1
|
Colpitts SJ, Jegatheeswaran S, Oakie A, Mashhouri S, Sachewsky N, Murshed H, Mathews JA, Reid KT, Misra PS, Fung VCW, Reichman TW, Nostro MC, Verchere CB, Levings MK, Crome SQ. Cell therapy with human IL-10-producing ILC2s enhances islet function and inhibits allograft rejection. Am J Transplant 2025:S1600-6135(25)00280-1. [PMID: 40412656 DOI: 10.1016/j.ajt.2025.05.023] [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: 11/20/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Group 2 innate lymphoid cells (ILC2s) that produce IL-10 (IL-10+ILC2s) have demonstrated regulatory and tissue-protective properties in murine studies, but preclinical studies are lacking that explore the potential of human IL-10+ILC2s as a tolerance-promoting cell therapy for transplantation or autoimmunity. Here, we investigated whether human IL-10+ILC2s could enhance islet function and prevent allograft rejection in humanized mouse models of islet transplantation. In vitro, human IL-10+ILC2s did not display cytotoxicity towards allogeneic deceased-donor islets or stem cell-derived islet-like cells, and co-transplantation with IL-10+ILC2s significantly improved glucose control post-transplantation. Allogeneic IL10+ILC2s directly inhibited T cell-mediated cytotoxicity against islet-like cells in vitro, and in an antigen-specific transplant rejection model, prevented T cell-mediated rejection of deceased donor islet grafts. Effects were greater with allogeneic IL-10+ILC2s, as autologous cells did not inhibit T cell IFN-γ production or cytotoxic activity in vitro, and were not sufficient to prevent islet rejection in vivo.Collectively, these studies provide proof-of-principle that human IL-10+ILC2s have therapeutic potential for islet transplantation and type 1 diabetes, and support their use as an allogeneic regulatory cell therapy.
Collapse
Affiliation(s)
- Sarah J Colpitts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Sinthuja Jegatheeswaran
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Siavash Mashhouri
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Nadia Sachewsky
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Humaira Murshed
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Vivian C W Fung
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Trevor W Reichman
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - C Bruce Verchere
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Cao Q, Wang Y, Chen J, Wang R, Chen T, Gloss B, Read SA, Wang X, Lee VWS, Clancy L, Rogers NM, Alexander SI, Zheng G, Yu D, Harris DCH. Targeting inflammation with chimeric antigen receptor macrophages using a signal switch. Nat Biomed Eng 2025:10.1038/s41551-025-01387-8. [PMID: 40335685 DOI: 10.1038/s41551-025-01387-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/01/2025] [Indexed: 05/09/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy has shown great success in clinical cancer, bringing hope to apply CAR strategies to other clinical settings. Here we developed a CAR macrophage (CAR-M) that recognizes the major inflammatory molecule tumour necrosis factor (TNF) and activates an intracellular IL-4 signalling pathway, thereby programming engineered macrophages for an anti-inflammatory function. CAR-M therapy has exhibited efficacy in mouse models of both acute and chronic inflammatory diseases. In kidney ischaemia reperfusion injury (IRI), infused CAR-Ms switched to an anti-inflammatory phenotype in inflamed kidney and attenuated kidney IRI. The anti-inflammatory phenotype of infused CAR-Ms switched off during the recovery phase of kidney IRI, coinciding with the disappearance of TNF. In Adriamycin-induced nephropathy, a model of chronic inflammatory disease, infused CAR-Ms maintained an anti-inflammatory phenotype for several weeks in response to sustained high levels of TNF and improved kidney function and structure. CAR-Ms also effectively reduced tissue injury in another organ, the liver. Human anti-TNF CAR-Ms exhibit anti-inflammatory phenotype and function in response to TNF. The CAR-M design, using signal switching, holds promise for the treatment of a broad range of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Jianwei Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Ruifeng Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Titi Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Westmead Hospital, Sydney, New South Wales, Australia
| | - Brian Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Scott A Read
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Xuerong Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Vincent W S Lee
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Westmead Hospital, Sydney, New South Wales, Australia
| | - Leighton Clancy
- Blood Transplant and Cell Therapies Laboratory, NSW Health Pathology-ICPMR Westmead, Sydney, New South Wales, Australia
- Bone Marrow Transplant & Cell Therapies, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Westmead Hospital, Sydney, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Di Yu
- Faculty of Medicine, The University of Queensland Diamantina Institute, St Lucia, Queensland, Australia
| | - David C H Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.
- Department of Renal Medicine, Westmead Hospital, Sydney, New South Wales, Australia.
| |
Collapse
|
3
|
Jin X, Jian Z, Ma Y, Wen J, Chao N, Chen X, Xiang L, Yuan Y, Liu L, Li Y, Wei J, Liao B, Zhang L, Wang K. Single-Cell RNA Sequencing Analysis Reveals the Role of Macrophage-Mediated CD44-AKT-CCL2 Pathways in Renal Tubule Injury during Calcium Oxalate Crystal Formation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0690. [PMID: 40330661 PMCID: PMC12053376 DOI: 10.34133/research.0690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
Oxalate-induced crystalline kidney injury is a common form of crystal nephropathy. The accumulation of calcium oxalate (CaOx) crystal could lead to renal epithelium injury and inflammation. The underlying cellular events in kidney after CaOx crystal formation are largely unknown. This study was aimed to gain a better understanding of mouse kidney function in the development of renal CaOx formation. The study utilized a mouse CaOx model to analyze the cellular response at 5 time points using single-cell RNA sequencing and investigate the interaction of different cells during renal CaOx crystal formation. Additionally, the study investigated the communication between these cells and macrophages, as well as the role of chemokines in recruiting infiltrating macrophages. RNA velocity analysis uncovered an alternative differentiation pathway for injured and S1 proximal tubule cells, which mainly communicate with macrophages through the SPP1-CD44 pair, along with the expression of proinflammatory factors and stone matrix genes during renal CaOx crystal formation. Furthermore, resident Fn1 macrophages were found to express chemokines, such as CCL2, which recruited infiltrating macrophages. The CCL2 secretion was mediated by the CD44-AKT pathway. Blocking CCL2 decreased the expression of injury markers in the kidney, including CLU, LCN2, and KIM-1, and inhibited CaOx crystal deposition. The study identified potential cell types and target genes involved in renal tubule injury in oxalate-related crystal nephropathy. The findings shed light on the cellular processes that contribute to the formation and damage caused by CaOx crystals within the kidney, which could lead to the development of potential cell types and target genes for treating this condition.
Collapse
Affiliation(s)
- Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Zhongyu Jian
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yucheng Ma
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jun Wen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ningning Chao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, Frontiers Science Center for Disease-related Molecular Network, Sichuan Provincial Engineering Laboratory of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, West China School of Medicine,
Sichuan University, Chengdu 610041, Sichuan Province, P.R. China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Liyuan Xiang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yiqiong Yuan
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Linhu Liu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ya Li
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jingwen Wei
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Banghua Liao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health, State Key Laboratory of Respiratory Health and Multimorbidity, Frontiers Science Center for Disease-related Molecular Network, Sichuan Provincial Engineering Laboratory of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, West China School of Medicine,
Sichuan University, Chengdu 610041, Sichuan Province, P.R. China
| | - Kunjie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital,
Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
4
|
Zhang C, Xiang Z, Yang P, Zhang L, Deng J, Liao X. Advances in Nano-Immunomodulatory Systems for the Treatment of Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409190. [PMID: 40145715 PMCID: PMC12061249 DOI: 10.1002/advs.202409190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/26/2025] [Indexed: 03/28/2025]
Abstract
Acute kidney injury (AKI) occurs when there is an imbalance in the immune microenvironment, leading to ongoing and excessive inflammation. Numerous immunomodulatory therapies have been suggested for the treatment of AKI, the current immunomodulatory treatment delivery systems are suboptimal and lack efficiency. Given the lack of effective treatment, AKI can result in multi-organ dysfunction and even death, imposing a significant healthcare burden on both the family and society. This underscores the necessity for innovative treatment delivery systems, such as nanomaterials, to better control pathological inflammation, and ultimately enhance AKI treatment outcomes. Despite the modification of numerous immunomodulatory nanomaterials to target the AKI immune microenvironment with promising therapeutic results, the literature concerning their intersection is scarce. In this article, the pathophysiological processes of AKI are outlined, focusing on the immune microenvironment, discuss significant advances in the comprehension of AKI recovery, and describe the multifunctionality and suitability of nanomaterial-based immunomodulatory treatments in managing AKI. The main obstacles and potential opportunities in the swiftly advancing research field are also clarified.
Collapse
Affiliation(s)
- Chenli Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Zeli Xiang
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Pengfei Yang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Ling Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Jun Deng
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Institute of Burn Research, Southwest HospitalState Key Lab of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xiaohui Liao
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| |
Collapse
|
5
|
Kojima H, Morinelli TA, Wang Y, Chin JL, Meyer AS, Kao YC, Kadono K, Yao S, Torgerson T, Dery KJ, Bhat A, Reed EF, Kaldas FM, van der Windt DJ, Farmer DG, Kupiec-Weglinski JW, Zhai Y. Group 1 innate lymphoid cells protect liver transplants from ischemia-reperfusion injury via an interferon gamma-mediated pathway. Am J Transplant 2025; 25:969-984. [PMID: 39736469 PMCID: PMC12043423 DOI: 10.1016/j.ajt.2024.11.035] [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: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/23/2024] [Indexed: 01/01/2025]
Abstract
As important immune regulatory cells, whether innate lymphoid cells (ILCs) are involved in liver transplantation (LT) remains unclear. In a murine orthotopic LT model, we dissected roles of ILCs in liver ischemia-reperfusion injury (IRI). Wild-type (WT) grafts suffered significantly higher IRI in Rag2-γc double knockout (DKO) than Rag2 knockout (KO) recipients, in association with downregulation of group 1 ILCs genes, including interferon gamma. Antibody-mediated ILC depletion or interferon gamma neutralization in Rag2 KO recipients increased, while interferon gamma treatment in DKO recipients reduced, liver graft injuries. At the donor side, grafts from DKO mice or anti-NK1.1-treated WT mice suffered significantly higher IRI, while grafts treated with interferon gamma during cold preservation decreased IRI. Thus, both recipient and donor group 1 ILCs protect liver grafts from IRI. Low-dose interferon gamma upregulated c-FLIP expression in vitro and in vivo and protected hepatocytes from inflammatory cell death. In human liver graft biopsies, single-cell RNA-sequencing analysis revealed group 1 ILCs produce interferon gamma. The c-FLIP levels were positively correlated with interferon gamma in pretransplant biopsies. Grafts with higher c-FLIP were associated with lower caspase-8 activation, IRI gradings, and frequency of early allograft dysfunction post-LT. Our study reveals a novel interferon gamma-mediated cytoprotective role of group 1 ILCs in LT.
