1
|
Churchill MJ, Pandeya A, Bauer R, Christopher T, Krug S, Honodel R, Smita S, Warner L, Mooney BM, Gibson AR, Mitchell PS, Tait Wojno ED, Rauch I. Enteric tuft cell inflammasome activation drives NKp46+ILC3 IL22 via PGD2 and inhibits Salmonella. J Exp Med 2025; 222:e20230803. [PMID: 40079814 PMCID: PMC11905811 DOI: 10.1084/jem.20230803] [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: 05/11/2023] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
To distinguish pathogens from commensals, the intestinal epithelium employs cytosolic innate immune sensors. Activation of the NAIP-NLRC4 inflammasome initiates extrusion of infected intestinal epithelial cells (IEC) upon cytosolic bacterial sensing. We previously reported that activation of the inflammasome in tuft cells, which are primarily known for their role in parasitic infections, leads to the release of prostaglandin D2 (PGD2). We observe that NAIP-NLRC4 inflammasome activation in tuft cells leads to an antibacterial response with increased IL-22 and antimicrobial protein levels within the small intestine, which is dependent on PGD2 signaling. A NKp46+ subset of ILC3 expresses the PGD2 receptor CRTH2 and is the source of the increased IL-22. Inflammasome activation in tuft cells also leads to better control of Salmonella Typhimurium in the distal small intestine. However, tuft cells in the cecum and colon are dispensable for antibacterial immunity. These data support that intestinal tuft cells can also induce antibacterial responses, possibly in a tissue-specific manner.
Collapse
Affiliation(s)
- Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Ankit Pandeya
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Renate Bauer
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Tighe Christopher
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Stefanie Krug
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Roslyn Honodel
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Shuchi Smita
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Lindsey Warner
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Bridget M. Mooney
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Alexis R. Gibson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Patrick S. Mitchell
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | | | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
2
|
Rodrigues PF, Wu S, Trsan T, Panda SK, Fachi JL, Liu Y, Du S, de Oliveira S, Antonova AU, Khantakova D, Sudan R, Desai P, Diamond MS, Gilfillan S, Anderson SK, Cella M, Colonna M. Rorγt-positive dendritic cells are required for the induction of peripheral regulatory T cells in response to oral antigens. Cell 2025; 188:2720-2737.e22. [PMID: 40185101 DOI: 10.1016/j.cell.2025.03.020] [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: 08/26/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
The intestinal immune system maintains tolerance to harmless food proteins and gut microbiota through peripherally derived RORγt+ Tregs (pTregs), which prevent food intolerance and inflammatory bowel disease. Recent studies suggested that RORγt+ antigen-presenting cells (APCs), which encompass rare dendritic cell (DC) subsets and type 3 innate lymphoid cells (ILC3s), are key to pTreg induction. Here, we developed a mouse with reduced RORγt+ APCs by deleting a specific cis-regulatory element of Rorc encoding RORγt. Single-cell RNA sequencing and flow cytometry analyses confirmed the depletion of a RORγt+ DC subset and ILC3s. These mice showed a secondary reduction in pTregs, impaired tolerance to oral antigens, and an increase in T helper (Th)2 cells. Conversely, ILC3-deficient mice showed no pTregs or Th2 cell abnormalities. Lineage tracing revealed that RORγt+ DCs share a lymphoid origin with ILC3s, consistent with their similar phenotypic traits. These findings highlight the role of lymphoid RORγt+ DCs in maintaining intestinal immune balance and preventing conditions like food allergies.
Collapse
Affiliation(s)
- Patrick Fernandes Rodrigues
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Shitong Wu
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Santosh K Panda
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - José Luís Fachi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Yizhou Liu
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Siling Du
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Sarah de Oliveira
- Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Darya Khantakova
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Raki Sudan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA; Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Stephen K Anderson
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|
3
|
Xiong L, Diwakarla S, Chatzis R, Artaiz O, Macowan M, Zhang S, Garnham A, Morgan PK, Mellett NA, Meikle PJ, Lancaster GI, Marsland BJ, Nutt SL, Seillet C. Acute exposure to high-fat diet impairs ILC3 functions and gut homeostasis. Immunity 2025; 58:1185-1200.e8. [PMID: 40233759 DOI: 10.1016/j.immuni.2025.03.017] [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: 07/04/2024] [Revised: 12/17/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Prolonged exposure to a high-fat diet (HFD) exacerbates intestinal disease pathology, yet the early events preceding the development of gut inflammation remain poorly understood. Here, we show that within 48 h, HFD impairs intestinal group 3 innate lymphoid cells (ILC3s) and their capacity to produce interleukin-22 (IL-22), critical for maintaining gut homeostasis. This loss of function was associated with rapid dysbiosis, increased gut permeability, and reduced production of antimicrobial peptides, mucus, and tight-junction proteins. While saturated fatty acids metabolized through oxidation impaired ILC3 function, unsaturated fatty acids sustained IL-22 secretion by ILC3s through the formation of lipid droplets using diacylglycerol O-acyltransferase (DGAT) enzymes. Upon inflammation, saturated fatty acids impaired IL-22 production by ILC3s and increased the susceptibility of the gut to injury. Our findings reveal the differential acute impact of saturated and unsaturated fatty acids on gut homeostasis through distinct metabolic pathways in ILC3s.
Collapse
Affiliation(s)
- Le Xiong
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shanti Diwakarla
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Roxanne Chatzis
- Department of Immunology, University of Monash, Melbourne, Melbourne, VIC 3004, Australia
| | - Olivia Artaiz
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Matthew Macowan
- Department of Immunology, University of Monash, Melbourne, Melbourne, VIC 3004, Australia
| | - Shengbo Zhang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexandra Garnham
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
| | - Graeme I Lancaster
- Department of Immunology, University of Monash, Melbourne, Melbourne, VIC 3004, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Benjamin J Marsland
- Department of Immunology, University of Monash, Melbourne, Melbourne, VIC 3004, Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cyril Seillet
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Immunology, University of Monash, Melbourne, Melbourne, VIC 3004, Australia.
| |
Collapse
|
4
|
Sun L, Spiteri AG, Griffith BD, Zhang Y, Di Magliano MP, Olivei AC, McGue JJ, Edwards J, Frankel TL. IL-22BP Modulates Injury in Acute Pancreatitis but Delays Tissue Recovery in Chronic Pancreatitis. Cell Mol Gastroenterol Hepatol 2025:101520. [PMID: 40274099 DOI: 10.1016/j.jcmgh.2025.101520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND & AIMS In acute pancreatitis, interleukin (IL)-22 signaling is increased, whereas overall expression of the cytokine paradoxically drops, suggesting an additional level of control. Here, we investigate the regulation of IL-22 signaling by its soluble neutralizing receptor interleukin-22 binding protein (IL-22BP) in the context of both acute and chronic pancreatitis. METHODS Cerulein was used to induce acute and chronic pancreatitis in both wild-type mice and IL-22BP knockout mice. Histology, multiplex immunofluorescence and flow cytometry were performed to compare differences in tissue injury, recovery, fibrosis, and inflammation at various times of recovery. RESULTS Loss of IL-22BP resulted in increased canonical IL-22 signaling and the expression of the anti-autophagy protein Bcl-XL. This was associated with decreased severity of acute pancreatitis, as evidenced by lower serum amylase and tissue injury. In chronic pancreatitis, IL-22BP expression was induced in the inflammatory and recovery phases and genetic deletion resulted in unchecked IL-22 signaling, as demonstrated by persistent p-Stat3 signaling and proliferation of both epithelial cells and fibroblasts. Loss of IL-22BP increased myeloid cell infiltration, which persisted throughout recovery. Mechanistically, IL-22 activity forced persistent acinar to ductal metaplasia and delayed tissue recovery. CONCLUSIONS IL-22BP plays an important role in modulating IL-22 activity during tissue injury and recovery after pancreatitis. Loss of IL-22BP attenuated acute pancreatitis but promoted chronic fibrosis and inflammation through uncontrolled IL-22 signaling and subsequent deleterious effects on epithelial cells, fibroblasts, and immune infiltration.
Collapse
Affiliation(s)
- Lei Sun
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Andrew G Spiteri
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Brian D Griffith
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Marina Pasca Di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Alberto C Olivei
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jake J McGue
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jacob Edwards
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
5
|
Roberts LB, Kelly AM, Hepworth MR. There's no place like home: How local tissue microenvironments shape the function of innate lymphoid cells. Mucosal Immunol 2025; 18:279-289. [PMID: 39900201 DOI: 10.1016/j.mucimm.2025.01.012] [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: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Innate lymphoid cells (ILC) have emerged as critical immune effectors with key roles in orchestrating the wider immune response. While ILC are relatively rare cells they are found enriched within discrete microenvironments, predominantly within barrier tissues. An emerging body of evidence implicates complex and multi-layered interactions between cell types, tissue structure and the external environment as key determinants of ILC function within these niches. In this review we will discuss the specific components that constitute ILC-associated microenvironments and consider how they act to determine health and disease. The development of holistic, integrated models of ILC function within complex tissue environments will inform new understanding of the contextual cues and mechanisms that determine the protective versus disease-causing roles of this immune cell family.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Alanna M Kelly
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Matthew R Hepworth
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom.
| |
Collapse
|
6
|
Cherrier M, Teo TH, Corrêa RO, Picard M, Couesnon A, Lebreton C, Carbone F, Masson C, Schnupf P, Cerf-Bensussan N, Gaboriau-Routhiau V. Hematopoietic MyD88 orchestrates the control of gut colonization by segmented filamentous bacteria. Mucosal Immunol 2025:S1933-0219(25)00028-5. [PMID: 40090466 DOI: 10.1016/j.mucimm.2025.03.002] [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: 04/26/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
Host-microbiota cooperation is critical for successful intestinal homeostasis. The commensal segmented filamentous bacteria (SFB) are crucial for orchestrating the post-natal maturation of the host gut immune system and establishing a healthy state of physiological inflammation, which largely depends on their intimate attachment to the ileal mucosa. However, the signaling pathways used by SFB to induce gut immune responses and how such responses ultimately control SFB colonization remain controversial. Using gnotobiotic approaches, we showed that SFB load is controlled by complex interactions involving the gut microbiota and the host immune system. Therefore, to clearly determine the role of host immune responses induced by SFB in directly controlling their growth, immunodeficient mice monocolonized with SFB were used. Here, we show that in the absence of a complex microbiota, the humoral immune response is dispensable to control SFB growth in the jejunum and ileum, shortly and later after colonization. In contrast, MyD88 signaling in myeloid cells is critical for licensing interleukin (IL)-22 production by type 3 innate lymphoid cells (ILC3) and CD4+ T cells, which ultimately limits SFB expansion. Thus, by revisiting the hierarchy of immune mechanisms that directly control SFB growth, our results emphasize the necessary and sufficient role of a hematopoietic MyD88/IL-22 axis.
