, macrophage phagocytosis, and recruitment of neutrophils. Alcohol consumption is also known to reduce peripheral natural killer (NK) cell numbers and compromises NK cell cytolytic activity, especially NK cells with a mature phenotype. However, the role of innate lymphocytes, such as NK cells during host defense against alcohol-associated bacterial pneumonia is essentially unknown. We have previously shown that indole supplementation mitigates increases in pulmonary bacterial burden and improves pulmonary NK cell recruitment in alcohol-fed mice, which were dependent of aryl hydrocarbon receptor (AhR) signaling. Employing a binge-on-chronic alcohol-feeding model we sought to define the role and interaction of indole and NK cells during pulmonary host defense against alcohol-associated pneumonia. We demonstrate that alcohol dysregulates NK cell effector function and pulmonary recruitment via alterations in two key signaling pathways. We found that alcohol increases transforming growth factor beta (TGF-β) signaling, while suppressing AhR signaling. We further demonstrated that NK cells isolated from alcohol-fed mice have a reduced ability to kill Klebsiella pneumoniae. NK cell migratory capacity to chemokines was also significantly altered by alcohol, as NK cells isolated from alcohol-fed mice exhibited preferential migration in response to CXCR3 chemokines but exhibited reduced migration in response to CCR2, CXCR4, and CX3CR1 chemokines. Together this data suggests that alcohol disrupts NK cell specific TGF-β and AhR signaling pathways leading to decreased pulmonary recruitment and cytolytic activity thereby increasing susceptibility to alcohol-associated bacterial pneumonia.
Collapse
|
49
|
Koprivica I, Stanisavljević S, Mićanović D, Jevtić B, Stojanović I, Miljković Đ. ILC3: a case of conflicted identity. Front Immunol 2023; 14:1271699. [PMID: 37915588 PMCID: PMC10616800 DOI: 10.3389/fimmu.2023.1271699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Innate lymphoid cells type 3 (ILC3s) are the first line sentinels at the mucous tissues, where they contribute to the homeostatic immune response in a major way. Also, they have been increasingly appreciated as important modulators of chronic inflammatory and autoimmune responses, both locally and systemically. The proper identification of ILC3 is of utmost importance for meaningful studies on their role in immunity. Flow cytometry is the method of choice for the detection and characterization of ILC3. However, the analysis of ILC3-related papers shows inconsistency in ILC3 phenotypic definition, as different inclusion and exclusion markers are used for their identification. Here, we present these discrepancies in the phenotypic characterization of human and mouse ILC3s. We discuss the pros and cons of using various markers for ILC3 identification. Furthermore, we consider the possibilities for the efficient isolation and propagation of ILC3 from different organs and tissues for in-vitro and in-vivo studies. This paper calls upon uniformity in ILC3 definition, isolation, and propagation for the increased possibility of confluent interpretation of ILC3's role in immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
50
|
Liang B, Xing D. Unveiling the mystery of ILC3s: Their functions and interactions in mucosal immunity. Int Immunopharmacol 2023; 123:110772. [PMID: 37552906 DOI: 10.1016/j.intimp.2023.110772] [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: 03/26/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
Innate lymphoid cells (ILCs) are a recently discovered subset of immune cells that play a crucial role in preserving tissue health and combating infections. Among these, ILC3s are particularly vital in regulating mucosal immunity across multiple organs such as the gut, lungs, and skin. The purpose of this article is to present a comprehensive and detailed overview of current knowledge on ILC3s, with a specific emphasis on their intricate interactions with various components of the intestinal microenvironment. Recent research on the complex, bidirectional communication pathways between ILC3s and intestinal epithelial cells, stromal cells, immune cells, microbiota, their metabolites, and diet are highlighted. Furthermore, this review comprehensively examines the diverse functions of ILC3s, which include lymphoid tissue development, tissue repair, infection, inflammation, and metabolic diseases, as well as the effector molecules that facilitate these functions. Overall, this review provides valuable insights into the biological and functional aspects of ILC3s and underscores their potential for developing innovative therapies for immune-mediated disorders, while also acknowledging the remaining knowledge gaps and challenges that need to be addressed.
Collapse
Affiliation(s)
- Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|