Collapse
Affiliation(s)
- Hidenobu Kojima
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Thomas A Morinelli
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yue Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jackson L Chin
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Yi-Chu Kao
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kentaro Kadono
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Siyuan Yao
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Taylor Torgerson
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kenneth J Dery
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Adil Bhat
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Fady M Kaldas
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Dirk J van der Windt
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Douglas G Farmer
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jerzy W Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yuan Zhai
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
6
|
Naz F, Uddin MJ, Hagspiel N, Young MK, Tyus D, Boone R, Brown AC, Ramakrishnan G, Rigo I, Fleming C, Madden GR, Petri WA. IL-33 protects from recurrent C. difficile infection by restoration of humoral immunity. J Clin Invest 2025; 135:e184659. [PMID: 40048372 PMCID: PMC12043089 DOI: 10.1172/jci184659] [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: 07/09/2024] [Accepted: 02/27/2025] [Indexed: 03/12/2025] Open
Abstract
Clostridioides difficile infection (CDI) recurs in 1 of 5 patients. Monoclonal antibodies targeting the virulence factor TcdB reduce disease recurrence, suggesting that an inadequate anti-TcdB response to CDI leads to recurrence. In patients with CDI, we discovered that IL-33 measured at diagnosis predicts future recurrence, leading us to test the role of IL-33 signaling in the induction of humoral immunity during CDI. Using a mouse recurrence model, IL-33 was demonstrated to be integral for anti-TcdB antibody production. IL-33 acted via ST2+ ILC2 cells, facilitating germinal center T follicular helper (GC-Tfh) cell generation of antibodies. IL-33 protection from reinfection was antibody-dependent, as μMT KO mice and mice treated with anti-CD20 mAb were not protected. These findings demonstrate the critical role of IL-33 in generating humoral immunity to prevent recurrent CDI.
Collapse
Affiliation(s)
- Farha Naz
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Md Jashim Uddin
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Nicholas Hagspiel
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Mary K. Young
- Department of Medicine, Division of Infectious Diseases and International Health
| | - David Tyus
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Rachel Boone
- Department of Microbiology, Immunology, and Cancer Biology, and
| | - Audrey C. Brown
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Girija Ramakrishnan
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Isaura Rigo
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Claire Fleming
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Gregory R. Madden
- Department of Medicine, Division of Infectious Diseases and International Health
| | - William A. Petri
- Department of Medicine, Division of Infectious Diseases and International Health
- Department of Microbiology, Immunology, and Cancer Biology, and
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
7
|
Meng XM, Wang L, Nikolic-Paterson DJ, Lan HY. Innate immune cells in acute and chronic kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00958-x. [PMID: 40263532 DOI: 10.1038/s41581-025-00958-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/24/2025]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are inter-related clinical and pathophysiological disorders. Cells of the innate immune system, such as granulocytes and macrophages, can induce AKI through the secretion of pro-inflammatory mediators such as cytokines, chemokines and enzymes, and the release of extracellular traps. In addition, macrophages and dendritic cells can drive the progression of CKD through a wide range of pro-inflammatory and pro-fibrotic mechanisms, and by regulation of the adaptive immune response. However, innate immune cells can also promote kidney repair after acute injury. These actions highlight the multifaceted nature of the way by which innate immune cells respond to signals within the kidney microenvironment, including interaction with the complement and coagulation cascades, cells of the adaptive immune system, intrinsic renal cells and infiltrating mesenchymal cells. The factors and mechanisms that underpin the ability of innate immune cells to contribute to renal injury or repair and to drive the progression of CKD are of great interest for understanding disease processes and for developing new therapeutic approaches to limit AKI and the AKI-to-CKD transition.
Collapse
Affiliation(s)
- Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre and Monash University Centre for Inflammatory Diseases, Melbourne, Victoria, Australia
| | - Hui-Yao Lan
- Research Center of Integrated Traditional Chinese and Western Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
- Departments of Medicine & Therapeutics, the Chinese University of Hong Kong, Hong Kong, and Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China.
| |
Collapse
|
8
|
He A, Yang Y, Kotsch K, Sattler A. Impact of Organ Donor Pretreatment With Anti-Thymocyte Globulin in a Murine Model of Allogenic Kidney Transplantation. Transpl Int 2025; 37:13997. [PMID: 39839912 PMCID: PMC11745874 DOI: 10.3389/ti.2024.13997] [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: 10/28/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025]
Abstract
Kidney transplantation is the treatment of choice for end-stage organ failure. To improve transplantation outcomes, particularly of "marginal" organs from extended criteria donors (ECD), attempts have been made to therapeutically modulate donor or graft pre-transplantation. Anti-thymocyte globulin (ATG) has a history as lymphocyte-depleting, immunosuppressive drug for treating rejection episodes post transplantation. In this study, however, we aimed to comprehensively analyze the effects of ATG donor pre-conditioning in a mouse model of kidney transplantation. ATG pre-treatment of potential donors led to a broad depletion of T- and NK cells in peripheral blood, non-lymphoid (including kidney) and lymphoid organs within 48 h, whereas myeloid cells were spared. ATG was also effectively depleting renal innate lymphoid type 1 and 2 cells. Importantly, transplantation of kidneys from ATG pre-treated donors into fully mismatched recipients showed only mild effects on leukocyte re-composition post transplantation. In line with this, serum creatinine and urea levels were similar in animals receiving kidneys from ATG treated donors or controls, demonstrating that donor treatment had no effect on allograft function in the early post-transplantation phase. In summary, our findings are suggestive of a more cell-type-specific depletion strategy in concert with an experimental model better reflecting aspects of clinical transplantation.
Collapse
Affiliation(s)
- An He
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
- Department of Urology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yiren Yang
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - Katja Kotsch
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - Arne Sattler
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| |
Collapse
|
9
|
Sabapathy V, Price A, Cheru NT, Venkatadri R, Dogan M, Costlow G, Mohammad S, Sharma R. ST2 + T-Regulatory Cells in Renal Inflammation and Fibrosis after Ischemic Kidney Injury. J Am Soc Nephrol 2025; 36:73-86. [PMID: 39186386 PMCID: PMC11706559 DOI: 10.1681/asn.0000000000000471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
Key Points IL-33/ST2 alarmin pathway regulates inflammation, fibrosis, and resolution of ischemia-reperfusion injury of kidneys. ST2 regulates the transcriptome of T-regulatory cells related to suppressive and reparative functions. The secretome of ST2+ T-regulatory cells regulates hypoxic injury in an amphiregulin-dependent manner. Background Inflammation is a major cause of kidney injury. IL-1 family cytokine IL-33 is released from damaged cells and modulates the immune response through its receptor ST2 expressed on many cell types, including regulatory T cells (Tregs). Although a proinflammatory role of IL-33 has been proposed, exogenous IL-33 expanded Tregs and suppressed renal inflammation. However, the contribution of endogenous IL-33/ST2 for the role of Tregs in the resolution of kidney injury has not been investigated. Methods We used murine renal ischemia-reperfusion injury and kidney organoids (KDOs) to delineate the role of the ST2 and amphiregulin (AREG) specifically in Tregs using targeted deletion. Bulk and single-cell RNA sequencing were performed on flow-sorted Tregs from spleen and CD4 T cells from postischemic kidneys, respectively. The protective role of ST2-sufficient Tregs was analyzed using a novel coculture system of syngeneic KDOs and Tregs under hypoxic conditions. Results Bulk RNA sequencing of splenic and single-cell RNA sequencing of kidney CD4 T cells showed that ST2+ Tregs are enriched for genes related to Treg proliferation and function. Genes for reparative factors, such as Areg , were also enriched in ST2+ Tregs. Treg-specific deletion of ST2 or AREG exacerbated kidney injury and fibrosis in the unilateral ischemia-reperfusion injury model. In coculture studies, wild-type but not ST2-deficient Tregs preserved hypoxia-induced loss of kidney organoid viability, which was restored by AREG supplementation. Conclusions Our study identified the role of the IL-33/ST2 pathway in Tregs for resolution of kidney injury. The transcriptome of ST2+ Tregs was enriched for reparative factors including Areg . Lack of ST2 or AREG in Tregs worsened kidney injury. Tregs protected KDOs from hypoxia in a ST2- and AREG-dependent manner.
Collapse
Affiliation(s)
- Vikram Sabapathy
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Airi Price
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of International Health, Georgetown University, Washington, DC
| | - Nardos Tesfaye Cheru
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut
| | - Rajkumar Venkatadri
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Immunology Research Unit, GlaxoSmithKline (GSK), Collegeville, Pennsylvania
| | - Murat Dogan
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Transplant Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Gabrielle Costlow
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Saleh Mohammad
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
10
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024; 20:789-805. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
11
|
Naz F, Hagspiel N, Young MK, Uddin J, Tyus D, Boone R, Brown AC, Ramakrishnan G, Rigo I, Madden GR, Petri WA. IL-33 protects from recurrent C. difficile infection by restoration of humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.623943. [PMID: 39605647 PMCID: PMC11601440 DOI: 10.1101/2024.11.16.623943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Clostridioides difficile infection (CDI) recurs in one of five patients. Monoclonal antibodies targeting the virulence factor TcdB reduce disease recurrence, suggesting that an inadequate anti-TcdB response to CDI leads to recurrence. In patients with CDI, we discovered that IL-33 measured at diagnosis predicts future recurrence, leading us to test the role of IL-33 signaling in the induction of humoral immunity during CDI. Using a mouse recurrence model, IL-33 was demonstrated to be integral for anti-TcdB antibody production. IL-33 acted via ST2+ ILC2 cells, facilitating germinal center T follicular helper (GC-Tfh) cell generation of antibodies. IL-33 protection from reinfection was antibody-dependent, as μMT KO mice and mice treated with anti-CD20 mAb were not protected. These findings demonstrate the critical role of IL-33 in generating humoral immunity to prevent recurrent CDI.
Collapse
Affiliation(s)
- Farha Naz
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Nicholas Hagspiel
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Mary K. Young
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Jashim Uddin
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - David Tyus
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Rachel Boone
- Department of Microbiology, Immunology and Cancer Biology, Charlottesville, Virginia, USA
| | - Audrey C. Brown
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Girija Ramakrishnan
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Isaura Rigo
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Gregory R. Madden
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - William A. Petri
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
12
|
Amin G, Ghali R, Habeichi NJ, Mallat Z, Booz GW, Zouein FA. Dual Time-Dependent Effects of Interleukin-33 Administration on the Kidney Postmyocardial Infarction. J Interferon Cytokine Res 2024; 44:496-509. [PMID: 39311715 PMCID: PMC11631796 DOI: 10.1089/jir.2024.0127] [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: 06/21/2024] [Accepted: 08/03/2024] [Indexed: 10/04/2024] Open
Abstract
Kidney damage is a serious prevalent complication that occurs after a myocardial infarction (MI) and is associated with worse outcomes. Interleukin-33 (IL-33), a member of the IL-1 superfamily, functions as an alarmin that is released upon necrosis or tissue damage to alert immune cells expressing the ST2L receptor. IL-33 is increased in kidney disease, and recent studies have shown that the IL-33/ST2 axis is instrumental in both disease progression and repair. In this study, we investigated the effect of IL-33 administration on kidneys in C57BL6/J male mice 4 and 7 days after the induction of MI. The mice received either IL-33 or vehicle (PBS) treatment. Cardiac systolic function and systemic inflammation were assessed, and kidneys were subjected to histological and molecular analysis. The administration of IL-33 for 4 days post-MI improved renal structure consistent with reduced expression of profibrotic markers, reduced apoptosis, and increased expression of the anti-inflammatory cytokine IL-4. In addition, IL-33 administration enhanced the levels of Sirtuin3, nicotinamide phosphoribosyltransferase, and the renal nicotinamide adenine dinucleotide pool which are critical for mitochondrial function and energy production, indicating metabolic benefits. However, this protection seems to be lost with the continued administration of IL-33 for 7 days post-MI coinciding with aggravated cardiac dysfunction and increased systemic inflammation. These findings demonstrate that while IL-33 treatment can help improve kidney damage post-MI in the short term, extended treatment may not be beneficial. This may be due to the direct effects of IL-33 on the kidneys or indirectly mediated by adverse cardiac remodeling influencing the cardiorenal crosstalk.