Collapse
Affiliation(s)
- Marie Cherrier
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Teck Hui Teo
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France; A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore
| | - Renan Oliveira Corrêa
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Marion Picard
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Aurélie Couesnon
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Corinne Lebreton
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Francesco Carbone
- Université Paris Cité, Imagine Institute, INSERM UMR 1163, Labtech Single-Cell@Imagine, 75015 Paris, France
| | - Cécile Masson
- Université Paris Cité, Imagine Institute, Structure Fédérative de Recherche Necker, Bioinformatics Core Facility, 75015 Paris, France
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Valérie Gaboriau-Routhiau
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| |
Collapse
|
7
|
Wils P, Habibi Kavashkohie MR, Sélos Guerra F, Landais S, Rubio M, Mehta H, Sarfati M, Chapuy L. Single-Cell Transcriptomic Profile of Innate Cell Populations in Mesenteric Lymph Nodes of Inflammatory Bowel Disease Patients. Inflamm Bowel Dis 2025:izaf017. [PMID: 39982469 DOI: 10.1093/ibd/izaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND AIMS Innate immune cells, including dendritic cells (DCs), monocytes (Mono), macrophages (Mac), natural killer (NK), and innate lymphoid cells (ILC), contribute to chronic inflammation in lymphoid tissues. Here, we characterized the innate immune cell landscape in inflamed mesenteric lymph nodes (MLNs) of patients with inflammatory bowel diseases (IBD) at the single-cell level. METHODS Surgically resected colonic MLNs were obtained from patients with Crohn's disease (CD; n = 3), ulcerative colitis (UC; n = 3), non-inflamed UC (n = 1), and non-IBD (n = 2). CD45+CD3-CD19- non-T/non-B cells were FACS-sorted to capture rare innate immune cells. Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) was performed on the BD Rhapsody platform alongside multiparameter flow cytometry staining. RESULTS CITE-seq analysis unveiled the molecular signature of 11 Mono/Mac/DC (MMDC) and 7 NK/ILC enriched clusters in human MLNs. DC clusters included 3 newly characterized DC clusters such as CD1c/CD163/VCAN/CD64-expressing DC3; AXL-expressing DCs; and a CD103+ DC subset, expressing LTB, S100B, and IL22RA2 (encoding IL22BP). Mono/Mac clusters comprised inflammatory monocytes, which accumulated in IBD compared to non-IBD MLNs. Among NK/ILC clusters, we identified a cytotoxic ILC subset (IL7R, KLRD1, GNLY), previously not reported in MLNs, reminiscent of cytotoxic ILC1-like cells found in inflamed gut mucosa. CONCLUSION CITE-seq and flow-cytometry analyses of colonic MLNs from patients with active IBD reveal the molecular signature and cell distribution of previously uncharacterized DC and ILC subpopulations in human MLNs. These findings expand our understanding of immune responses during chronic inflammation in IBD.
Collapse
Affiliation(s)
- Pauline Wils
- Hepato-Gastroenterology Department, Claude Huriez Hospital, University of Lille 2, 59000 Lille, France
- INFINITE, University of Lille, INSERM U1286-Institute for Translational Research in Inflammation, 59000 Lille, France
| | | | - Fabiana Sélos Guerra
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
| | - Séverine Landais
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Heena Mehta
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
| | - Laurence Chapuy
- Department of Pediatrics, Centre de Recherche du CHU Sainte-Justine, Université de Montréal, H3T 1C5 Montréal, Québec, Canada
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CR-CHUM), Université de Montréal, H2X 0A9 Montréal, Québec, Canada
- Department of Pediatrics, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, H4A 3J1 Montreal, Quebec, Canada
| |
Collapse
|
8
|
Kanaya T, Jinnohara T, Sakakibara S, Tachibana N, Sasaki T, Kato T, Riemann M, Jin J, Shiroguchi K, Kawakami E, Ohno H. RelB and C/EBPα critically regulate the development of Peyer's patch mononuclear phagocytes. Mucosal Immunol 2025; 18:151-161. [PMID: 39413971 DOI: 10.1016/j.mucimm.2024.10.005] [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: 12/28/2023] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
To establish protection against harmful foreign antigens, the small intestine harbors guardian sites called Peyer's patches (PPs). PPs take up antigens through microfold (M) cells and transfer them to the sub-epithelial dome (SED), which contains a high density of mononuclear phagocytes (MPs), for T cell-priming. Accumulating evidence indicates that SED-MPs have unique functions other than T cell-priming to facilitate mucosal immune responses; however, the crucial factors regulating the functions of SED-MPs have not been determined. Here we performed transcriptome analysis, and identified the gene signatures of SED-MPs. Further data interpretation with transcription factor (TF) enrichment analysis estimated TFs responsible for the functions of SED-MPs. Among them, we found that RelB and C/EBPα were preferentially activated in SED-MPs. RelB-deficiency silenced the expression of IL-22BP and S100A4 by SED-MPs. On the other hand, C/EBPα-deficiency decreased the expression of lysozyme by SED-MPs, resulting the increased invasion of orally administered pathogenic bacteria into PPs and mesenteric lymph nodes. Our findings thus demonstrate that RelB and C/EBPα are essential to regulate the functions of SED-MPs.
Collapse
Affiliation(s)
- Takashi Kanaya
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan.
| | - Toshi Jinnohara
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Sayuri Sakakibara
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Naoko Tachibana
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Takaharu Sasaki
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Marc Riemann
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Jianshi Jin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Katsuyuki Shiroguchi
- Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Eiryo Kawakami
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Yokohama, Kanagawa, Japan; Department of Artificial intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan.
| |
Collapse
|
9
|
Dudziak D, Heger L, Agace WW, Bakker J, de Gruijl TD, Dress RJ, Dutertre C, Fenton TM, Fransen MF, Ginhoux F, Heyman O, Horev Y, Hornsteiner F, Kandiah V, Kles P, Lubin R, Mizraji G, Prokopi A, Saar O, Sopper S, Stoitzner P, Strandt H, Sykora MM, Toffoli EC, Tripp CH, van Pul K, van de Ven R, Wilensky A, Yona S, Zelle‐Rieser C. Guidelines for preparation and flow cytometry analysis of human nonlymphoid tissue DC. Eur J Immunol 2025; 55:e2250325. [PMID: 39668411 PMCID: PMC11739683 DOI: 10.1002/eji.202250325] [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: 12/09/2022] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 12/14/2024]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs, and various nonlymphoid tissues. Within this article, detailed protocols are presented that allow for the generation of single-cell suspensions from human nonlymphoid tissues including lung, skin, gingiva, intestine as well as from tumors and tumor-draining lymph nodes with a subsequent analysis of dendritic cells by flow cytometry. Further, prepared single-cell suspensions can be subjected to other applications including cellular enrichment procedures, RNA sequencing, functional assays, etc. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Diana Dudziak
- Institute of ImmunologyJena University HospitalFriedrich‐Schiller‐UniversityJenaGermany
- Laboratory of Dendritic Cell BiologyDepartment of DermatologyUniversity Hospital ErlangenErlangenGermany
| | - Lukas Heger
- Laboratory of Dendritic Cell BiologyDepartment of DermatologyUniversity Hospital ErlangenErlangenGermany
- Department of Transfusion Medicine and HemostaseologyUniversity Hospital ErlangenErlangenGermany
| | - William W Agace
- LEO Foundation Skin Immunology Research CenterDepartment of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- Immunology SectionLund UniversityLundSweden
| | - Joyce Bakker
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Tanja D. de Gruijl
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Regine J. Dress
- Institute of Systems ImmunologyHamburg Center for Translational Immunology (HCTI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | | | | | - Marieke F. Fransen
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Department of Pulmonary DiseasesAmsterdam UMC location Vrije UniversiteitAmsterdamThe Netherlands
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and ResearchSingaporeSingapore
- Department of Immunology and MicrobiologyShanghai Institute of ImmunologyShanghai Jiao Tong University School of MedicineShanghaiChina
- SingHealth Duke‐NUS Academic Medical CentreTranslational Immunology InstituteSingaporeSingapore
- INSERM U1015, Gustave Roussy Cancer CampusVillejuifFrance
| | - Oded Heyman
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Yael Horev
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Florian Hornsteiner
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Vinitha Kandiah
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Paz Kles
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Ruth Lubin
- Faculty of Dental MedicineThe Institute of Biomedical and Oral ResearchHebrew University of JerusalemIsrael
| | - Gabriel Mizraji
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Anastasia Prokopi
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Or Saar
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Sieghart Sopper
- Internal Medicine V, Hematology and OncologyMedical University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research CenterInnsbruckAustria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Helen Strandt
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Martina M Sykora
- Internal Medicine V, Hematology and OncologyMedical University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research CenterInnsbruckAustria
| | - Elisa C. Toffoli
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Christoph H. Tripp
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Kim van Pul
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Rieneke van de Ven
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
- Department of Otolaryngology, Head and Neck SurgeryAmsterdam UMC location Vrije UniversiteitAmsterdamThe Netherlands
| | - Asaf Wilensky
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Simon Yona
- Faculty of Dental MedicineThe Institute of Biomedical and Oral ResearchHebrew University of JerusalemIsrael
| | - Claudia Zelle‐Rieser
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
10
|
Ohara D, Takeuchi Y, Hirota K. Type 17 immunity: novel insights into intestinal homeostasis and autoimmune pathogenesis driven by gut-primed T cells. Cell Mol Immunol 2024; 21:1183-1200. [PMID: 39379604 PMCID: PMC11528014 DOI: 10.1038/s41423-024-01218-x] [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: 04/15/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
The IL-23 signaling pathway in both innate and adaptive immune cells is vital for orchestrating type 17 immunity, which is marked by the secretion of signature cytokines such as IL-17, IL-22, and GM-CSF. These proinflammatory mediators play indispensable roles in maintaining intestinal immune equilibrium and mucosal host defense; however, their involvement has also been implicated in the pathogenesis of chronic inflammatory disorders, such as inflammatory bowel diseases and autoimmunity. However, the implications of type 17 immunity across diverse inflammation models are complex. This review provides a comprehensive overview of the multifaceted roles of these cytokines in maintaining gut homeostasis and in perturbing gut barrier integrity, leading to acute and chronic inflammation in various models of gut infection and colitis. Additionally, this review focuses on type 17 immunity interconnecting multiple organs in autoimmune conditions, with a particular emphasis on the pathogenesis of autoimmune arthritis and neuroinflammation driven by T cells primed within the gut microenvironment.