Collapse
Affiliation(s)
- Ghadir Amin
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rana Ghali
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
| | - Nada J. Habeichi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Signaling and Cardiovascular Pathophysiology, Institute Paris-Saclay for Therapeutic Innovation, Faculty of Pharmacy, University Paris Saclay, Gif-sur-Yvette, France
| | - Ziad Mallat
- Department of Medicine, Section of Cardio Respiratory Medicine, Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- Paris Cardiovascular Research Center, Institut National de la Sante et de la Recherche Medicale (Inserm), Paris, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Signaling and Cardiovascular Pathophysiology, Institute Paris-Saclay for Therapeutic Innovation, Faculty of Pharmacy, University Paris Saclay, Gif-sur-Yvette, France
| |
Collapse
|
13
|
Chen L, Gao C, Yin X, Mo L, Cheng X, Chen H, Jiang C, Wu B, Zhao Y, Li H, Li Y, Li J, Chen L, Deng Q, Yao P, Tang Y. Partial reduction of interleukin-33 signaling improves senescence and renal injury in diabetic nephropathy. MedComm (Beijing) 2024; 5:e742. [PMID: 39465143 PMCID: PMC11502718 DOI: 10.1002/mco2.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 10/29/2024] Open
Abstract
Diabetic nephropathy (DN) is a frequent and costly complication of diabetes with limited understandings of mechanisms and therapies. Emerging evidence points to the important roles of interleukin-33 (IL-33) in acute kidney injury, yet its contribution to DN is still unclear. We here found a ubiquitous increase of IL-33 and its receptor (ST2) in murine models and patients with DN. Surprisingly, both IL-33 and ST2 knockdown aggravated renal lesions in DN, while overexpression of IL-33 also exacerbated the condition. Further population-based analyses revealed a positive correlation of IL-33 expression with renal dysfunction in DN patients. Individuals with high IL-33 expression-related polygenic risk score had a higher DN risk. These findings confirmed the harmful effects of IL-33 on DN. Conversely, endogenous and exogenous partial reduction of IL-33 signaling conferred renoprotective effects in vivo and in vitro. Mechanistically, IL-33 induced senescence by regulating cell cycle factors in HK-2 cells, and accordingly senescence led to renal cell damage through the secretion of senescence-related secretory phenotype (SASP) including IL-33 and prostaglandins. Together, elevated IL-33 accelerates cellular senescence to drive DN possibly by SASP production, while a partial blockage improves renal injury and senescence. Our findings pinpoint a possible and new avenue for DN interventions.
Collapse
Affiliation(s)
- Li Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Oil Crops Research Institute of the Chinese Academy of Agricultural SciencesHubei Key Laboratory of Lipid Chemistry and Nutritionand Key Laboratory of Oilseeds ProcessingMinistry of AgricultureOil Crops and Lipids Process Technology National & Local Joint Engineering LaboratoryWuhanHubeiChina
| | - Chao Gao
- National Institute for Nutrition and HealthChinese Center for Disease Control and Prevention BeijingBeijingChina
| | - Xingzhu Yin
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Mo
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xueer Cheng
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huimin Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chunjie Jiang
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bangfu Wu
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ying Zhao
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongxia Li
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yanyan Li
- Shenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jiansha Li
- Institute of PathologyTongji HospitalWuhanChina
- Department of PathologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Liangkai Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qianchun Deng
- Oil Crops Research Institute of the Chinese Academy of Agricultural SciencesHubei Key Laboratory of Lipid Chemistry and Nutritionand Key Laboratory of Oilseeds ProcessingMinistry of AgricultureOil Crops and Lipids Process Technology National & Local Joint Engineering LaboratoryWuhanHubeiChina
| | - Ping Yao
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuhan Tang
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
14
|
Fan Y, Wang C, Dai W, Zhou Y, Lu G, Li W, Li L, Lin T. DNA Origami Enhanced Cytokine Immunotherapy for Alleviating Renal Ischemia-Reperfusion Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38979-38988. [PMID: 39029244 DOI: 10.1021/acsami.4c06110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) is a major contributing factor to the development of acute kidney injury (AKI) and has resulted in considerable morbidity and mortality. Persistent inflammatory responses and excessive reactive oxygen species (ROS) in the kidney following IRI can severely delay tissue repair, making it challenging to effectively promote IRI regeneration. Herein, we report an approach to enhance immunotherapy using interleukin-10 (IL-10) to promote IRI regeneration by loading IL-10 onto rectangular DNA origami nanostructures (rDON). rDON can significantly enhance the renal accumulation and retention time of IL-10, enabling it to effectively polarize type 1 macrophages into type 2 macrophages, thereby significantly reducing proinflammatory factors and increasing anti-inflammatory factors. In addition, DNA origami helps mitigate the harmful effects of ROS during renal IRI. The administration of IL-10-loaded DNA origami effectively improves kidney function, resulting in a notable reduction in blood urea nitrogen, serum uric acid, and serum creatinine levels. Our study demonstrates that the integration of anti-inflammatory cytokines within DNA origami holds promise as a strategic approach for cytokine immunotherapy in patients with AKI and other renal disorders.
Collapse
Affiliation(s)
- Yu Fan
- Department of Urology, Institute of Urology and Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengshi Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshu Dai
- NHC Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaojia Zhou
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gonggong Lu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Tao Lin
- Department of Urology, Institute of Urology and Organ Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
15
|
Chen Z, Li J, Liu X, Liu X, Zhu J, Tang X, Deng Y, Chen C. Predictive Value of Serum Soluble ST2 in Adult Patients Undergoing Cardiac Surgery for Acute Kidney Injury. Cardiorenal Med 2024; 14:498-507. [PMID: 39074462 DOI: 10.1159/000540529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
INTRODUCTION Cardiac surgery is related to an increased risk of postoperative acute kidney injury (AKI). Serum soluble ST2 (sST2) is highly predictive of several cardiovascular diseases and may also be involved in renal injury. This study explored the relationship between serum sST2 levels measured at intensive care unit (ICU) admission and the development of AKI after cardiac surgery. METHODS We prospectively conducted an investigation on consecutive patients who underwent cardiac surgery. sST2 was immediately measured at ICU admission. The relationship between the levels of sST2 and the development of AKI was explored using stepwise logistic regression. RESULTS Among the 500 patients enrolled, AKI was observed in 207 (41%) patients. Serum sST2 levels in AKI patients were higher than those without AKI (61.46 ng/mL [46.52, 116.25] vs. 38.91 ng/mL [28.74, 50.93], p < 0.001). Additionally, multivariable logistic regression analysis showed that as progressively higher tertiles of serum sST2, the odds ratios (ORs) of AKI gradually increased (adjusted ORs of 1.97 [95% CI, 1.13-3.45], and 4.27 [95% CI, 2.36-7.71] for tertiles 2 and 3, respectively, relative to tertile 1, p < 0.05). The addition of sST2 further improved reclassification (p < 0.001) and discrimination (p < 0.001) over the basic model, which included established risk factors. CONCLUSION Serum sST2 levels at ICU admission were associated with the development of postoperative AKI and improved the identification of AKI after cardiac surgery.
Collapse
Affiliation(s)
- Zeling Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaxin Li
- Department of Intensive Care Unit of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xicheng Liu
- Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Xiaolong Liu
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Junjiang Zhu
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xuanhe Tang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yiyu Deng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
16
|
陶 怀, 骆 金, 闻 志, 虞 亘, 苏 萧, 王 鑫, 关 翰, 陈 志. [High STING expression exacerbates renal ischemia-reperfusion injury in mice by regulating the TLR4/NF-κB/NLRP3 pathway and promoting inflammation and apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1345-1354. [PMID: 39051080 PMCID: PMC11270667 DOI: 10.12122/j.issn.1673-4254.2024.07.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Indexed: 07/27/2024]
Abstract
OBJECTIVE To investigate renal expression level of STING in mice with renal ischemia-reperfusion injury (IRI) and its regulatory role in IRI. METHODS C57BL/6 mice were divided into sham operation group, IRI (induced by clamping the renal artery) model group, IRI+DMSO treatment group, and IRI+SN-011 treatment group. Serum creatinine and blood urea nitrogen of the mice were analyzed, and pathological changes in the renal tissue were assessed with PAS staining. RT-qPCR, ELISA, Western blotting, and immunohistochemistry were used to detect the expression levels of STING, KIM-1, Bcl-2, Bax, caspase-3, TLR4, P65, NLRP3, caspase-1, CD68, MPO, IL-1β, IL-6, and TNF-α in the renal tissues. In the cell experiment, HK-2 cells exposed to hypoxia-reoxygenation (H/R) were treated with DMSO or SN-011, and cellular STING expression levels and cell apoptosis were analyzed using RT-qPCR, Western blotting or flow cytometry. RESULTS In C57BL/6 mice, renal IRI induced obvious renal tissue damage, elevation of serum creatinine and blood urea nitrogen levels and renal expression levels of KIM-1, STING, TLR4, P65, NLRP3, caspase-1, caspase-3, Bax, CD68, MPO, IL-1β, IL-6, and TNF-α, and reduction of Bcl-2 expression level. Treatment of the mouse models with SN-011 for inhibiting STING expression significantly alleviated these changes. In HK-2 cells, H/R exposure caused significant elevation of cellular STING expression and obviously increased cell apoptosis rate, which was significantly lowered by treatment with SN-011. CONCLUSION Renal STING expression is elevated in mice with renal IRI to exacerbate renal injury by regulating the TLR4/NF-κB/NLRP3 pathway and promoting inflammation and apoptosis in the renal tissues.
Collapse
|
17
|
Ryu S, Kim KA, Kim J, Lee DH, Bae YS, Lee H, Kim BC, Kim HY. The protective roles of integrin α4β7 and Amphiregulin-expressing innate lymphoid cells in lupus nephritis. Cell Mol Immunol 2024; 21:723-737. [PMID: 38806623 PMCID: PMC11214630 DOI: 10.1038/s41423-024-01178-2] [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: 08/03/2023] [Accepted: 04/27/2024] [Indexed: 05/30/2024] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) have emerged as key regulators of the immune response in renal inflammatory diseases such as lupus nephritis. However, the mechanisms underlying ILC2 adhesion and migration in the kidney remain poorly understood. Here, we revealed the critical role of integrin α4β7 in mediating renal ILC2 adhesion and function. We found that integrin α4β7 enables the retention of ILC2s in the kidney by binding to VCAM-1, E-cadherin, or fibronectin on structural cells. Moreover, integrin α4β7 knockdown reduced the production of the reparative cytokine amphiregulin (Areg) by ILC2s. In lupus nephritis, TLR7/9 signaling within the kidney microenvironment downregulates integrin α4β7 expression, leading to decreased Areg production and promoting the egress of ILC2s. Notably, IL-33 treatment upregulated integrin α4β7 and Areg expression in ILC2s, thereby enhancing survival and reducing inflammation in lupus nephritis. Together, these findings highlight the potential of targeting ILC2 adhesion as a therapeutic strategy for autoimmune kidney diseases.
Collapse
Affiliation(s)
- Seungwon Ryu
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Kyung Ah Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Jinwoo Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, 03080, South Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, 03080, South Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, South Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Hajeong Lee
- Division of Nephrology, Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Byoung Choul Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, South Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
18
|
Chen Y, Gu X, Cao K, Tu M, Liu W, Ju J. The role of innate lymphoid cells in systemic lupus erythematosus. Cytokine 2024; 179:156623. [PMID: 38685155 DOI: 10.1016/j.cyto.2024.156623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Systemic lupus erythematosus (SLE) is a connective tissue disorder that affects various body systems. Both the innate and adaptive immunity contribute to the onset and progression of SLE. The main mechanism of SLE is an excessive immune response of immune cells to autoantigens, which leads to systemic inflammation and inflammation-induced organ damage. Notably, a subset of innate immune cells known as innate lymphoid cells (ILCs) has recently emerged. ILCs are pivotal in the early stages of infection; participate in immune responses, inflammation, and tissue repair; and regulate the immune function of the body by resisting pathogens and regulating autoimmune inflammation and metabolic homeostasis. Thus, ILCs dysfunction can lead to autoimmune diseases. This review discusses the maturation of ILCs, the potential mechanisms by which ILCs exacerbate SLE pathogenesis, and their contributions to organ inflammatory deterioration in SLE.
Collapse
Affiliation(s)
- Yong Chen
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Xiaotian Gu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Kunyu Cao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Miao Tu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China
| | - Wan Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Jiyu Ju
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
19
|
Nagashima R, Ishikawa H, Kuno Y, Kohda C, Eshima K, Iyoda M. Group2 innate lymphoid cells ameliorate renal fibrosis and dysfunction associated with adenine-induced CKD. Cell Immunol 2024; 401-402:104828. [PMID: 38759328 DOI: 10.1016/j.cellimm.2024.104828] [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: 01/23/2024] [Revised: 03/27/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Renal fibrosis is a common pathway of chronic kidney disease (CKD) progression involving primary kidney injury and kidney diseases. Group 2 innate lymphoid cells (ILC2s) mediate type 2 immune responses irrespective of antigen presentation and play a reno-protective role in kidney injury and disease. In the present study, we observed a decrease in kidney-resident ILC2s in CKD and found that enrichment of ILC2s in the kidney ameliorates renal fibrosis. In CKD kidney, ILC2s preferentially produced IL-13 over IL-5 in response to IL-33 stimulation, regardless of ST2L expression. Moreover, GATA3 expression was decreased in ILC2s, and T-bet+ ILC1s and RORγt+ ILC3s were increased in CKD kidney. Adoptive transfer of kidney ILC2s into adenine-induced CKD model mouse improved renal function and fibrosis. Renal fibroblasts cultured with IL33-activated kidney ILC2s suppressed myofibroblast trans-differentiation through Acta2 and Fn-1 regulation. These results suggest that kidney ILC2s prevent CKD progression via improvement of renal fibrosis. Our findings also suggest that ILC2s may contribute to the development of new therapeutic agents and strategies for tissue fibroses.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Immunology, Department of Biosciences, Kitasato University School of Science, Japan.
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan
| | - Koji Eshima
- Division of Immunology, Department of Biosciences, Kitasato University School of Science, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Japan
| |
Collapse
|
20
|
Hofherr A, Liarte Marin E, Musial B, Seth A, Slidel T, Conway J, Baker D, Hansen PB, Challis B, Bartesaghi S, Bhat M, Pecoits-Filho R, Tu X, Selvarajah V, Woollard K, Heerspink HJ. Inhibition of Interleukin-33 to Reduce Glomerular Endothelial Inflammation in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1876-1891. [PMID: 38899206 PMCID: PMC11184260 DOI: 10.1016/j.ekir.2024.03.009] [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: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Inflammation is a significant contributor to cardiorenal morbidity and mortality in diabetic kidney disease (DKD). The pathophysiological mechanisms linking systemic, subacute inflammation and local, kidney injury-initiated immune maladaptation is partially understood. Methods Here, we explored the expression of proinflammatory cytokines in patients with DKD; investigated mouse models of type 1 and type 2 diabetes (T2D); evaluated glomerular signaling in vitro; performed post hoc analyses of systemic and urinary markers of inflammation; and initiated a phase 2b clinical study (FRONTIER-1; NCT04170543). Results Transcriptomic profiling of kidney biopsies from patients with DKD revealed significant glomerular upregulation of interleukin-33 (IL-33). Inhibition of IL-33 signaling reduced glomerular damage and albuminuria in the uninephrectomized db/db mouse model (T2D/DKD). On a cellular level, inhibiting IL-33 improved glomerular endothelial health by decreasing cellular inflammation and reducing release of proinflammatory cytokines. Therefore, FRONTIER-1 was designed to test the safety and efficacy of the IL-33-targeted monoclonal antibody tozorakimab in patients with DKD. So far, 578 patients are enrolled in FRONTIER-1. The baseline inflammation status of participants (N > 146) was assessed in blood and urine. Comparison to independent reference cohorts (N > 200) validated the distribution of urinary tumor necrosis factor receptor 1 (TNFR1) and C-C motif chemokine ligand 2 (CCL2). Treatment with dapagliflozin for 6 weeks did not alter these biomarkers significantly. Conclusion We show that blocking the IL-33 pathway may mitigate glomerular endothelial inflammation in DKD. The findings from the FRONTIER-1 study will provide valuable insights into the therapeutic potential of IL-33 inhibition in DKD.
Collapse
Affiliation(s)
- Alexis Hofherr
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elena Liarte Marin
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Barbara Musial
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Asha Seth
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Tim Slidel
- Bioinformatics, Oncology R&D, AstraZeneca, Cambridge, UK
| | - James Conway
- Bioinformatics, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - David Baker
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Pernille B.L. Hansen
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benjamin Challis
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefano Bartesaghi
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Bhat
- Translational Science and Experimental Medicine, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Roberto Pecoits-Filho
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
- School of Medicine, Pontificia Universidade de Catolica do Parana, Curitiba, Brazil
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Xiao Tu
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Viknesh Selvarajah
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kevin Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Hiddo J.L. Heerspink
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Xu L, Xing Z, Yuan J, Han Y, Jiang Z, Han M, Hou X, Xing W, Li Z. Ultrasmall Nanoparticles Regulate Immune Microenvironment by Activating IL-33/ST2 to Alleviate Renal Ischemia-Reperfusion Injury. Adv Healthc Mater 2024; 13:e2303276. [PMID: 38335143 DOI: 10.1002/adhm.202303276] [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: 09/27/2023] [Revised: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) is a common disease with high morbidity and mortality. Renal IRI can cause the disorder of immune microenvironment and reprograming the immune microenvironment to alleviate excessive inflammatory response is crucial for its treatment. Cytokine IL-33 can improve the immune inflammatory microenvironment by modulating both innate and adaptive immune cells, and serve as an important target for modulating immune microenvironment of renal IRI. Herein, we report that bilobetin-functionalized ultrasmall Cu2- xSe nanoparticles (i.e., CSPB NPs) can activate the PKA/p-CREB/IL-33/ST2 signaling pathway to regulate innate and adaptive immune cells for reprograming the immune microenvironment of IRI-induced acute kidney injury. The biocompatible CSPB NPs can promote the polarization of M1-like macrophages into M2-like macrophages, and the expansion of ILC2 and Treg cells by activating IL-33/ST2 to modulate the excessive immune inflammatory response of renal IRI. More importantly, they can rapidly accumulate at the injured kidney to significantly alleviate IRI. This work demonstrates that modulating the expression of cytokines to reprogram immune microenvironment has great potential in the treatment of renal IRI and other ischemic diseases.
Collapse
Affiliation(s)
- Liyao Xu
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Zhaoyu Xing
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
| | - Jiaxin Yuan
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Xianao Hou
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| | - Wei Xing
- Department of Radiology, Affiliated Hospital 3, Soochow University, Changzhou, 213003, P. R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P. R. China
| |
Collapse
|
22
|
Tian X, Wu L, Li X, Zheng W, Zuo H, Song H. Exosomes derived from bone marrow mesenchymal stem cells alleviate biliary ischemia reperfusion injury in fatty liver transplantation by inhibiting ferroptosis. Mol Cell Biochem 2024; 479:881-894. [PMID: 37243945 PMCID: PMC11016128 DOI: 10.1007/s11010-023-04770-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
Fatty liver grafts are susceptible to ischemia reperfusion injury (IRI), increasing the risk of biliary complications after liver transplantation (LT). Ferroptosis, a newly recognized programmed cell death, is expected to be a novel therapeutic target for IRI. We investigated whether exosomes derived from heme oxygenase 1-modified bone marrow mesenchymal stem cells (HExos) relieve ferroptosis and protect biliary tracts from IRI in a rat fatty liver transplantation model. Rats were fed with a methionine choline deficient (MCD) diet for 2 weeks to induce severe hepatic steatosis. Steatotic grafts were implanted and HExos were administered after liver transplantation. A series of functional assays and pathological analysis were performed to assess ferroptosis and biliary IRI. The HExos attenuated IRI following liver transplantation, as demonstrated by less ferroptosis, improved liver function, less Kupffer and T cell activation, and less long-term biliary fibrosis. MicroRNA (miR)-204-5p delivered by HExos negatively regulated ferroptosis by targeting a key pro-ferroptosis enzyme, ACSL4. Ferroptosis contributes to biliary IRI in fatty liver transplantation. HExos protect steatotic grafts by inhibiting ferroptosis, and may become a promising strategy to prevent biliary IRI and expand the donor pool.
Collapse
Affiliation(s)
- Xuan Tian
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Longlong Wu
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xiang Li
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
- NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, People's Republic of China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
- Tianjin Key Laboratory of Organ Transplantation, Tianjin, People's Republic of China.
| |
Collapse
|
23
|
Jang JY, Kim HW, Yan J, Kang TK, Lee W, Kim BS, Yang J. Interleukin-2/anti-interleukin-2 immune complex attenuates cold ischemia-reperfusion injury after kidney transplantation by increasing renal regulatory T cells. Clin Transl Med 2024; 14:e1631. [PMID: 38504554 PMCID: PMC10951489 DOI: 10.1002/ctm2.1631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Cold ischemia-reperfusion injury (IRI) is an unavoidable complication of kidney transplantation. We investigated the role of regulatory T cells (Treg) in cold IRI and whether the interleukin (IL)-2/anti-IL-2 antibody complex (IL-2C) can ameliorate cold IRI. METHODS We developed a cold IRI mouse model using kidney transplantation and analyzed the IL-2C impact on cold IRI in acute, subacute and chronic phases. RESULTS Treg transfer attenuated cold IRI, while Treg depletion aggravated cold IRI. Next, IL-2C administration prior to IRI mitigated acute renal function decline, renal tissue damage and apoptosis and inhibited infiltration of effector cells into kidneys and pro-inflammatory cytokine expression on day 1 after IRI. On day 7 after IRI, IL-2C promoted renal regeneration and reduced subacute renal damage. Furthermore, on day 28 following IRI, IL-2C inhibited chronic fibrosis. IL-2C decreased reactive oxygen species-mediated injury and improved antioxidant function. When IL-2C was administered following IRI, it also increased renal regeneration with Treg infiltration and suppressed renal fibrosis. In contrast, Treg depletion in the presence of IL-2C eliminated the positive effects of IL-2C on IRI. CONCLUSION Tregs protect kidneys from cold IRI and IL-2C inhibited cold IRI by increasing the renal Tregs, suggesting a potential of IL-2C in treating cold IRI. KEY POINTS Interleukin (IL)-2/anti-IL-2 antibody complex attenuated acute renal injury, facilitated subacute renal regeneration and suppressed chronic renal fibrosis after cold ischemia-reperfusion injury (IRI) by increasing the renal Tregs. IL-2/anti-IL-2 antibody complex decreased reactive oxygen species-mediated injury and improved antioxidant function. This study suggests the therapeutic potential of the IL-2/anti-IL-2 antibody complex in kidney transplantation-associated cold IR.