Collapse
Affiliation(s)
- Daiya Ohara
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- ImmunoSensation Cluster of Excellence, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Nguyen HH, Talbot J, Li D, Raghavan V, Littman DR. Modulating intestinal neuroimmune VIPergic signaling attenuates the reduction in ILC3-derived IL-22 and hepatic steatosis in MASLD. Hepatol Commun 2024; 8:e0528. [PMID: 39761015 PMCID: PMC11495769 DOI: 10.1097/hc9.0000000000000528] [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: 05/01/2024] [Accepted: 07/18/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD, formerly known as NAFLD) is a major driver of cirrhosis and liver-related mortality. However, therapeutic options for MASLD, including prevention of liver steatosis, are limited. We previously described that vasoactive intestinal peptide-producing neurons (VIP-neurons) regulate the efficiency of intestinal dietary fat absorption and IL-22 production by type 3 innate lymphoid cells (ILC3) in the intestine. Given the described hepatoprotective role of IL-22, we hypothesize that modulation of this neuroimmune circuit could potentially be an innovative approach for the control of liver steatosis. METHODS We used a model of diet-induced MASLD by exposing mice to a high-fat diet (HFD) for 16 weeks, when the development of liver steatosis was first observed in our animals. We characterized IL-22 production by intestinal ILC3 at this dietary endpoint. We then evaluated whether communication between VIP-neurons and ILC3 affected IL-22 production and MASLD development by exposing mice with a conditional genetic deletion of Vipr2 in ILC3 (Rorc(t)CreVipr2fl/fl) to the HFD. We also performed intermittent global inhibition of VIP-neurons using a chemogenetic inhibitory approach (VipIres-CrehM4DiLSL) in HFD-fed mice. RESULTS Production of IL-22 by intestinal ILC3 is reduced in steatotic mice that were exposed to an HFD for 16 weeks. Targeted deletion of VIP receptor 2 in ILC3 resulted in higher production of IL-22 in ILC3 and was associated with a significant reduction in liver steatosis in mice under HFD. Global inhibition of VIP-producing neurons also resulted in a significant reduction in liver steatosis. CONCLUSIONS Modulating VIPergic neuroimmune signaling can ameliorate the development of hepatic steatosis induced by a surplus of fat ingestion in the diet. This neuroimmune pathway should be further investigated as a potential therapeutic avenue in MASLD.
Collapse
Affiliation(s)
- Henry H. Nguyen
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Department of Medicine and Department of Microbiology, Immunology, and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jhimmy Talbot
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Dayi Li
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Varsha Raghavan
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Howard Hughes Medical Institute, New York, New York, USA
| |
Collapse
|
12
|
Gogoleva VS, Drutskaya MS, Vorontsov AI, Atretkhany KSN, Belogurov AA, Kruglov AA, Nedospasov SA. Lymphotoxins from distinct types of lymphoid cells differentially contribute to neuroinflammation. Eur J Immunol 2024; 54:e2350977. [PMID: 39210647 DOI: 10.1002/eji.202350977] [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: 12/30/2023] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Lymphotoxin α and lymphotoxin β (LTs), TNF superfamily members, are expressed in either soluble (LTα3) or membrane-bound (LTα1β2 or LTα2β1) forms. In the pathological context, LT-mediated signaling is known to exacerbate autoimmunity by perpetuating inflammation and promoting the formation of tertiary lymphoid organs. Despite this understanding, the exact roles of LTα and LTβ in the pathogenesis of the murine model of multiple sclerosis, and experimental autoimmune encephalomyelitis (EAE), remain controversial. Here, we employed a panel of gene-modified mice with cell-type restricted ablation of LTα (targeting both membrane-bound and soluble forms of LTs) to unravel the contributions of LTs from various lymphoid cells, namely T cells, type 3 innate lymphoid cells (ILC3) and B cells, in EAE. We found that the effects of LTα deletion were dependent on the cellular source. ILC3-derived lymphotoxins exerted a protective role in EAE by regulating the accumulation of IFN-ɣ- and GM-CSF-producing TH cells in the CNS. In contrast, T-cell-derived lymphotoxins promoted IL-17A- and GM-CSF-mediated TH responses in the periphery, whereas B-cell-derived lymphotoxins were pathogenic only in the autoantibody-mediated EAE model. Collectively, our findings unveil the multifaceted involvement of lymphotoxins in EAE pathogenesis and challenge the view that lymphotoxins play a solely pathogenic role in neuroinflammation.
Collapse
Affiliation(s)
- Violetta S Gogoleva
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
| | - Marina S Drutskaya
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius, Krasnodarsky Krai, 354349, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alexander I Vorontsov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Kamar-Sulu N Atretkhany
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexey A Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Andrey A Kruglov
- AG Chronic Inflammation, German Rheumatism Research Center, a Leibniz Institute, Berlin, 10117, Germany
| | - Sergei A Nedospasov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius, Krasnodarsky Krai, 354349, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
13
|
Geiselhöringer AL, Kolland D, Patt AJ, Hammann L, Köhler A, Kreft L, Wichmann N, Hils M, Ruedl C, Riemann M, Biedermann T, Anz D, Diefenbach A, Voehringer D, Schmidt-Weber CB, Straub T, Pasztoi M, Ohnmacht C. Dominant immune tolerance in the intestinal tract imposed by RelB-dependent migratory dendritic cells regulates protective type 2 immunity. Nat Commun 2024; 15:9143. [PMID: 39443450 PMCID: PMC11500181 DOI: 10.1038/s41467-024-53112-9] [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: 09/22/2023] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Dendritic cells (DCs) are crucial for initiating protective immune responses and have also been implicated in the generation and regulation of Foxp3+ regulatory T cells (Treg cells). Here, we show that in the lamina propria of the small intestine, the alternative NF-κB family member RelB is necessary for the differentiation of cryptopatch and isolated lymphoid follicle-associated DCs (CIA-DCs). Moreover, single-cell RNA sequencing reveals a RelB-dependent signature in migratory DCs in mesenteric lymph nodes favoring DC-Treg cell interaction including elevated expression and release of the chemokine CCL22 from RelB-deficient conventional DCs (cDCs). In line with the key role of CCL22 to facilitate DC-Treg cell interaction, RelB-deficient DCs have a selective advantage to interact with Treg cells in an antigen-specific manner. In addition, DC-specific RelB knockout animals show increased total Foxp3+ Treg cell numbers irrespective of inflammatory status. Consequently, DC-specific RelB knockout animals fail to mount protective Th2-dominated immune responses in the intestine after infection with Heligmosomoides polygyrus bakeri. Thus, RelB expression in cDCs acts as a rheostat to establish a tolerogenic set point that is maintained even during strong type 2 immune conditions and thereby is a key regulator of intestinal homeostasis.
Collapse
Affiliation(s)
- Anna-Lena Geiselhöringer
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Arisha Johanna Patt
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Linda Hammann
- Division of Clinical Pharmacology, LMU University Hospital, LMU, Munich, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Luisa Kreft
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076, Tuebingen, Germany
| | - Nina Wichmann
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Marc Riemann
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745, Jena, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, LMU University Hospital, LMU, Munich, Germany
- Department of Medicine II, LMU University Hospital, LMU, Munich, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, 12203, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, 91054, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Tobias Straub
- Bioinformatics Core Unit, Biomedical Center, Ludwig-Maximilians-University, 82152, Planegg, Germany
| | - Maria Pasztoi
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
14
|
Hunzeker ZE, Zhao L, Kim AM, Parker JM, Zhu Z, Xiao H, Bai Q, Wakefield MR, Fang Y. The role of IL-22 in cancer. Med Oncol 2024; 41:240. [PMID: 39231878 DOI: 10.1007/s12032-024-02481-8] [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: 06/23/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024]
Abstract
Interleukin-22, discovered in the year of 2000, is a pleiotropic Th17 cytokine from the IL-10 family of cytokines. IL-22 signals through the type 2 cytokine receptor complex IL-22R and predominantly activates STAT3. This pathway leads to the transcription of several different types of genes, giving IL-22 context-specific functions ranging from inducing antimicrobial peptide expression to target cell proliferation. In recent years, it has been shown that IL-22 is involved in the pathogenesis of neoplasia in some cancers through its pro-proliferative and anti-apoptotic effects. This review highlights studies with recent discoveries and conclusions drawn on IL-22 and its involvement and function in various cancers. Such a study may be helpful to better understand the role of IL-22 in cancer so that new treatment could be developed targeting IL-22.
Collapse
Affiliation(s)
- Zachary E Hunzeker
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Department of Internal Medicine, University of Texas Houston Health Science Center, Houston, TX, USA
| | - Lei Zhao
- Department of Respiratory Medicine, the 2nd People's Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Austin M Kim
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Jacob M Parker
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
| | - Ziwen Zhu
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Huaping Xiao
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, 50312, USA.
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
15
|
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
|
16
|
Nelius E, Fan Z, Sobecki M, Krzywinska E, Nagarajan S, Ferapontova I, Gotthardt D, Takeda N, Sexl V, Stockmann C. The transcription factor HIF-1α in NKp46+ ILCs limits chronic intestinal inflammation and fibrosis. Life Sci Alliance 2024; 7:e202402593. [PMID: 38876796 PMCID: PMC11178940 DOI: 10.26508/lsa.202402593] [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: 01/13/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
Innate lymphoid cells (ILCs) are critical for intestinal adaptation to microenvironmental challenges, and the gut mucosa is characterized by low oxygen. Adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs), and the HIF-1α subunit shapes an ILC phenotype upon acute colitis that contributes to intestinal damage. However, the impact of HIF signaling in NKp46+ ILCs in the context of repetitive mucosal damage and chronic inflammation, as it typically occurs during inflammatory bowel disease, is unknown. In chronic colitis, mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in NKp46+ ILC1s but a concomitant rise in neutrophils and Ly6Chigh macrophages. Single-nucleus RNA sequencing suggests enhanced interaction of mesenchymal cells with other cell compartments in the colon of HIF-1α KO mice and a loss of mucus-producing enterocytes and intestinal stem cells. This was, furthermore, associated with increased bone morphogenetic pathway-integrin signaling, expansion of fibroblast subsets, and intestinal fibrosis. In summary, this suggests that HIF-1α-mediated ILC1 activation, although detrimental upon acute colitis, protects against excessive inflammation and fibrosis during chronic intestinal damage.
Collapse
Affiliation(s)
- Eric Nelius
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Zheng Fan
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Michal Sobecki
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | | | | | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | | | - Christian Stockmann
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Zurich, Switzerland
- Zurich Kidney Center, Zurich, Switzerland
| |
Collapse
|
17
|
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
|
18
|
Chen Q, Muñoz AR, Korchagina AA, Shou Y, Vallecer J, Todd AW, Shein SA, Tumanov AV, Koroleva E. LTβR-RelB signaling in intestinal epithelial cells protects from chemotherapy-induced mucosal damage. Front Immunol 2024; 15:1388496. [PMID: 38873613 PMCID: PMC11169669 DOI: 10.3389/fimmu.2024.1388496] [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: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
The intricate immune mechanisms governing mucosal healing following intestinal damage induced by cytotoxic drugs remain poorly understood. The goal of this study was to investigate the role of lymphotoxin beta receptor (LTβR) signaling in chemotherapy-induced intestinal damage. LTβR deficient mice exhibited heightened body weight loss, exacerbated intestinal pathology, increased proinflammatory cytokine expression, reduced IL-22 expression, and proliferation of intestinal epithelial cells following methotrexate (MTX) treatment. Furthermore, LTβR-/-IL-22-/- mice succumbed to MTX treatment, suggesting that LTβR- and IL-22- dependent pathways jointly promote mucosal repair. Although both LTβR ligands LIGHT and LTβ were upregulated in the intestine early after MTX treatment, LIGHT-/- mice, but not LTβ-/- mice, displayed exacerbated disease. Further, we revealed the critical role of T cells in mucosal repair as T cell-deficient mice failed to upregulate intestinal LIGHT expression and exhibited increased body weight loss and intestinal pathology. Analysis of mice with conditional inactivation of LTβR revealed that LTβR signaling in intestinal epithelial cells, but not in Lgr5+ intestinal stem cells, macrophages or dendritic cells was critical for mucosal repair. Furthermore, inactivation of the non-canonical NF-kB pathway member RelB in intestinal epithelial cells promoted MTX-induced disease. Based on these results, we propose a model wherein LIGHT produced by T cells activates LTβR-RelB signaling in intestinal epithelial cells to facilitate mucosal repair following chemotherapy treatment.
Collapse
Affiliation(s)
- Qiangxing Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Gastroenterology, Second Xiangya Hospital, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| | - Amanda R. Muñoz
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Anna A. Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Yajun Shou
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Gastroenterology, Second Xiangya Hospital, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, China
| | - Jensine Vallecer
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Austin W. Todd
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Sergey A. Shein
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V. Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
19
|
Yang H, Cao R, Zhou F, Wang B, Xu Q, Li R, Zhang C, Xu H. The role of Interleukin-22 in severe acute pancreatitis. Mol Med 2024; 30:60. [PMID: 38750415 PMCID: PMC11097471 DOI: 10.1186/s10020-024-00826-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Severe acute pancreatitis (SAP) begins with premature activation of enzymes, promoted by the immune system, triggering a potential systemic inflammatory response that leads to organ failure with increased mortality and a bleak prognosis. Interleukin-22 (IL-22) is a cytokine that may have a significant role in SAP. IL-22, a member of the IL-10 cytokine family, has garnered growing interest owing to its potential tissue-protective properties. Recently, emerging research has revealed its specific effects on pancreatic diseases, particularly SAP. This paper provides a review of the latest knowledge on the role of IL-22 and its viability as a therapeutic target in SAP.
Collapse
Affiliation(s)
- Hongli Yang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P.R. China
| | - Ruofan Cao
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P.R. China
| | - Feifei Zhou
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China
| | - Ben Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China
| | - Qianqian Xu
- Department of Gastroenterology, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Ji'nan, Shandong, 250021, P.R. China
| | - Rui Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P.R. China
| | - ChunHua Zhang
- Shandong First Medical University, Ji'nan, Shandong, 250117, P.R. China
| | - Hongwei Xu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong, 250021, P.R. China.
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, P.R. China.
| |
Collapse
|
20
|
Kloc M, Halasa M, Ghobrial RM. Macrophage niche imprinting as a determinant of macrophage identity and function. Cell Immunol 2024; 399-400:104825. [PMID: 38648700 DOI: 10.1016/j.cellimm.2024.104825] [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: 01/25/2024] [Revised: 03/22/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Macrophage niches are the anatomical locations within organs or tissues consisting of various cells, intercellular and extracellular matrix, transcription factors, and signaling molecules that interact to influence macrophage self-maintenance, phenotype, and behavior. The niche, besides physically supporting macrophages, imposes a tissue- and organ-specific identity on the residing and infiltrating monocytes and macrophages. In this review, we give examples of macrophage niches and the modes of communication between macrophages and surrounding cells. We also describe how macrophages, acting against their immune defensive nature, can create a hospitable niche for pathogens and cancer cells.
Collapse
Affiliation(s)
- Malgorzata Kloc
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Department of Genetics, Houston, TX, USA.
| | - Marta Halasa
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| | - Rafik M Ghobrial
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX, USA; Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| |
Collapse
|
21
|
Ulmert I, Lahl K. IL-23 to see: Gut DCs shine bright in inductive sites. J Exp Med 2024; 221:e20232144. [PMID: 38180806 PMCID: PMC10770875 DOI: 10.1084/jem.20232144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2024] Open
Abstract
The cytokine IL-23 plays important roles in intestinal barrier protection and integrity, but is also linked to chronic inflammation. In this issue of JEM, Ohara et al. (https://doi.org/10.1084/jem.20230923) provide clarity on the much-debated question of which cells produce IL-23.
Collapse
Affiliation(s)
- Isabel Ulmert
- Technical University of Denmark, Kongens Lyngby, Denmark
| | - Katharina Lahl
- Technical University of Denmark, Kongens Lyngby, Denmark
- Lund University, Lund, Sweden
- University of Calgary, Calgary, Canada
| |
Collapse
|
22
|
Ohara D, Takeuchi Y, Watanabe H, Lee Y, Mukoyama H, Ohteki T, Kondoh G, Hirota K. Notch2 with retinoic acid license IL-23 expression by intestinal EpCAM+ DCIR2+ cDC2s in mice. J Exp Med 2024; 221:e20230923. [PMID: 38180443 PMCID: PMC10770806 DOI: 10.1084/jem.20230923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/06/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Despite the importance of IL-23 in mucosal host defense and disease pathogenesis, the mechanisms regulating the development of IL-23-producing mononuclear phagocytes remain poorly understood. Here, we employed an Il23aVenus reporter strain to investigate the developmental identity and functional regulation of IL-23-producing cells. We showed that flagellin stimulation or Citrobacter rodentium infection led to robust induction of IL-23-producing EpCAM+ DCIR2+ CD103- cDC2s, termed cDCIL23, which was confined to gut-associated lymphoid tissues, including the mesenteric lymph nodes, cryptopatches, and isolated lymphoid follicles. Furthermore, we demonstrated that Notch2 signaling was crucial for the development of EpCAM+ DCIR2+ cDC2s, and the combination of Notch2 signaling with retinoic acid signaling controlled their terminal differentiation into cDCIL23, supporting a two-step model for the development of gut cDCIL23. Our findings provide fundamental insights into the developmental pathways and cellular dynamics of IL-23-producing cDC2s at steady state and during pathogen infection.
Collapse
Affiliation(s)
- Daiya Ohara
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoonha Lee
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroki Mukoyama
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Pervizaj-Oruqaj L, Selvakumar B, Ferrero MR, Heiner M, Malainou C, Glaser RD, Wilhelm J, Bartkuhn M, Weiss A, Alexopoulos I, Witte B, Gattenlöhner S, Vadász I, Morty RE, Seeger W, Schermuly RT, Vazquez-Armendariz AI, Herold S. Alveolar macrophage-expressed Plet1 is a driver of lung epithelial repair after viral pneumonia. Nat Commun 2024; 15:87. [PMID: 38167746 PMCID: PMC10761876 DOI: 10.1038/s41467-023-44421-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Influenza A virus (IAV) infection mobilizes bone marrow-derived macrophages (BMDM) that gradually undergo transition to tissue-resident alveolar macrophages (TR-AM) in the inflamed lung. Combining high-dimensional single-cell transcriptomics with complex lung organoid modeling, in vivo adoptive cell transfer, and BMDM-specific gene targeting, we found that transitioning ("regenerative") BMDM and TR-AM highly express Placenta-expressed transcript 1 (Plet1). We reveal that Plet1 is released from alveolar macrophages, and acts as important mediator of macrophage-epithelial cross-talk during lung repair by inducing proliferation of alveolar epithelial cells and re-sealing of the epithelial barrier. Intratracheal administration of recombinant Plet1 early in the disease course attenuated viral lung injury and rescued mice from otherwise fatal disease, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Learta Pervizaj-Oruqaj
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Balachandar Selvakumar
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Maximiliano Ruben Ferrero
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Monika Heiner
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Christina Malainou
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Rolf David Glaser
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus Liebig University, Giessen, Germany
| | - Jochen Wilhelm
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Marek Bartkuhn
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus Liebig University, Giessen, Germany
| | - Astrid Weiss
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Biruta Witte
- Department of General and Thoracic Surgery, University Hospital of Giessen, Giessen, Germany
| | | | - István Vadász
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Rory Edward Morty
- Department of Translational Pulmonology and the Translational Lung Research Center, University Hospital Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Werner Seeger
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Buenos Aires, Argentina
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ralph Theo Schermuly
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ana Ivonne Vazquez-Armendariz
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- University of Bonn, Transdisciplinary Research Area Life and Health, Organoid Biology, Life & Medical Sciences Institute, Bonn, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany.
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.
| |
Collapse
|
24
|
Hildreth AD, Padilla ET, Gupta M, Wong YY, Sun R, Legala AR, O'Sullivan TE. Adipose cDC1s contribute to obesity-associated inflammation through STING-dependent IL-12 production. Nat Metab 2023; 5:2237-2252. [PMID: 37996702 DOI: 10.1038/s42255-023-00934-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Obesity is associated with chronic low-grade white adipose tissue (WAT) inflammation that can contribute to the development of insulin resistance in mammals. Previous studies have identified interleukin (IL)-12 as a critical upstream regulator of WAT inflammation and metabolic dysfunction during obesity. However, the cell types and mechanisms that initiate WAT IL-12 production remain unclear. Here we show that conventional type 1 dendritic cells (cDC1s) are the cellular source of WAT IL-12 during obesity through analysis of mouse and human WAT single-cell transcriptomic datasets, IL-12 reporter mice and IL-12p70 protein levels by enzyme-linked immunosorbent assay. We demonstrate that cDC1s contribute to obesity-associated inflammation by increasing group 1 innate lymphocyte interferon-γ production and inflammatory macrophage accumulation. Inducible depletion of cDC1s increased WAT insulin sensitivity and systemic glucose tolerance during diet-induced obesity. Mechanistically, endocytosis of apoptotic bodies containing self-DNA by WAT cDC1s drives stimulator of interferon genes (STING)-dependent IL-12 production. Together, these results suggest that WAT cDC1s act as critical regulators of adipose tissue inflammation and metabolic dysfunction during obesity.