Collapse
Affiliation(s)
- Joon Young Jang
- Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Hyung Woo Kim
- Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Ji‐Jing Yan
- Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Tae Kyeom Kang
- Natural Product Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Wook‐Bin Lee
- Natural Product Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Beom Seok Kim
- Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Jaeseok Yang
- Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
24
|
Noel S, Kurzhagen JT, Lee SA, Sadasivam M, Hamad AR, Pierorazio PM, Rabb H. Kidney Immune Cell Characterization of Humanized Mouse Models. KIDNEY360 2024; 5:96-109. [PMID: 38037230 PMCID: PMC10833610 DOI: 10.34067/kid.0000000000000300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Experimental studies often fail to translate to clinical practice. Humanized mouse models are an important tool to close this gap. We immunophenotyped the kidneys of NOG (EXL) and NSG mouse strains engrafted with human CD34 + hematopoietic stem cells or PBMCs and compared with immune cell composition of normal human kidney. Human CD34 + hematopoietic stem cell engraftment results in steady renal immune cell populations in mouse kidney with key similarities in composition compared with human kidney. Successful translation of experimental mouse data to human diseases is limited because of biological differences and imperfect disease models. Humanized mouse models are being used to bring murine models closer to humans. However, data for application in renal immune cell-mediated diseases are rare. We therefore studied immune cell composition of three different humanized mouse kidneys and compared them with human kidney. NOG and NOGEXL mice engrafted with human CD34 + hematopoietic stem cells were compared with NSG mice engrafted with human PBMCs. Engraftment was confirmed with flow cytometry, and immune cell composition in kidney, blood, spleen, and bone marrow was analyzed in different models. The results from immunophenotyping of kidneys from different humanized mouse strains were compared with normal portions of human kidneys. We found significant engraftment of human immune cells in blood and kidney of all tested models. huNSG mice showed highest frequencies of hTCR + cells compared with huNOG and huNOGEXL in blood. huNOGEXL was found to have the highest hCD4 + frequency among all tested models. Non-T cells such as hCD20 + and hCD11c + cells were decreased in huNSG mice compared with huNOG and huNOGEXL. Compared with normal human kidney, huNOG and huNOGEXL mice showed representative immune cell composition, rather than huNSG mice. In summary, humanization results in immune cell infiltration in the kidney with variable immune cell composition of tested humanized mouse models and partially reflects normal human kidneys, suggesting potential use for translational studies.
Collapse
Affiliation(s)
- Sanjeev Noel
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Johanna T. Kurzhagen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sul A Lee
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mohanraj Sadasivam
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Abdel R.A. Hamad
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Phillip M. Pierorazio
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hamid Rabb
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
25
|
Kanazawa N, Iyoda M, Suzuki T, Tachibana S, Nagashima R, Honda H. Exploring the significance of interleukin-33/ST2 axis in minimal change disease. Sci Rep 2023; 13:18776. [PMID: 37907612 PMCID: PMC10618262 DOI: 10.1038/s41598-023-45678-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Minimal change disease (MCD), a common cause of idiopathic nephrotic syndrome, has been postulated to exhibit an association with allergic conditions. Recent studies revealed the crucial role of interleukin (IL)-33 in type 2 innate immunity. We hypothesized that development of MCD involves an IL-33-related immune response. We examined 49 patients with biopsy-proven MCD, 6 healthy volunteers, and 29 patients in remission. In addition to clinical features, serum and urinary levels of IL-33 and soluble suppression of tumorigenicity 2 protein (sST2), a secreted form of the receptor of IL-33, were analyzed. Although IL-33 was barely detectable in either MCD or control samples, sST2 levels at diagnosis were elevated in MCD patients. Serum sST2 levels of MCD patients were correlated with serum total protein level (r = - 0.36, p = 0.010) and serum creatinine level (r = 0.34, p = 0.016). Furthermore, the elevated sST2 levels were observed to decrease following remission. Immunofluorescence revealed IL-33 expression in the podocytes among MCD patients, with a significant increase compared with controls. In vitro, mouse podocyte cells incubated with serum from a MCD patient at disease onset showed increased IL-33 secretion. These results suggest an IL-33-related immune response plays a role in MCD.
Collapse
Affiliation(s)
- Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
- Department of Microbiology and Immunology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Cao Q, Wang R, Niu Z, Chen T, Azmi F, Read SA, Chen J, Lee VW, Zhou C, Julovi S, Huang Q, Wang YM, Starkey MR, Zheng G, Alexander SI, George J, Wang Y, Harris DC. Type 2 innate lymphoid cells are protective against hepatic ischaemia/reperfusion injury. JHEP Rep 2023; 5:100837. [PMID: 37691688 PMCID: PMC10482753 DOI: 10.1016/j.jhepr.2023.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND AND AIMS Although type 2 innate lymphoid cells (ILC2s) were originally found to be liver-resident lymphocytes, the role and importance of ILC2 in liver injury remains poorly understood. In the current study, we sought to determine whether ILC2 is an important regulator of hepatic ischaemia/reperfusion injury (IRI). METHODS ILC2-deficient mice (ICOS-T or NSG) and genetically modified ILC2s were used to investigate the role of ILC2s in murine hepatic IRI. Interactions between ILC2s and eosinophils or macrophages were studied in coculture. The role of human ILC2s was assessed in an immunocompromised mouse model of hepatic IRI. RESULTS Administration of IL-33 prevented hepatic IRI in association with reduction of neutrophil infiltration and inflammatory mediators in the liver. IL-33-treated mice had elevated numbers of ILC2s, eosinophils, and regulatory T cells. Eosinophils, but not regulatory T cells, were required for IL-33-mediated hepatoprotection in IRI mice. Depletion of ILC2s substantially abolished the protective effect of IL-33 in hepatic IRI, indicating that ILC2s play critical roles in IL-33-mediated liver protection. Adoptive transfer of ex vivo-expanded ILC2s improved liver function and attenuated histologic damage in mice subjected to IRI. Mechanistic studies combining genetic and adoptive transfer approaches identified a protective role of ILC2s through promoting IL-13-dependent induction of anti-inflammatory macrophages and IL-5-dependent elevation of eosinophils in IRI. Furthermore, in vivo expansion of human ILC2s by IL-33 or transfer of ex vivo-expanded human ILC2s ameliorated hepatic IRI in an immunocompromised mouse model of hepatic IRI. CONCLUSIONS This study provides insight into the mechanisms of ILC2-mediated liver protection that could serve as therapeutic targets to treat acute liver injury. IMPACT AND IMPLICATIONS We report that type 2 innate lymphoid cells (ILC2s) are important regulators in a mouse model of liver ischaemia/reperfusion injury (IRI). Through manipulation of macrophage and eosinophil phenotypes, ILC2s mitigate liver inflammation and injury during liver IRI. We propose that ILC2s have the potential to serve as a therapeutic tool for protecting against acute liver injury and lay the foundation for translation of ILC2 therapy to human liver disease.
Collapse
Affiliation(s)
- Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Ruifeng Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
- Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Zhiguo Niu
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Titi Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Farhana Azmi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Scott A. Read
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Jianwei Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Vincent W.S. Lee
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Chunze Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Sohel Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Qingsong Huang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yuan Min Wang
- Centre for Kidney Research, Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Malcolm R. Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Stephen I. Alexander
- Centre for Kidney Research, Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - David C.H. Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
27
|
Yao HC, Zhu Y, Lu HY, Ju HM, Xu SQ, Qiao Y, Wei SJ. Type 2 innate lymphoid cell-derived amphiregulin regulates type II alveolar epithelial cell transdifferentiation in a mouse model of bronchopulmonary dysplasia. Int Immunopharmacol 2023; 122:110672. [PMID: 37480752 DOI: 10.1016/j.intimp.2023.110672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication in preterm infants characterized by alveolar growth arrest. Interleukin (IL)-33 and type 2 innate lymphoid cell (ILC2) affect type II alveolar epithelial cell (AECII) differentiation in BPD mice and may cause increased lung epithelial-mesenchymal transition (EMT). Amphiregulin (AREG) can be produced by ILC2 and is associated with tissue repair. However, the action mechanism of AREG produced by ILC2 to alveolar development in BPD is unclear. In this study, we aimed to demonstrate the role and mechanism of AREG in influencing AECII transdifferentiation in the lung tissue of BPD mice. The effects of ILC2-derived AREG on AECII transdifferentiation were verified in vivo and in vitro, and the role of IL-33 on ILC2-derived AREG in AECII transdifferentiation in BPD mice and a preliminary investigation of the role of AREG's receptor-epidermal growth factor receptor (EGFR) on AECII transdifferentiation. The results showed that neonatal mice developed severe lung injury after hyperoxia, and IL-33 induced AREG production via ILC2 affected normal AECII differentiation and promoted EMT. In addition, the blockade of EGFR was found to alleviate the impaired AECII differentiation under hyperoxia in an in vitro study. In summary, our study demonstrates that AREG secreted by ILC2 affects AECII transdifferentiation in BPD mice, which provides a new idea for the clinical treatment of BPD.
Collapse
Affiliation(s)
- Hui-Ci Yao
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yue Zhu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hong-Yan Lu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Hui-Min Ju
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Su-Qing Xu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qiao
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shan-Jie Wei
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
28
|
Zhu X, Lu J, Rao J, Ru D, Gao M, Shi D, Cao K, Wen S, Dai C, Wang X, Mi W, Liu L, Zhou H. Crosstalk between Interleukin-1 Receptor-Like 1 and Transforming Growth Factor-β Receptor Signaling Promotes Renal Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1029-1045. [PMID: 37236504 DOI: 10.1016/j.ajpath.2023.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
IL-33, a member of the IL-1 family, acts as an alarmin in immune response. Epithelial-mesenchymal transition and transforming growth factor-β (TGF-β)–induced fibroblast activation are key events in the development of renal interstitial fibrosis. The current study found increased expression of IL-33 and interleukin-1 receptor-like 1 (IL1RL1, alias ST2), the receptor for IL-33, in human fibrotic renal tissues. In addition, IL-33– or ST2-deficient mice showed significantly reduced levels of fibronectin, α-smooth muscle actin, and vimentin, and increased E-cadherin levels. In HK-2 cells, IL-33 promotes the phosphorylation of the TGF-β receptor (TGF-βR), Smad2, and Smad3, and the production of extracellular matrix (ECM), with reduced expression of E-cadherin. Blocking TGF-βR signaling or suppressing ST2 expression impeded Smad2 and Smad3 phosphorylation, thereby reducing ECM production, suggesting that IL-33–induced ECM synthesis requires cooperation between the two pathways. Mechanistically, IL-33 treatment induced a proximate interaction between ST2 and TGF-βRs, activating downstream Smad2 and Smad3 for ECM production in renal epithelial cells. Collectively, this study identified a novel and essential role for IL-33 in promoting TGF-β signaling and ECM production in the development of renal fibrosis. Therefore, targeting IL-33/ST2 signaling may be an effective therapeutic strategy for renal fibrosis.
Collapse
Affiliation(s)
- Xingxing Zhu
- Department of Immunology, Nanjing Medical University, Nanjing, China.
| | - Jiahui Lu
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Jia Rao
- Department of Immunology, Anhui Medical University, Hefei, China
| | - Dongqing Ru
- Department of Immunology, Nanjing Medical University, Nanjing, China; Central Laboratory, The Second Affiliated Hospital, Henan University of Science and Technology, Luoyang, China
| | - Mengru Gao
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Kelei Cao
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Shuang Wen
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Chunsun Dai
- Department of Clinical Pathology, The Fourth Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xuerong Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, Nanjing, China; Department of Immunology, Anhui Medical University, Hefei, China.
| |
Collapse
|
29
|
Mak ML, Reid KT, Crome SQ. Protective and pathogenic functions of innate lymphoid cells in transplantation. Clin Exp Immunol 2023; 213:23-39. [PMID: 37119279 PMCID: PMC10324558 DOI: 10.1093/cei/uxad050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 05/01/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a family of lymphocytes with essential roles in tissue homeostasis and immunity. Along with other tissue-resident immune populations, distinct subsets of ILCs have important roles in either promoting or inhibiting immune tolerance in a variety of contexts, including cancer and autoimmunity. In solid organ and hematopoietic stem cell transplantation, both donor and recipient-derived ILCs could contribute to immune tolerance or rejection, yet understanding of protective or pathogenic functions are only beginning to emerge. In addition to roles in directing or regulating immune responses, ILCs interface with parenchymal cells to support tissue homeostasis and even regeneration. Whether specific ILCs are tissue-protective or enhance ischemia reperfusion injury or fibrosis is of particular interest to the field of transplantation, beyond any roles in limiting or promoting allograft rejection or graft-versus host disease. Within this review, we discuss the current understanding of ILCs functions in promoting immune tolerance and tissue repair at homeostasis and in the context of transplantation and highlight where targeting or harnessing ILCs could have applications in novel transplant therapies.
Collapse
Affiliation(s)
- Martin L Mak
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| |
Collapse
|
30
|
Zhu Y, Mi L, Lu H, Ju H, Hao X, Xu S. ILC2 regulates hyperoxia-induced lung injury via an enhanced Th17 cell response in the BPD mouse model. BMC Pulm Med 2023; 23:188. [PMID: 37254088 DOI: 10.1186/s12890-023-02474-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 05/10/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUD Recent research has focused on the role of immune cells and immune responses in the pathogenesis of bronchopulmonary dysplasia (BPD), but the exact mechanisms have not yet been elucidated. Previously, the key roles of type 2 innate lymphoid cells (ILC2) in the lung immune network of BPD were explored. Here, we investigated the role Th17 cell response in hyperoxia-induced lung injury of BPD, as well as the relationship between ILC2 and Th17 cell response. METHODS A hyperoxia-induced BPD mouse model was constructed and the pathologic changes of lung tissues were evaluated by Hematoxylin-Eosin staining. Flow cytometry analysis was conducted to determine the levels of Th17 cell, ILC2 and IL-6+ILC2. The expression levels of IL-6, IL-17 A, IL-17 F, and IL-22 in the blood serum and lung tissues of BPD mice were measured by ELISA. To further confirm the relationship between ILC2 and Th17 cell differentiation, ILC2 depletion was performed in BPD mice. Furthermore, we used immunomagnetic beads to enrich ILC2 and then flow-sorted mouse lung CD45+Lin-CD90.2+Sca-1+ILC2. The sorted ILC2s were injected into BPD mice via tail vein. Following ILC2 adoptive transfusion, the changes of Th17 cell response and lung injury were detected in BPD mice. RESULTS The expression levels of Th17 cells and Th17 cell-related cytokines, including IL-17 A, IL-17 F, and IL-22, were significantly increased in BPD mice. Concurrently, there was a significant increase in the amount of ILC2 and IL-6+ILC2 during hyperoxia-induced lung injury, which was consistent with the trend for Th17 cell response. Compared to the control BPD group, ILC2 depletion was found to partially abolish the Th17 cell response and had protective effects against lung injury after hyperoxia. Furthermore, the adoptive transfer of ILC2 enhanced the Th17 cell response and aggravated lung injury in BPD mice. CONCLUSIONS This study found that ILC2 regulates hyperoxia-induced lung injury by targeting the Th17 cell response in BPD, which shows a novel strategy for BPD immunotherapy.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Lanlan Mi
- Department of Neonatology, Shanghai Children's Medical Center, No.1678 Dongfang Road, Pudong New Area, Shanghai, 200127, China
| | - Hongyan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China.
| | - Huimin Ju
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Xiaobo Hao
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Suqing Xu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| |
Collapse
|
31
|
Jiang S, Zheng Y, Lv B, He S, Yang W, Wang B, Zhou J, Liu S, Li D, Lin J. Single-cell landscape dissecting the transcription and heterogeneity of innate lymphoid cells in ischemic heart. Front Immunol 2023; 14:1129007. [PMID: 37228603 PMCID: PMC10203554 DOI: 10.3389/fimmu.2023.1129007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Background Until now, few articles have revealed the potential roles of innate lymphoid cells (ILCs) in cardiovascular diseases. However, the infiltration of ILC subsets in ischemic myocardium, the roles of ILC subsets in myocardial infarction (MI) and myocardial ischemia-reperfusion injury (MIRI) and the related cellular and molecular mechanisms have not been described with a sufficient level of detail. Method In the current study, 8-week-old male C57BL/6J mice were divided into three groups: MI, MIRI and sham group. Single-cell sequencing technology was used to perform dimensionality reduction clustering of ILC to analyze the ILC subset landscape at a single-cell resolution, and finally flow cytometry was used to confirm the existence of the new ILC subsets in different disease groups. Results Five ILC subsets were found, including ILC1, ILC2a, ILC2b, ILCdc and ILCt. It is worth noting that ILCdc, ILC2b and ILCt were identified as new ILC subclusters in the heart. The cellular landscapes of ILCs were revealed and signal pathways were predicted. Furthermore, pseudotime trajectory analysis exhibited different ILC statuses and traced related gene expression in normal and ischemic conditions. In addition, we established a ligand-receptor-transcription factor-target gene regulatory network to disclose cell communications among ILC clusters. Moreover, we further revealed the transcriptional features of the ILCdc and ILC2a subsets. Finally, the existence of ILCdc was confirmed by flow cytometry. Conclusion Collectively, by characterizing the spectrums of ILC subclusters, our results provide a new blueprint for understanding ILC subclusters' roles in myocardial ischemia diseases and further potential treatment targets.
Collapse
Affiliation(s)
- Shijiu Jiang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, The First Affiliated Hospital, Shihezi University, Shihezi, Xinjiang, China
| | - Yuqi Zheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenling Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boyuan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shangwei Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dazhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jibin Lin
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Ott LC, Cuenca AG. Innate immune cellular therapeutics in transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1067512. [PMID: 37994308 PMCID: PMC10664839 DOI: 10.3389/frtra.2023.1067512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Successful organ transplantation provides an opportunity to extend the lives of patients with end-stage organ failure. Selectively suppressing the donor-specific alloimmune response, however, remains challenging without the continuous use of non-specific immunosuppressive medications, which have multiple adverse effects including elevated risks of infection, chronic kidney injury, cardiovascular disease, and cancer. Efforts to promote allograft tolerance have focused on manipulating the adaptive immune response, but long-term allograft survival rates remain disappointing. In recent years, the innate immune system has become an attractive therapeutic target for the prevention and treatment of transplant organ rejection. Indeed, contemporary studies demonstrate that innate immune cells participate in both the initial alloimmune response and chronic allograft rejection and undergo non-permanent functional reprogramming in a phenomenon termed "trained immunity." Several types of innate immune cells are currently under investigation as potential therapeutics in transplantation, including myeloid-derived suppressor cells, dendritic cells, regulatory macrophages, natural killer cells, and innate lymphoid cells. In this review, we discuss the features and functions of these cell types, with a focus on their role in the alloimmune response. We examine their potential application as therapeutics to prevent or treat allograft rejection, as well as challenges in their clinical translation and future directions for investigation.
Collapse
Affiliation(s)
- Leah C Ott
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| | - Alex G Cuenca
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
33
|
Razi S, Yaghmoorian Khojini J, Kargarijam F, Panahi S, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Macrophage efferocytosis in health and disease. Cell Biochem Funct 2023; 41:152-165. [PMID: 36794573 DOI: 10.1002/cbf.3780] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Creating cellular homeostasis within a defined tissue typically relates to the processes of apoptosis and efferocytosis. A great example here is cell debris that must be removed to prevent unwanted inflammatory responses and then reduce autoimmunity. In view of that, defective efferocytosis is often assumed to be responsible for the improper clearance of apoptotic cells (ACs). This predicament triggers off inflammation and even results in disease development. Any disruption of phagocytic receptors, molecules as bridging groups, or signaling routes can also inhibit macrophage efferocytosis and lead to the impaired clearance of the apoptotic body. In this line, macrophages as professional phagocytic cells take the lead in the efferocytosis process. As well, insufficiency in macrophage efferocytosis facilitates the spread of a wide variety of diseases, including neurodegenerative diseases, kidney problems, types of cancer, asthma, and the like. Establishing the functions of macrophages in this respect can be thus useful in the treatment of many diseases. Against this background, this review aimed to recapitulate the knowledge about the mechanisms related to macrophage polarization under physiological or pathological conditions, and shed light on its interaction with efferocytosis.
Collapse
Affiliation(s)
- Shokufeh Razi
- Department of Genetics, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Kargarijam
- Department of Biotechnology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran
| | - Susan Panahi
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
34
|
Nagashima R, Ishikawa H, Kuno Y, Kohda C, Iyoda M. HIF-PHD inhibitor regulates the function of group2 innate lymphoid cells and polarization of M2 macrophages. Sci Rep 2023; 13:1867. [PMID: 36725898 PMCID: PMC9892566 DOI: 10.1038/s41598-023-29161-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/31/2023] [Indexed: 02/03/2023] Open
Abstract
Hypoxia-inducible factor-prolyl hydroxylase (HIF-PHD) inhibitors are therapeutic agents for renal anemia that work through HIF2-mediated upregulation of erythropoietin (EPO) and have also been reported to suppress renal fibrosis. Group 2 innate lymphoid cells (ILC2s) have been proven to be involved in the pathogenesis of fibrosis in various organs, including the kidney. However, the relationship between the HIF pathway, renal fibrosis, and kidney ILC2s remains unclear. In the present study, we found that HIF activation by HIF-PHD inhibitors suppressed type 2 cytokine production from kidney ILC2s. The enhanced HIF pathway downregulated the IL-33 receptor ST2L on ILC2s, and phosphorylation of downstream p38 MAPK was attenuated. M2 macrophages that promote renal fibrosis were polarized by ILC2 supernatants, but reduced cytokine production from ILC2s treated with HIF-PHD inhibitors suppressed this polarization. Our findings suggest that HIF-PHD inhibitors are potential therapeutic agents for renal fibrosis that are mediated by the alteration of ILC2 function.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan.
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan.,Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan.,Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
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
|
36
|
Abstract
When discovered in the early 2000s, interleukin-33 (IL-33) was characterized as a potent driver of type 2 immunity and implicated in parasite clearance, as well as asthma, allergy, and lung fibrosis. Yet research in other models has since revealed that IL-33 is a highly pleiotropic molecule with diverse functions. These activities are supported by elusive release mechanisms and diverse expression of the IL-33 receptor, STimulation 2 (ST2), on both immune and stromal cells. Interestingly, IL-33 also supports type 1 immune responses during viral and tumor immunity and after allogeneic hematopoietic stem cell transplantation. Yet the IL-33-ST2 axis is also critical to the establishment of systemic homeostasis and tissue repair and regeneration. Despite these recent findings, the mechanisms by which IL-33 governs the balance between immunity and homeostasis or can support both effective repair and pathogenic fibrosis are poorly understood. As such, ongoing research is trying to understand the potential reparative and regulatory versus pro-inflammatory and pro-fibrotic roles for IL-33 in transplantation. This review provides an overview of the emerging regenerative role of IL-33 in organ homeostasis and tissue repair as it relates to transplantation immunology. It also outlines the known impacts of IL-33 in commonly transplanted solid organs and covers the envisioned roles for IL-33 in ischemia-reperfusion injury, rejection, and tolerance. Finally, we give a comprehensive summary of its effects on different cell populations involved in these processes, including ST2 + regulatory T cells, innate lymphoid cell type 2, as well as significant myeloid cell populations.