Collapse
Affiliation(s)
- Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eddie T Padilla
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Meha Gupta
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yung Yu Wong
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan Sun
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Akshara R Legala
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Zhang Y, Gao X, Gao S, Liu Y, Wang W, Feng Y, Pei L, Sun Z, Liu L, Wang C. Effect of gut flora mediated-bile acid metabolism on intestinal immune microenvironment. Immunology 2023; 170:301-318. [PMID: 37317655 DOI: 10.1111/imm.13672] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/28/2023] [Indexed: 06/16/2023] Open
Abstract
According to reports, gut microbiota and metabolites regulate the intestinal immune microenvironment. In recent years, an increasing number of studies reported that bile acids (BAs) of intestinal flora origin affect T helper cells and regulatory T cells (Treg cells). Th17 cells play a pro-inflammatory role and Treg cells usually act in an immunosuppressive role. In this review, we emphatically summarised the influence and corresponding mechanism of different configurations of lithocholic acid (LCA) and deoxycholic acid (DCA) on intestinal Th17 cells, Treg cells and intestinal immune microenvironment. The regulation of BAs receptors G protein-coupled bile acid receptor 1 (GPBAR1/TGR5) and farnesoid X receptor (FXR) on immune cells and intestinal environment are elaborated. Furthermore, the potential clinical applications above were also concluded in three aspects. The above will help researchers better understand the effects of gut flora on the intestinal immune microenvironment via BAs and contribute to the development of new targeted drugs.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xueyan Gao
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuochen Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yudi Feng
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Pei
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
26
|
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
|
27
|
Maes B, Fayazpour F, Catrysse L, Lornet G, Van De Velde E, De Wolf C, De Prijck S, Van Moorleghem J, Vanheerswynghels M, Deswarte K, Descamps B, Vanhove C, Van der Schueren B, Vangoitsenhoven R, Hammad H, Janssens S, Lambrecht BN. STE20 kinase TAOK3 regulates type 2 immunity and metabolism in obesity. J Exp Med 2023; 220:e20210788. [PMID: 37347461 PMCID: PMC10287548 DOI: 10.1084/jem.20210788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 03/31/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023] Open
Abstract
Healthy adipose tissue (AT) contains ST2+ Tregs, ILC2s, and alternatively activated macrophages that are lost in mice or humans on high caloric diet. Understanding how this form of type 2 immunity is regulated could improve treatment of obesity. The STE20 kinase Thousand And One amino acid Kinase-3 (TAOK3) has been linked to obesity in mice and humans, but its precise function is unknown. We found that ST2+ Tregs are upregulated in visceral epididymal white AT (eWAT) of Taok3-/- mice, dependent on IL-33 and the kinase activity of TAOK3. Upon high fat diet feeding, metabolic dysfunction was attenuated in Taok3-/- mice. ST2+ Tregs disappeared from eWAT in obese wild-type mice, but this was not the case in Taok3-/- mice. Mechanistically, AT Taok3-/- Tregs were intrinsically more responsive to IL-33, through higher expression of ST2, and expressed more PPARγ and type 2 cytokines. Thus, TAOK3 inhibits adipose tissue Tregs and regulates immunometabolism under excessive caloric intake.
Collapse
Affiliation(s)
- Bastiaan Maes
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Farzaneh Fayazpour
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Leen Catrysse
- Cellular and Molecular (Patho)Physiology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Guillaume Lornet
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Evelien Van De Velde
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Caroline De Wolf
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sofie De Prijck
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Justine Van Moorleghem
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Manon Vanheerswynghels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Department of Electronics and Information Systems, IBiTech-MEDISIP-Infinity Lab, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Department of Electronics and Information Systems, IBiTech-MEDISIP-Infinity Lab, Ghent University, Ghent, Belgium
| | - Bart Van der Schueren
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Bart N. Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam Netherlands
| |
Collapse
|
28
|
Seth P, Dubey S. IL-22 as a target for therapeutic intervention: Current knowledge on its role in various diseases. Cytokine 2023; 169:156293. [PMID: 37441942 DOI: 10.1016/j.cyto.2023.156293] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/12/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
IL-22 has emerged as a crucial cytokine mediating protective response against pathogens and tissue regeneration. Dysregulated production of IL-22 has been shown to play a pivotal role in the pathogenesis of various diseases like malignant tumours, viral, cardiovascular, allergic and autoimmune disorders. Interleukin 22 belongs to IFN-IL-10 cytokine family. It is a major proinflammatory cytokine secreted by activated Th1 cells (Th22), though can also be secreted by many other immune cells like group 3 innate lymphocytes, γδ T cells, NK cells, NK T cells, and mucosal associated invariant T cells. Th22 cells exclusively release IL-22 but not IL-17 or IFN-γ (as Th1 cells releases IFN-γ along with IL-22 and Th17 cells releases IL-17 along with IL-22) and also express aryl hydrocarbon receptor as the key transcription factor. Th22 cells also exhibit expression of chemokine receptor CCR6 and skin-homing receptors CCR4 and CCR10 indicating the involvement of this subset in bolstering epithelial barrier immunity and promoting secretion of antimicrobial peptides (AMPs) from intestinal epithelial cells. The function of IL-22 is modulated by IL-22 binding protein (binds to IL-22 and inhibits it binding to its cell surface receptor); which serves as a competitor for IL-22R1 chain of IL-22 receptor. The pathogenic and protective nature of the Th22 cells is modulated both by the site of infected tissue and the type of disease pathology. This review aims to discuss key features of IL-22 biology, comparisons between IL and 22 and IFN-γ and its role as a potential immune therapy target in different maladies.
Collapse
Affiliation(s)
- Pranav Seth
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India
| | - Shweta Dubey
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India.
| |
Collapse
|
29
|
Chiaranunt P, Burrows K, Ngai L, Tai SL, Cao EY, Liang H, Hamidzada H, Wong A, Gschwend J, Flüchter P, Kuypers M, Despot T, Momen A, Lim SM, Mallevaey T, Schneider C, Conway T, Imamura H, Epelman S, Mortha A. Microbial energy metabolism fuels an intestinal macrophage niche in solitary isolated lymphoid tissues through purinergic signaling. Sci Immunol 2023; 8:eabq4573. [PMID: 37540734 PMCID: PMC11192171 DOI: 10.1126/sciimmunol.abq4573] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/07/2023] [Indexed: 08/06/2023]
Abstract
Maintaining macrophage (MΦ) heterogeneity is critical to ensure intestinal tissue homeostasis and host defense. The gut microbiota and host factors are thought to synergistically guide intestinal MΦ development, although the exact nature, regulation, and location of such collaboration remain unclear. Here, we report that microbial biochemical energy metabolism promotes colony-stimulating factor 2 (CSF2) production by group 3 innate lymphoid cells (ILC3s) within solitary isolated lymphoid tissues (SILTs) in a cell-extrinsic, NLRP3/P2X7R-dependent fashion in the steady state. Tissue-infiltrating monocytes accumulating around SILTs followed a spatially constrained, distinct developmental trajectory into SILT-associated MΦs (SAMs). CSF2 regulated the mitochondrial membrane potential and reactive oxygen species production of SAMs and contributed to the antimicrobial defense against enteric bacterial infections. Collectively, these findings identify SILTs and CSF2-producing ILC3s as a microanatomic niche for intestinal MΦ development and functional programming fueled by the integration of commensal microbial energy metabolism.
Collapse
Affiliation(s)
- Pailin Chiaranunt
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Kyle Burrows
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Eric Y. Cao
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Helen Liang
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Homaira Hamidzada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Anthony Wong
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Julia Gschwend
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Pascal Flüchter
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Tijana Despot
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Sung Min Lim
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | | | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, USA
| | - Hiromi Imamura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Slava Epelman
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Arroyo Portilla C, Fenouil R, Wagner C, Luciani C, Lagier M, Da Silva C, Hidalgo-Villeda F, Spinelli L, Fallet M, Tomas J, Gorvel JP, Lelouard H. Peyer's patch phagocytes acquire specific transcriptional programs that influence their maturation and activation profiles. Mucosal Immunol 2023; 16:527-547. [PMID: 37257775 DOI: 10.1016/j.mucimm.2023.05.009] [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: 04/26/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Peyer's patches (PPs) are secondary lymphoid organs in contact with the external environment via the intestinal lumen, thus combining antigen sampling and immune response initiation sites. Therefore, they provide a unique opportunity to study the entire process of phagocyte differentiation and activation in vivo. Here, we deciphered the transcriptional and spatial landscape of PP phagocyte populations from their emergence in the tissue to their final maturation state at homeostasis and under stimulation. Activation of monocyte-derived Lysozyme-expressing dendritic cells (LysoDCs) differs from that of macrophages by their upregulation of conventional DC (cDC) signature genes such as Ccr7 and downregulation of typical monocyte-derived cell genes such as Cx3cr1. We identified gene sets that distinguish PP cDCs from the villus ones and from LysoDCs. We also identified key immature, early, intermediate, and late maturation markers of PP phagocytes. Finally, exploiting the ability of the PP interfollicular region to host both villous and subepithelial dome emigrated cDCs, we showed that the type of stimulus, the subset, but also the initial location of cDCs shape their activation profile and thus direct the immune response. Our study highlights the importance of targeting the right phagocyte subset at the right place and time to manipulate the immune response.
Collapse
Affiliation(s)
- Cynthia Arroyo Portilla
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France; Departamento de Análisis Clínicos, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Romain Fenouil
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Cécilia Luciani
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Margaux Lagier
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Clément Da Silva
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Fanny Hidalgo-Villeda
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France; Escuela de Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Lionel Spinelli
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Mathieu Fallet
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Julie Tomas
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Jean-Pierre Gorvel
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Hugues Lelouard
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France.
| |
Collapse
|
31
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [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: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
32
|
Sciarra F, Franceschini E, Campolo F, Venneri MA. The Diagnostic Potential of the Human Blood Microbiome: Are We Dreaming or Awake? Int J Mol Sci 2023; 24:10422. [PMID: 37445600 DOI: 10.3390/ijms241310422] [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: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Human blood has historically been considered a sterile environment. Recently, a thriving microbiome dominated by Firmicutes, Actinobacteria, Proteobacteria, and Bacteroidetes phyla was detected in healthy blood. The localization of these microbes is restricted to some blood cell populations, particularly the peripheral blood mononuclear cells and erythrocytes. It was hypothesized that the blood microbiome originates from the skin-oral-gut axis. In addition, many studies have evaluated the potential of blood microbiome dysbiosis as a prognostic marker in cardiovascular diseases, cirrhosis, severe liver fibrosis, severe acute pancreatitis, type 2 diabetes, and chronic kidney diseases. The present review aims to summarize current findings and most recent evidence in the field.
Collapse
Affiliation(s)
- Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Edoardo Franceschini
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
33
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
34
|
Giannou AD, Kempski J, Zhang T, Lücke J, Shiri AM, Zazara DE, Belios I, Machicote A, Seeger P, Agalioti T, Tintelnot J, Sagebiel A, Tomczak M, Bauditz L, Bedke T, Kocheise L, Mercanoglu B, Fard-Aghaie M, Giorgakis E, Lykoudis PM, Pikouli A, Grass JK, Wahib R, Bardenhagen J, Brunswig B, Heumann A, Ghadban T, Duprée A, Tachezy M, Melling N, Arck PC, Stringa P, Gentilini MV, Gondolesi GE, Nakano R, Thomson AW, Perez D, Li J, Mann O, Izbicki JR, Gagliani N, Maroulis IC, Huber S. IL-22BP controls the progression of liver metastasis in colorectal cancer. Front Oncol 2023; 13:1170502. [PMID: 37324022 PMCID: PMC10265988 DOI: 10.3389/fonc.2023.1170502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Background The immune system plays a pivotal role in cancer progression. Interleukin 22 binding protein (IL-22BP), a natural antagonist of the cytokine interleukin 22 (IL-22) has been shown to control the progression of colorectal cancer (CRC). However, the role of IL-22BP in the process of metastasis formation remains unknown. Methods We used two different murine in vivo metastasis models using the MC38 and LLC cancer cell lines and studied lung and liver metastasis formation after intracaecal or intrasplenic injection of cancer cells. Furthermore, IL22BP expression was measured in a clinical cohort of CRC patients and correlated with metastatic tumor stages. Results Our data indicate that low levels of IL-22BP are associated with advanced (metastatic) tumor stages in colorectal cancer. Using two different murine in vivo models we show that IL-22BP indeed controls the progression of liver but not lung metastasis in mice. Conclusions We here demonstrate a crucial role of IL-22BP in controlling metastasis progression. Thus, IL-22 might represent a future therapeutic target against the progression of metastatic CRC.
Collapse
Affiliation(s)
- Anastasios D. Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Surgery, University of Patras Medical School, Patras, Greece
| | - Jan Kempski
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dimitra E. Zazara
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ioannis Belios
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andres Machicote
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philipp Seeger
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Theodora Agalioti
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joseph Tintelnot
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ll. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrian Sagebiel
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Miriam Tomczak
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Bauditz
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Bedke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lorenz Kocheise
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Baris Mercanoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mohammad Fard-Aghaie
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Emmanouil Giorgakis
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Division of Transplantation, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Panagis M. Lykoudis
- 3rd Department of Surgery, Attiko University Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom
| | - Anastasia Pikouli
- 3rd Department of Surgery, Attiko University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Julia-Kristin Grass
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramez Wahib
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Bardenhagen
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Brunswig
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tarik Ghadban
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Duprée
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C. Arck
- Laboratory for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pablo Stringa
- Department General Surgery, Liver, Pancreas and Intestinal Transplantation, Hospital Universitario, Fundacion Favaloro, Buenos Aires, Argentina
| | - Maria Virginia Gentilini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTyB, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro), Laboratorio de Inmunología Asociada al Trasplante, Buenos Aires, Argentina
| | - Gabriel E. Gondolesi
- Department General Surgery, Liver, Pancreas and Intestinal Transplantation, Hospital Universitario, Fundacion Favaloro, Buenos Aires, Argentina
| | - Ryosuke Nakano
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angus W. Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jun Li
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
35
|
Zhao C, Li Y, Tang J, Zhou Q, Lin X, Wen Z. Metaphocytes are IL-22BP-producing cells regulated by ETS transcription factor Spic and essential for zebrafish barrier immunity. Cell Rep 2023; 42:112483. [PMID: 37148242 DOI: 10.1016/j.celrep.2023.112483] [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: 10/06/2022] [Revised: 03/03/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Metaphocytes are tissue-resident macrophage (TRM)/dendritic cell (DC)-like cells of non-hematopoietic origin in zebrafish barrier tissues. One remarkable property of metaphocytes is their ability to capture soluble antigens from the external environment via transepithelial protrusions, a unique function manifested by specialized subpopulations of the TRMs/DCs in mammal barrier tissues. Yet, how metaphocytes acquire myeloid-like cell properties from non-hematopoietic precursors and how they regulate barrier immunity remains unknown. Here, we show that metaphocytes are in situ generated from local progenitors guided by the ETS transcription factor Spic, the deficiency of which results in the absence of metaphocytes. We further document that metaphocytes are the major IL-22BP-producing cells, and the depletion of metaphocytes causes dysregulated barrier immunity that resembles the phenotype of IL-22BP-deficient mice. These findings reveal the ontogeny, development, and function of metaphocytes in zebrafish, which facilitates our understanding of the nature and function of the mammalian TRM/DC counterparts.
Collapse
Affiliation(s)
- Changlong Zhao
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yunbo Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Jinlin Tang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Qiuxia Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Xi Lin
- Brigham and Women's Hospital, Harvard Medical School, Boston, MS 02115, USA
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China; Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
36
|
Wang X, Yu S, Liu W, Lv P, Zhao L, Wang Y, Fu C, Meng L, Yang Q, Wang X, Huang Y, Zuo Z, Liu X. Relationship between IL-22 and IL-22BP in diabetic cognitive dysfunction. Acta Diabetol 2023; 60:631-644. [PMID: 36717397 DOI: 10.1007/s00592-022-02024-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/21/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND CD4 + T helper (Th)22 cells play a regulatory role in autoimmune diseases such as type 1 diabetes mellitus. The Th22-related cytokine interleukin (IL)-22, the expression of which is increased in diabetes mellitus (DM), can act as a neurotrophic factor to protect neurons from apoptosis. Paradoxically, neuronal apoptosis and learning and memory decline occur in DM. In this study, we investigated the relationship between IL-22 and its receptors IL-22Rα1 and IL-22 binding protein (IL-22BP, a soluble inhibitor of IL-22) in diabetic encephalopathy (DE) and the effects of IL-22 on hippocampal neurons, learning and memory. METHODS A C57BL/6 mouse model of diabetes was constructed by intraperitoneal injection of streptozotocin. The mice were randomly divided into 4 groups: the control group, diabetes group, diabetes + recombinantIL-22 (rIL-22) group and diabetes + IL-22BP group. The Morris water maze test was used to evaluate learning and memory, the expression of IL-22 was measured by ELISA, and Evans Blue staining was used to evaluate blood-brain barrier permeability. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to measure the mRNA expression of IL-22 and IL-22Rα1 in the hippocampus. The morphology and number of hippocampal neurons were assessed by Nissl staining, and TUNEL staining was used to detect hippocampal neuronal apoptosis. Immunofluorescence was used to analyze IL-22Rα1 expression and localization in hippocampus, and Western blotting was used to evaluate the expression of IL-22, IL-22Rα1, IL-22BP, and the apoptosis related proteins Caspase-3 and C-caspase-3. RESULTS Compared with those in the control group, mice in the diabetes group showed cognitive decline; apoptosis of hippocampal neurons; increased expression of hippocampal Caspase-3, C-Caspase-3, IL-22, IL-22Rα1, and IL-22BP; and a decreased IL-22/IL-22BP ratio. Learning and memory were improved, neuronal apoptosis was attenuated, IL-22Rα1 expression and the IL-22/IL-22BP ratio were increased, and caspase-3 and C-caspase-3 expression was decreased in the rIL-22-treated group compared with the diabetes group. IL-22BP treatment aggravated diabetic cognitive dysfunction and pathological alterations in the hippocampus, decreased the IL-22/IL-22BP ratio, and increased the expression of caspase-3 and C-caspase-3 in mice with diabetes. CONCLUSION A decrease in the IL-22/IL-22BP ratio plays an important role in diabetic cognitive dysfunction, and rIL-22 can effectively alleviate DE. Herein, we shed light on the interaction between IL-22 and IL-22BP as therapeutic targets for DM.
Collapse
Affiliation(s)
- Xiaobai Wang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Shengxue Yu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Wenqiang Liu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Pan Lv
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Lipan Zhao
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Yufei Wang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Cong Fu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Lu Meng
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Qi Yang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Xuehua Wang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Ying Huang
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Zhongfu Zuo
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China.
- Department of Anatomy, Histology and Embryology, Postdoctoral Research Station, Guangxi Medical University, Nanning, China.
| | - Xuezheng Liu
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
37
|
Zhuang YP, Pan ZY, Huang JM, Zhou HG, Yang XR, Zhong HJ. Negative correlation between circulating integrin α4 + group 3 innate lymphoid cells and the severity of type 2 diabetes. Int Immunopharmacol 2023; 119:110237. [PMID: 37121112 DOI: 10.1016/j.intimp.2023.110237] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/27/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Impaired intestinal barrier and immune dysfunction promote the development of type 2 diabetes (T2D). Group 3 innate lymphoid cells (ILC3s), which are enriched in the intestinal lamina propria, are key for intestinal barrier integrity. However, there is a paucity of data on circulating ILC3s in patients with T2D. PURPOSE To examine the characteristics of ILC3s in patients with T2D and identify the relationship between ILC3s and clinical indicators of T2D. METHODS Fifty-nine patients with T2D and thirty controls were enrolled in this retrospective study. Peripheral blood mononuclear cells were isolated and analyzed by flow cytometry and plasma cytokine levels were measured by enzyme-linked immunosorbent assays. RESULTS The proportion of circulating ILC3s in the T2D group was significantly lower than that in controls and showed a negative correlation with fasting glucose and glycated hemoglobin and a positive correlation with granulocyte-macrophage colony-stimulating factor (GM-CSF). Similarly, the proportion of circulating integrin α4+ ILC3s was also significantly lower in the T2D group and showed a negative correlation with fasting glucose and glycated hemoglobin and a positive correlation with GM-CSF. Moreover, the level of circulating integrin α4+ ILC3s showed a positive correlation with the proportion of circulating dendritic cells (DCs), which was also decreased in patients with T2D and positively associated with GM-CSF. CONCLUSION ILC3s, especially integrin α4+ ILC3s, were decreased in patients with T2D and showed a negative correlation with disease severity. These cell subsets may delay the progression of T2D by promoting DC differentiation via the secretion of GM-CSF.