Collapse
|
37
|
Wang Z, Li W, Gou L, Zhou Y, Peng G, Zhang J, Liu J, Li R, Ni H, Zhang W, Cao T, Cao Q, Su H, Han YP, Tong N, Fu X, Ilegems E, Lu Y, Berggren PO, Zheng X, Wang C. Biodegradable and Antioxidant DNA Hydrogel as a Cytokine Delivery System for Diabetic Wound Healing. Adv Healthc Mater 2022; 11:e2200782. [PMID: 36101484 DOI: 10.1002/adhm.202200782] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/30/2022] [Indexed: 01/28/2023]
Abstract
Impaired diabetic wound healing is associated with the persistence of chronic inflammation and excessive oxidative stress, which has become one of the most serious clinical challenges. Wound dressings with anti-inflammatory and reactive oxygen species (ROS)-scavenging properties are desirable for diabetic wound treatment. In this study, a shape-adaptable, biodegradable, biocompatible, antioxidant, and immunomodulatory interleukin-33 (IL-33)-cytogel is developed by encapsulating IL-33 into physically cross-linked DNA hydrogels and used as wound dressings to promote diabetic wound healing. The porous microstructures and biodegradable properties of the IL-33-cytogel ensure the local sustained-release of IL-33 in the wound area, where the sustained-release of IL-33 is maintained for at least 7 days. IL-33-cytogel can induce local accumulation of group 2 innate lymphoid cells (ILC2s) and regulatory T cells (Tregs), as well as M1-to-M2 transition at the wound sites. Additionally, the antioxidant and biocompatible characteristics of DNA hydrogels promote the scavenging of intracellular ROS without affecting cell viability. As a result, local inflammation in the diabetic wound area is resolved upon IL-33-cytogel treatment, which is accompanied by improved granulation tissue regeneration and accelerated wound closure. This study demonstrates a promising strategy in tissue engineering and regenerative medicine by incorporating DNA hydrogels and cytokine immunotherapy for promoting diabetic wound healing.
Collapse
Affiliation(s)
- Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, SE-17176, Sweden
| | - Wei Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liping Gou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ge Peng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiayi Zhang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaye Liu
- Department of thyroid and parathyroid surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruoqing Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Hengfan Ni
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wanli Zhang
- Core facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Cao
- Laboratory of Infectious Diseases and Vaccine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia
| | - Hong Su
- Department of Dermatology, Chengdu First People's Hospital, Chengdu, 610041, China
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, The College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Erwin Ilegems
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, SE-17176, Sweden
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Per-Olof Berggren
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, SE-17176, Sweden
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengshi Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
38
|
Di Carmine S, Scott MM, McLean MH, McSorley HJ. The role of interleukin-33 in organ fibrosis. DISCOVERY IMMUNOLOGY 2022; 1:kyac006. [PMID: 38566909 PMCID: PMC10917208 DOI: 10.1093/discim/kyac006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/06/2022] [Accepted: 09/22/2022] [Indexed: 04/04/2024]
Abstract
Interleukin (IL)-33 is highly expressed in the nucleus of cells present at barrier sites and signals via the ST2 receptor. IL-33 signalling via ST2 is essential for return to tissue homeostasis after acute inflammation, promoting fibrinogenesis and wound healing at injury sites. However, this wound-healing response becomes aberrant during chronic or sustained inflammation, leading to transforming growth factor beta (TGF-β) release, excessive extracellular matrix deposition, and fibrosis. This review addresses the role of the IL-33 pathway in fibrotic diseases of the lung, liver, gastrointestinal tract, skin, kidney and heart. In the lung and liver, IL-33 release leads to the activation of pro-fibrotic TGF-β, and in these sites, IL-33 has clear pro-fibrotic roles. In the gastrointestinal tract, skin, and kidney, the role of IL-33 is more complex, being both pro-fibrotic and tissue protective. Finally, in the heart, IL-33 serves cardioprotective functions by favouring tissue healing and preventing cardiomyocyte death. Altogether, this review indicates the presence of an unclear and delicate balance between resolving and pro-fibrotic capabilities of IL-33, which has a central role in the modulation of type 2 inflammation and fibrosis in response to tissue injury.
Collapse
Affiliation(s)
- Samuele Di Carmine
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, UK
| | - Molly M Scott
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Mairi H McLean
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, University of Dundee, Dundee, UK
| |
Collapse
|
39
|
Yi XM, Lian H, Li S. Signaling and functions of interleukin-33 in immune regulation and diseases. CELL INSIGHT 2022; 1:100042. [PMID: 37192860 PMCID: PMC10120307 DOI: 10.1016/j.cellin.2022.100042] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 05/18/2023]
Abstract
Interleukin-33 (IL-33) which belongs to the interleukin-1 (IL-1) family is an alarmin cytokine with critical roles in tissue homeostasis, pathogenic infection, inflammation, allergy and type 2 immunity. IL-33 transmits signals through its receptor IL-33R (also called ST2) which is expressed on the surface of T helper 2 (Th2) cells and group 2 innate lymphoid cells (ILC2s), thus inducing transcription of Th2-associated cytokine genes and host defense against pathogens. Moreover, the IL-33/IL-33R axis is also involved in development of multiple types of immune-related diseases. In this review, we focus on current progress on IL-33-trigggered signaling events, the important functions of IL-33/IL-33R axis in health and diseases as well as the promising therapeutic implications of these findings.
Collapse
Affiliation(s)
- Xue-Mei Yi
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huan Lian
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Shu Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
40
|
Zhang HM, Chen XJ, Li SP, Zhang JM, Sun J, Zhou LX, Zhou GP, Cui B, Sun LY, Zhu ZJ. ILC2s expanded by exogenous IL-33 regulate CD45+CD11b+F4/80high macrophage polarization to alleviate hepatic ischemia-reperfusion injury. Front Immunol 2022; 13:869365. [PMID: 35967407 PMCID: PMC9372719 DOI: 10.3389/fimmu.2022.869365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatic ischemia-reperfusion injury (IRI) is an adverse consequence of hepatectomy or liver transplantation. Recently, immune mechanisms involved in hepatic IRI have attracted increased attention of investigators working in this area. In specific, group 2 innate lymphoid cells (ILC2s), have been strongly implicated in mediating type 2 inflammation. However, their immune mechanisms as involved with hepatic IRI remain unclear. Here, we reported that the population of ILC2s is increased with the development of hepatic IRI as shown in a mouse model in initial stage. Moreover, M2 type CD45+CD11b+F4/80high macrophages increased and reached maximal levels at 24 h followed by a significant elevation in IL-4 levels. We injected exogenous IL-33 into the tail vein of mice as a mean to stimulate ILC2s production. This stimulation of ILC2s resulted in a protective effect upon hepatic IRI along with an increase in M2 type CD45+CD11b+F4/80high macrophages. In contrast, depletion of ILC2s as achieved with use of an anti-CD90.2 antibody substantially abolished this protective effect of exogenous IL-33 and M2 type CD45+CD11b+F4/80high macrophage polarization in hepatic IRI. Therefore, this exogenous IL-33 induced potentiation of ILC2s appears to regulate the polarization of CD45+CD11b+F4/80high macrophages to alleviate IRI. Such findings provide the foundation for the development of new targets and strategies in the treatment of hepatic IRI.
Collapse
Affiliation(s)
- Hai-Ming Zhang
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Xiao-Jie Chen
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Shi-Peng Li
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Jin-Ming Zhang
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Jie Sun
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Liu-Xin Zhou
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Guang-Peng Zhou
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Bin Cui
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
| | - Li-Ying Sun
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
- Department of Critical Liver Disease, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Li-Ying Sun, ; Zhi-Jun Zhu,
| | - Zhi-Jun Zhu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Pediatric Liver Transplantation of Capital Medical University, Beijing, China
- *Correspondence: Li-Ying Sun, ; Zhi-Jun Zhu,
| |
Collapse
|
41
|
Nagashima R, Ishikawa H, Kuno Y, Kohda C, Iyoda M. IL-33 attenuates renal fibrosis via group2 innate lymphoid cells. Cytokine 2022; 157:155963. [PMID: 35868116 DOI: 10.1016/j.cyto.2022.155963] [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: 03/28/2022] [Revised: 06/20/2022] [Accepted: 07/04/2022] [Indexed: 11/03/2022]
Abstract
Renal fibrosis is a common pathway in the progression of various kidney diseases and injuries. Unilateral ureteral obstruction (UUO) induces renal fibrosis, and immune responses profoundly affect its pathogenesis. Group2 innate lymphoid cells (ILC2s) are strongly activated by interleukin (IL) -33, which is a member of IL-1 family and recognize as alarmin. ILC2s quickly produce large amounts of type 2 cytokines including IL-5 and IL-13, which are involved in inflammation, tissue homeostasis, and wound healing. However, the relationship between renal fibrosis and ILC2s has been unclear. In the present study, we investigated the roles of the ILC2/L-33 axis in renal fibrosis using a UUO model. We found that kidney ILC2s decreased in UUO-affected kidneys compared with their counterpart kidneys despite IL-33 upregulation. There was no effect of reactive oxygen species or TGF-β from reduced ILC2 caused by UUO. Pretreatment with IL-33 before UUO induced ILC2s and Tregs in kidneys and alleviated renal fibrosis. Furthermore, this protective effect was maintained even when CD4+T cells was depleted. These findings demonstrated that ILC2s play a predominant role in the suppressive function of renal fibrosis mediated by pretreatment with IL-33. In contrast, post-treatment with IL-33 after UUO increased ILC2s in kidneys but had no therapeutic effect on renal fibrosis. Our findings suggest that ILC2s have potential roles in the prevention of renal fibrosis and can serve as a therapeutic and diagnostic target.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
miR-29a-3p in Exosomes from Heme Oxygenase-1 Modified Bone Marrow Mesenchymal Stem Cells Alleviates Steatotic Liver Ischemia-Reperfusion Injury in Rats by Suppressing Ferroptosis via Iron Responsive Element Binding Protein 2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6520789. [PMID: 35720183 PMCID: PMC9203237 DOI: 10.1155/2022/6520789] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/03/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
Hepatic ischemia-reperfusion injury (IRI) is an inevitable result of liver surgery. Steatotic livers are extremely sensitive to IRI and have worse tolerance. Ferroptosis is considered to be one of the main factors of organ IRI. This study is aimed at exploring the role of ferroptosis in the effect of heme oxygenase-1-modified bone marrow mesenchymal stem cells (HO-1/BMMSCs) on steatotic liver IRI and its mechanism. An IRI model of a steatotic liver and a hypoxia reoxygenation (HR) model of steatotic hepatocytes (SHPs) were established. Rat BMMSCs were extracted and transfected with the Ho1 gene to establish HO-1/BMMSCs, and their exosomes were extracted by ultracentrifugation. Ireb2 was knocked down to verify its role in ferroptosis and cell injury in SHP-HR. Public database screening combined with quantitative real-time reverse transcription PCR identified microRNAs (miRNAs) targeting Ireb2 in HO-1/BMMSCs exosomes. miR-29a-3p mimic and inhibitor were used for functional verification experiments. Liver function, histopathology, terminal deoxynulceotidyl transferase nick-end-labeling staining, cell viability, mitochondrial membrane potential, and cell death were measured to evaluate liver tissue and hepatocyte injury. Ferroptosis was assessed by detecting the levels of IREB2, Fe2+, malondialdehyde, glutathione, lipid reactive oxygen species, glutathione peroxidase 4, prostaglandin-endoperoxide synthase 2 mRNA, and mitochondrial morphology. The results revealed that HO-1/BMMSCs improved liver tissue and hepatocyte injury and suppressed ferroptosis in vivo and in vitro. The expression of IREB2 was increased in steatotic liver IRI and SHP-HR. Knocking down Ireb2 reduced the level of Fe2+ and inhibited ferroptosis. HO-1/BMMSC exosomes reduced the expression of IREB2 and inhibited ferroptosis and cell damage. Furthermore, we confirmed high levels of miR-29a-3p in HO-1/BMMSCs exosomes. Overexpression of miR-29a-3p downregulated the expression of Ireb2 and inhibited ferroptosis. Downregulation of miR-29a-3p blocked the protective effect of HO-1/BMMSC exosomes on SHP-HR cell injury. In conclusion, ferroptosis plays an important role in HO-1/BMMSC-mediated alleviation of steatotic liver IRI. HO-1/BMMSCs could suppress ferroptosis by targeting Ireb2 via the exosomal transfer of miR-29a-3p.