Collapse
Affiliation(s)
- Yu-Pei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, China
| | - Zhao-Yu Pan
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, China
| | - Jian-Ming Huang
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, China
| | - Hong-Guang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xiao-Rong Yang
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, China.
| | - Hao-Jie Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, China; Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
38
|
Sorini C, Tripathi KP, Wu S, Higdon SM, Wang J, Cheng L, Banerjee S, Reinhardt A, Kreslavsky T, Thorell A, Engstrand L, Du J, Villablanca EJ. Metagenomic and single-cell RNA-Seq survey of the Helicobacter pylori-infected stomach in asymptomatic individuals. JCI Insight 2023; 8:161042. [PMID: 36810249 PMCID: PMC9977493 DOI: 10.1172/jci.insight.161042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 01/11/2023] [Indexed: 02/23/2023] Open
Abstract
Helicobacter pylori colonization of the gastric niche can persist for years in asymptomatic individuals. To deeply characterize the host-microbiota environment in H. pylori-infected (HPI) stomachs, we collected human gastric tissues and performed metagenomic sequencing, single-cell RNA-Seq (scRNA-Seq), flow cytometry, and fluorescent microscopy. HPI asymptomatic individuals had dramatic changes in the composition of gastric microbiome and immune cells compared with noninfected individuals. Metagenomic analysis uncovered pathway alterations related to metabolism and immune response. scRNA-Seq and flow cytometry data revealed that, in contrast to murine stomachs, ILC2s are virtually absent in the human gastric mucosa, whereas ILC3s are the dominant population. Specifically, proportion of NKp44+ ILC3s out of total ILCs were highly increased in the gastric mucosa of asymptomatic HPI individuals, and correlated with the abundance of selected microbial taxa. In addition, CD11c+ myeloid cells and activated CD4+ T cells and B cells were expanded in HPI individuals. B cells of HPI individuals acquired an activated phenotype and progressed into a highly proliferating germinal-center stage and plasmablast maturation, which correlated with the presence of tertiary lymphoid structures within the gastric lamina propria. Our study provides a comprehensive atlas of the gastric mucosa-associated microbiome and immune cell landscape when comparing asymptomatic HPI and uninfected individuals.
Collapse
Affiliation(s)
- Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Kumar P Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Shengru Wu
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Shawn M Higdon
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Jing Wang
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Liqin Cheng
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Sanghita Banerjee
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Taras Kreslavsky
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | | | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Juan Du
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
39
|
Pan ZY, Liu HQ, Zhuang YP, Tan HB, Yang XY, Zhong HJ, He XX. Reduced type 3 innate lymphoid cells related to worsening kidney function in renal dysfunction. Exp Biol Med (Maywood) 2023; 248:242-252. [PMID: 36670544 PMCID: PMC10107398 DOI: 10.1177/15353702221147561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/22/2022] [Indexed: 01/22/2023] Open
Abstract
Intestinal mucosa barrier injury and immunity imbalance contribute to chronic kidney disease (CKD) progression. Type 3 innate lymphoid cells (ILC3s) are essential for normal intestinal homeostasis. Nevertheless, the relationship between ILC3s and CKD remains largely unknown. The aim of this study was to investigate the relationship linking ILC3s to clinical indicators among patients with renal dysfunction. The levels of circulating ILC3s and dendritic cells, as well as their subsets, in patients with renal dysfunction and healthy controls were determined through flow cytometry. The levels of human plasma granulocyte-macrophage colony-stimulating factor (GM-CSF) were measured using enzyme-linked immunosorbent assay. Renal function was evaluated by measuring the estimated glomerular filtration rate (eGFR), as well as the levels of serum creatinine, blood urea nitrogen (BUN), and uric acid. The results revealed that the proportion of peripheral ILC3s was significantly decreased in patients with renal dysfunction. This reduction was positively associated with the levels of eGFR, and inversely associated with the levels of BUN and uric acid. Similarly, the percentage of circulating C-C motif chemokine receptor 6-positive (CCR6 +) ILC3s was also obviously reduced, and demonstrated positive and negative associations with the levels of eGFR and BUN, respectively. Furthermore, the levels of CCR6 + ILC3s correlated positively with those of GM-CSF, as well as type 1 conventional dendritic cells (cDC1s), which also decreased in parallel with kidney function. Thus, the reduction of ILC3s, particularly CCR6 + ILC3s, was related to worsening kidney function in patients with renal dysfunction. This effect may delay renal function impairment by regulating cDC1s via the secretion of GM-CSF, indicating that CCR6 + ILC3s may serve as efficient biomarkers for evaluating kidney function.
Collapse
Affiliation(s)
- Zhao-Yu Pan
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
| | - Hong-Qian Liu
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
| | - Yu-Pei Zhuang
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- The First Clinical Medical College,
Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hai-Bo Tan
- Shenzhen Traditional Chinese Medicine
Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033,
China
| | - Xiao-Ya Yang
- Department of Physiology, Guangzhou
Health Science College, Guangzhou 510450, China
| | - Hao-Jie Zhong
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- School of Biology and Biological
Engineering, South China University of Technology, Guangzhou 510080, China
| | - Xing-Xiang He
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- Xing-Xiang He.
| |
Collapse
|
40
|
Filardy AA, Ferreira JRM, Rezende RM, Kelsall BL, Oliveira RP. The intestinal microenvironment shapes macrophage and dendritic cell identity and function. Immunol Lett 2023; 253:41-53. [PMID: 36623708 PMCID: PMC9907447 DOI: 10.1016/j.imlet.2023.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
The gut comprises the largest body interface with the environment and is continuously exposed to nutrients, food antigens, and commensal microbes, as well as to harmful pathogens. Subsets of both macrophages and dendritic cells (DCs) are present throughout the intestinal tract, where they primarily inhabit the gut-associate lymphoid tissue (GALT), such as Peyer's patches and isolated lymphoid follicles. In addition to their role in taking up and presenting antigens, macrophages and DCs possess extensive functional plasticity and these cells play complementary roles in maintaining immune homeostasis in the gut by preventing aberrant immune responses to harmless antigens and microbes and by promoting host defense against pathogens. The ability of macrophages and DCs to induce either inflammation or tolerance is partially lineage imprinted, but can also be dictated by their activation state, which in turn is determined by their specific microenvironment. These cells express several surface and intracellular receptors that detect danger signals, nutrients, and hormones, which can affect their activation state. DCs and macrophages play a fundamental role in regulating T cells and their effector functions. Thus, modulation of intestinal mucosa immunity by targeting antigen presenting cells can provide a promising approach for controlling pathological inflammation. In this review, we provide an overview on the characteristics, functions, and origins of intestinal macrophages and DCs, highlighting the intestinal microenvironmental factors that influence their functions during homeostasis. Unraveling the mechanisms by which macrophages and DCs regulate intestinal immunity will deepen our understanding on how the immune system integrates endogenous and exogenous signals in order to maintain the host's homeostasis.
Collapse
Affiliation(s)
- Alessandra A Filardy
- Laboratório de Imunologia Celular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Brazil.
| | - Jesuino R M Ferreira
- Laboratório de Imunologia Celular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Brazil
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Brian L Kelsall
- Laboratory of Molecular Immunology, NIAID, National Institutes of Health, USA
| | | |
Collapse
|
41
|
Gu J, Zhou P, Liu Y, Xu Q, Chen X, Chen M, Lu C, Qu C, Tong Y, Yu Q, Lu X, Yu C, Liu Z. Down-regulating Interleukin-22/Interleukin-22 binding protein axis promotes inflammation and aggravates diet-induced metabolic disorders. Mol Cell Endocrinol 2022; 557:111776. [PMID: 36108991 DOI: 10.1016/j.mce.2022.111776] [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: 04/20/2022] [Revised: 08/25/2022] [Accepted: 09/08/2022] [Indexed: 11/26/2022]
Abstract
The prevalence of metabolic diseases has become a severe public health problem. Previously, we reported that Interleukin-22 (IL-22) was independently associated with type 2 diabetes mellitus and cardiovascular disease, and could protect endothelial cells from glucose- and lysophosphatidylcholine-induced injury. The activity of IL-22 is strongly regulated by IL-22-binding protein (IL-22BP). The aim of this investigation was to determine the effect of IL-22/IL-22BP axis on glucolipid metabolism. Serum IL-22 and IL-22BP expression in metabolic syndrome (MetS) patients and healthy controls was examined. IL-22BP-knockout (IL-22ra2-/-) and wild-type (WT) mice were fed with control diet (CTD) and high-fat diet (HFD) for 12 weeks. The IL-22 related pathway expression, the glucolipid metabolism, and inflammatory markers in mice were examined. Serum IL-22 and IL-22BP levels were found significantly increased in MetS patients (p < 0.001). IL-22BP deficiency down-regulated IL-22-related pathway, aggravated glucolipid metabolism disorder, and promoted inflammation in mice. Collectively, this work deepens the understanding of the relationship between IL-22/IL-22BP axis and metabolism disorders, and identified that down-regulation of IL-22/IL-22BP axis promotes metabolic disorders in mice.
Collapse
Affiliation(s)
- Jiayi Gu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Ping Zhou
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Ying Liu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Qiao Xu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Xi Chen
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Mengqi Chen
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Chen Lu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, 109 Longmian Avenue, Jiangning District, Nanjing, China
| | - Chen Qu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Yanli Tong
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xiang Lu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China.
| | - Chunzhao Yu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, 109 Longmian Avenue, Jiangning District, Nanjing, China; Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, PR China.
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, 210011, PR China.
| |
Collapse
|
42
|
Ghilas S, O’Keefe R, Mielke LA, Raghu D, Buchert M, Ernst M. Crosstalk between epithelium, myeloid and innate lymphoid cells during gut homeostasis and disease. Front Immunol 2022; 13:944982. [PMID: 36189323 PMCID: PMC9524271 DOI: 10.3389/fimmu.2022.944982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/29/2022] [Indexed: 12/05/2022] Open
Abstract
The gut epithelium not only provides a physical barrier to separate a noxious outside from a sterile inside but also allows for highly regulated interactions between bacteria and their products, and components of the immune system. Homeostatic maintenance of an intact epithelial barrier is paramount to health, requiring an intricately regulated and highly adaptive response of various cells of the immune system. Prolonged homeostatic imbalance can result in chronic inflammation, tumorigenesis and inefficient antitumor immune control. Here we provide an update on the role of innate lymphoid cells, macrophages and dendritic cells, which collectively play a critical role in epithelial barrier maintenance and provide an important linkage between the classical innate and adaptive arm of the immune system. These interactions modify the capacity of the gut epithelium to undergo continuous renewal, safeguard against tumor formation and provide feedback to the gut microbiome, which acts as a seminal contributor to cellular homeostasis of the gut.
Collapse
Affiliation(s)
- Sonia Ghilas
- Mucosal Immunity Laboratory, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Ryan O’Keefe
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Lisa Anna Mielke
- Mucosal Immunity Laboratory, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Dinesh Raghu
- Mucosal Immunity Laboratory, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Michael Buchert
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
- *Correspondence: Michael Buchert, ; Matthias Ernst,
| | - Matthias Ernst
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, and La Trobe University - School of Cancer Medicine, Heidelberg, VIC, Australia
- *Correspondence: Michael Buchert, ; Matthias Ernst,
| |
Collapse
|
43
|
Crosstalk between macrophages and innate lymphoid cells (ILCs) in diseases. Int Immunopharmacol 2022; 110:108937. [PMID: 35779490 DOI: 10.1016/j.intimp.2022.108937] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022]
Abstract
Innate lymphoid cells (ILCs) and macrophages are tissue-resident cells that play important roles in tissue-immune homeostasis and immune regulation. ILCs are mainly distributed on the barrier surfaces of mammals to ensure immunity or tissue homeostasis following host, microbial, or environmental stimulation. Their complex relationships with different organs enable them to respond quickly to disturbances in environmental conditions and organ homeostasis, such as during infections and tissue damage. Gradually emerging evidence suggests that ILCs also play complex and diverse roles in macrophage development, homeostasis, polarization, inflammation, and viral infection. In turn, macrophages also determine the fate of ILCs to some extent, which indicates that network crossover between these interactions is a key determinant of the immune response. More work is needed to better define the crosstalk of ILCs with macrophages in different tissues and demonstrate how it is affected during inflammation and other diseases. Here, we summarize current research on the functional interactions between ILCs and macrophages and consider the potential therapeutic utility of these interactions for the benefit of human health.
Collapse
|
44
|
Intestinal fibroblastic reticular cell niches control innate lymphoid cell homeostasis and function. Nat Commun 2022; 13:2027. [PMID: 35440118 PMCID: PMC9018819 DOI: 10.1038/s41467-022-29734-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Innate lymphoid cells (ILCs) govern immune cell homeostasis in the intestine and protect the host against microbial pathogens. Various cell-intrinsic pathways have been identified that determine ILC development and differentiation. However, the cellular components that regulate ILC sustenance and function in the intestinal lamina propria are less known. Using single-cell transcriptomic analysis of lamina propria fibroblasts, we identify fibroblastic reticular cells (FRCs) that underpin cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Genetic ablation of lymphotoxin-β receptor expression in Ccl19-expressing FRCs blocks the maturation of CPs into mature ILFs. Interactome analysis shows the major niche factors and processes underlying FRC-ILC crosstalk. In vivo validation confirms that a sustained lymphotoxin-driven feedforward loop of FRC activation including IL-7 generation is critical for the maintenance of functional ILC populations. In sum, our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure protective gut immunity. Fibroblastic reticular cells (FRCs) support localisation of immune cells in secondary lymphoid tissues but less is known about the lamina propria. Here the authors use scRNA-seq and intestinal infection to characterise FRCs in the intestinal lamina propria and show specialised niches that foster innate lymphoid cells during homeostasis and infection.
Collapse
|
45
|
Zeiser R, Warnatz K, Rosshart S, Sagar, Tanriver Y. GVHD, IBD and primary immunodeficiencies: The gut as a target of immunopathology resulting from impaired immunity. Eur J Immunol 2022; 52:1406-1418. [PMID: 35339113 DOI: 10.1002/eji.202149530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 01/21/2022] [Indexed: 11/11/2022]
Abstract
The intestinal tract is the largest immunological organ in the body and has a central function of regulating local immune responses, as the intestinal epithelial barrier is a location where the immune system interacts with the gut microbiome including bacteria, fungi and viruses. Impaired immunity in the intestinal tract can lead to immunopathology, which manifests in different diseases such as inflammatory bowel disease (IBD) or intestinal graft-versus-host disease (GVHD). A disturbed communication between epithelial cells, immune cells and microbiome will shape pathogenic immune responses to antigens, which need to be counterbalanced by tolerogenic mechanisms and repair mechanisms. Here, we review how impaired intestinal immune function leads to immunopathology with a specific focus on innate immune cells, the role of the microbiome and the resulting clinical manifestations including intestinal GVHD, IBD and enteropathy in primary immunodeficiency. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology - Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Rosshart
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yakup Tanriver
- Department of Medicine IV (Nephrology and Primary Care), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Microbiology and Hygiene, Institute for Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
46
|
Murphy JM, Ngai L, Mortha A, Crome SQ. Tissue-Dependent Adaptations and Functions of Innate Lymphoid Cells. Front Immunol 2022; 13:836999. [PMID: 35359972 PMCID: PMC8960279 DOI: 10.3389/fimmu.2022.836999] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-resident immune cells reside in distinct niches across organs, where they contribute to tissue homeostasis and rapidly respond to perturbations in the local microenvironment. Innate lymphoid cells (ILCs) are a family of innate immune cells that regulate immune and tissue homeostasis. Across anatomical locations throughout the body, ILCs adopt tissue-specific fates, differing from circulating ILC populations. Adaptations of ILCs to microenvironmental changes have been documented in several inflammatory contexts, including obesity, asthma, and inflammatory bowel disease. While our understanding of ILC functions within tissues have predominantly been based on mouse studies, development of advanced single cell platforms to study tissue-resident ILCs in humans and emerging patient-based data is providing new insights into this lymphocyte family. Within this review, we discuss current concepts of ILC fate and function, exploring tissue-specific functions of ILCs and their contribution to health and disease across organ systems.
Collapse
Affiliation(s)
- Julia M. Murphy
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
47
|
Luciani C, Hager FT, Cerovic V, Lelouard H. Dendritic cell functions in the inductive and effector sites of intestinal immunity. Mucosal Immunol 2022; 15:40-50. [PMID: 34465895 DOI: 10.1038/s41385-021-00448-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 02/04/2023]
Abstract
The intestine is constantly exposed to foreign antigens, which are mostly innocuous but can sometimes be harmful. Therefore, the intestinal immune system has the delicate task of maintaining immune tolerance to harmless food antigens while inducing tailored immune responses to pathogens and regulating but tolerating the microbiota. Intestinal dendritic cells (DCs) play a central role in these functions as sentinel cells able to prime and polarize the T cell responses. DCs are deployed throughout the intestinal mucosa but with local specializations along the gut length and between the diffuse effector sites of the gut lamina propria (LP) and the well-organized immune inductive sites comprising isolated lymphoid follicles (ILFs), Peyer's patches (PPs), and other species-specific gut-associated lymphoid tissues (GALTs). Understanding the specificities of each intestinal DC subset, how environmental factors influence DC functions, and how these can be modulated is key to harnessing the therapeutic potential of mucosal adaptive immune responses, whether by enhancing the efficacy of mucosal vaccines or by increasing tolerogenic responses in inflammatory disorders. In this review, we summarize recent findings related to intestinal DCs in steady state and upon inflammation, with a special focus on their functional specializations, highly dependent on their microenvironment.
Collapse
Affiliation(s)
| | | | - Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | | |
Collapse
|
48
|
Chen H, Sun L, Feng L, Yin Y, Zhang W. Role of Innate lymphoid Cells in Obesity and Insulin Resistance. Front Endocrinol (Lausanne) 2022; 13:855197. [PMID: 35574038 PMCID: PMC9091334 DOI: 10.3389/fendo.2022.855197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity, a growing chronic metabolic disease, greatly increases the risk of metabolic syndrome which includes type 2 diabetes, fatty liver and cardiovascular diseases. Obesity-associated metabolic diseases significantly contribute to mortality and reduce life expectancy. Recently, innate lymphoid cells (ILCs) have emerged as crucial regulators of metabolic homeostasis and tissue inflammation. This review focuses on the roles of ILCs in different metabolic tissues, including adipose tissue, liver, pancreas, and intestine. We briefly outline the relationship between obesity, inflammation, and insulin resistance. We then discuss how ILCs in distinct metabolic organs may function to maintain metabolic homeostasis and contribute to obesity and its associated metabolic diseases. The potential of ILCs as the therapeutic target for obesity and insulin resistance is also addressed.
Collapse
Affiliation(s)
- Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lu Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| |
Collapse
|
49
|
Overview: Themes in Innate Lymphoid Cell Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:1-6. [DOI: 10.1007/978-981-16-8387-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
50
|
Zenewicz LA. IL-22 Binding Protein (IL-22BP) in the Regulation of IL-22 Biology. Front Immunol 2021; 12:766586. [PMID: 34868019 PMCID: PMC8634938 DOI: 10.3389/fimmu.2021.766586] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/25/2021] [Indexed: 01/21/2023] Open
Abstract
Cytokines are powerful mediators of inflammation. Consequently, their potency is regulated in many ways to protect the host. Several cytokines, including IL-22, have coordinating binding proteins or soluble receptors that bind to the cytokine, block the interaction with the cellular receptor, and thus prevent cellular signaling. IL-22 is a critical cytokine in the modulation of tissue responses during inflammation and is highly upregulated in many chronic inflammatory disease patients, including those with psoriasis, rheumatoid arthritis, and inflammatory bowel disease (IBD). In healthy individuals, low levels of IL-22 are secreted by immune cells, mainly in the gastrointestinal (GI) tract. However, much of this IL-22 is likely not biologically active due to the high levels of IL-22 binding protein (IL-22BP) produced by intestinal dendritic cells (DCs). IL-22BP is a soluble receptor homolog that binds to IL-22 with greater affinity than the membrane spanning receptor. Much is known regarding the regulation and function of IL-22 in health and disease. However, less is known about IL-22BP. In this review, we will focus on IL-22BP, including its regulation, role in IL-22 biology and inflammation, and promise as a therapeutic. IL-22 can be protective or pathogenic, depending on the context of inflammation. IL-22BP also has divergent roles. Ongoing and forthcoming studies will expand our knowledge of IL-22BP and IL-22 biology, and suggest that IL-22BP holds promise as a way to regulate IL-22 biology in patients with chronic inflammatory disease.
Collapse
Affiliation(s)
- Lauren A. Zenewicz
- Department of Microbiology and Immunology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|