Collapse
|
43
|
Wang R, Zhang J, Li D, Liu G, Fu Y, Li Q, Zhang L, Qian L, Hao L, Wang Y, Harris DCH, Wang D, Cao Q. Imbalance of circulating innate lymphoid cell subpopulations in patients with chronic kidney disease. Clin Immunol 2022; 239:109029. [PMID: 35525476 DOI: 10.1016/j.clim.2022.109029] [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: 07/22/2021] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022]
Abstract
Innate lymphoid cells (ILCs) are a newly identified heterogeneous family of innate immune cells. We conducted this study to investigate the frequency of circulating ILC subsets in various chronic kidney diseases (CKD). In DN, the proportion of total ILCs and certain ILC subgroups increased significantly. Positive correlations between proportion of total ILCs, ILC1s and body mass index, glycated hemoglobin were observed in DN. In LN, a significantly increased proportion of ILC1s was found in parallel with a reduced proportion of ILC2s. The proportions of total ILCs and ILC1s were correlated with WBC count and the level of C3. In all enrolled patients, the proportion of total ILCs and ILC1s was significantly correlated with the levels of ACR and GFR. In the present study, the proportion of circulating ILC subsets increased significantly in various types of CKD and correlated with clinico-pathological features, which suggests a possible role for ILCs in CKD.
Collapse
Affiliation(s)
- Ruifeng Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia; Department of Nephrology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dandan Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guiling Liu
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuqin Fu
- Department of Nephrology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qing Li
- The Central Laboratory of Medical Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lei Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Long Qian
- Department of Rheumatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Hao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - David C H Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
44
|
Wang YM, Shaw K, Zhang GY, Chung EY, Hu M, Cao Q, Wang Y, Zheng G, Wu H, Chadban SJ, McCarthy HJ, Harris DC, Mackay F, Grey ST, Alexander SI. Interleukin-33 Exacerbates IgA Glomerulonephritis in Transgenic Mice Overexpressing B Cell Activating Factor. J Am Soc Nephrol 2022; 33:966-984. [PMID: 35387873 PMCID: PMC9063894 DOI: 10.1681/asn.2021081145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 02/06/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The cytokine IL-33 is an activator of innate lymphoid cells 2 (ILC2s) in innate immunity and allergic inflammation. B cell activating factor (BAFF) plays a central role in B cell proliferation and differentiation, and high levels of this protein cause excess antibody production, including IgA. BAFF-transgenic mice overexpress BAFF and spontaneously develop glomerulonephritis that resembles human IgA nephropathy. METHODS We administered IL-33 or PBS to wild-type and BAFF-transgenic mice. After treating Rag1-deficient mice with IL-33, with or without anti-CD90.2 to preferentially deplete ILC2s, we isolated splenocytes, which were adoptively transferred into BAFF-transgenic mice. RESULTS BAFF-transgenic mice treated with IL-33 developed more severe kidney dysfunction and proteinuria, glomerular sclerosis, tubulointerstitial damage, and glomerular deposition of IgA and C3. Compared with wild-type mice, BAFF-transgenic mice exhibited increases of CD19+ B cells in spleen and kidney and ILC2s in kidney and intestine, which were further increased by administration of IL-33. Administering IL-33 to wild-type mice had no effect on kidney function or histology, nor did it alter the number of ILC2s in spleen, kidney, or intestine. To understand the role of ILC2s, splenocytes were transferred from IL-33-treated Rag1-deficient mice into BAFF-transgenic mice. Glomerulonephritis and IgA deposition were exacerbated by transfer of IL-33-stimulated Rag1-deficient splenocytes, but not by ILC2 (anti-CD90.2)-depleted splenocytes. Wild-type mice infused with IL-33-treated Rag1-deficient splenocytes showed no change in kidney function or ILC2 numbers or distribution. CONCLUSIONS IL-33-expanded ILC2s exacerbated IgA glomerulonephritis in a mouse model. These findings indicate that IL-33 and ILC2s warrant evaluation as possible mediators of human IgA nephropathy.
Collapse
Affiliation(s)
- Yuan Min Wang
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Karli Shaw
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Geoff Yu Zhang
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Edmund Y.M. Chung
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Min Hu
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Qi Cao
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Huiling Wu
- Kidney Node Laboratory, The Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Steven J. Chadban
- Kidney Node Laboratory, The Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Hugh J. McCarthy
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - David C.H. Harris
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Fabienne Mackay
- QIMR, University of Queensland, Brisbane, Queensland, Australia
| | - Shane T. Grey
- Transplantation Immunology Group, Garvan Institute of Medical Research, Sydney, Australia
| | - Stephen I. Alexander
- Centre for Kidney Research, The Children’s Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
45
|
Melderis S, Warkotsch MT, Dang J, Hagenstein J, Ehnold LI, Herrnstadt GR, Niehus CB, Feindt FC, Kylies D, Puelles VG, Berasain C, Avila MA, Neumann K, Tiegs G, Huber TB, Tharaux PL, Steinmetz OM. The Amphiregulin/EGFR axis protects from lupus nephritis via downregulation of pathogenic CD4 + T helper cell responses. J Autoimmun 2022; 129:102829. [PMID: 35468361 DOI: 10.1016/j.jaut.2022.102829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disorder with a complex and poorly understood immuno-pathogenesis. Lupus nephritis (LN) is a frequent and difficult to treat complication, which causes high morbidity and mortality. The multifunctional cytokine amphiregulin (AREG) has been implicated in SLE pathogenesis, but its function in LN currently remains unknown. We thus studied the model of pristane-induced LN and found increasing renal and systemic AREG expression during the course of disease. Importantly, renal injury was significantly aggravated in the absence of AREG, revealing a net anti-inflammatory role. Analyses of immune responses showed dual effects. On the one hand, AREG enhanced activation of pro-inflammatory myeloid cells, which however did not play a major role for the course of LN. More importantly, on the other hand, AREG strongly suppressed pathogenic cytokine production by T helper effector cells. This effect was more general in nature and could be reproduced in response to antigen immunization. Since AREG has been postulated to downregulate T cell responses via enhancing Treg suppressive capacity, we followed up on this aspect. Interestingly, however, in vitro studies revealed potential direct and Treg independent effects of AREG on T helper effector cells. In favor of this notion, we found significantly enhanced T cell responses and consecutive aggravation of LN, only if epidermal growth factor receptor (EGFR) signaling was abrogated in total T cells, but not if the EGFR was absent on Tregs alone. Finally, we also found enhanced AREG expression in plasma and renal biopsies of patients with LN, supporting the relevance of our findings for human disease. In summary, our data identify AREG as an anti-inflammatory mediator of LN via broad downregulation of pathogenic T cell immunity. These findings further highlight the AREG/EGFR axis as a potential therapeutic target.
Collapse
Affiliation(s)
- Simon Melderis
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias T Warkotsch
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julien Dang
- Paris Cardiovascular Research Center, Inserm, Université Paris Cité, Paris, France
| | - Julia Hagenstein
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura-Isabell Ehnold
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg R Herrnstadt
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph B Niehus
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederic C Feindt
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Kylies
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carmen Berasain
- Program of Hepatology, CIMA, University of Navarra, CIBERehd and IdiSNA, Pamplona, Spain
| | - Matias A Avila
- Program of Hepatology, CIMA, University of Navarra, CIBERehd and IdiSNA, Pamplona, Spain
| | - Katrin Neumann
- Institut für Experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Institut für Experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pierre-Louis Tharaux
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Paris Cardiovascular Research Center, Inserm, Université Paris Cité, Paris, France
| | - Oliver M Steinmetz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
46
|
Cutaneous Wound Healing: A Review about Innate Immune Response and Current Therapeutic Applications. Mediators Inflamm 2022; 2022:5344085. [PMID: 35509434 PMCID: PMC9061066 DOI: 10.1155/2022/5344085] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/22/2021] [Accepted: 03/25/2022] [Indexed: 12/22/2022] Open
Abstract
Skin wounds and compromised wound healing are major concerns for the public. Although skin wound healing has been studied for decades, the molecular and cellular mechanisms behind the process are still not completely clear. The systemic responses to trauma involve the body’s inflammatory and immunomodulatory cellular and humoral networks. Studies over the years provided essential insights into a complex and dynamic immunity during the cutaneous wound healing process. This review will focus on innate cell populations involved in the initial phase of this orchestrated process, including innate cells from both the skin and the immune system.
Collapse
|
47
|
Wu L, Tian X, Zuo H, Zheng W, Li X, Yuan M, Tian X, Song H. miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J Nanobiotechnology 2022; 20:196. [PMID: 35459211 PMCID: PMC9026664 DOI: 10.1186/s12951-022-01407-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/30/2022] [Indexed: 02/08/2023] Open
Abstract
Background Steatotic livers tolerate ischemia–reperfusion injury (IRI) poorly, increasing the risk of organ dysfunction. Ferroptosis is considered the initiating factor of organ IRI. Heme oxygenase oxygen-1 (HO-1)-modified bone marrow mesenchymal stem cells (BMMSCs) (HO-1/BMMSCs) can reduce hepatic IRI; however, the role of ferroptosis in IRI of steatotic grafts and the effect of HO-1/BMMSCs-derived exosomes (HM-exos) on ferroptosis remain unknown. Methods A model of rat liver transplantation (LT) with a severe steatotic donor liver and a model of hypoxia and reoxygenation (H/R) of steatotic hepatocytes were established. Exosomes were obtained by differential centrifugation, and the differentially expressed genes (DEGs) in liver after HM-exo treatment were detected using RNA sequencing. The expression of ferroptosis markers was analyzed. microRNA (miRNA) sequencing was used to analyze the miRNA profiles in HM-exos. Results We verified the effect of a candidate miRNA on ferroptosis of H/R treated hepatocytes, and observed the effect of exosomes knockout of the candidate miRNA on hepatocytes ferroptosis. In vitro, HM-exo treatment reduced the IRI in steatotic grafts, and enrichment analysis of DEGs suggested that HM-exos were involved in the regulation of the ferroptosis pathway. In vitro, inhibition of ferroptosis by HM-exos reduced hepatocyte injury. HM-exos contained more abundant miR-124-3p, which reduced ferroptosis of H/R-treated cells by inhibiting prostate six transmembrane epithelial antigen 3 (STEAP3), while overexpression of Steap3 reversed the effect of mir-124-3p. In addition, HM-exos from cell knocked out for miR-124-3p showed a weakened inhibitory effect on ferroptosis. Similarly, HM-exo treatment increased the content of miR-124-3p in grafts, while decreasing the level of STEAP3 and reducing the degree of hepatic ferroptosis. Conclusion Ferroptosis is involved in the IRI during LT with a severe steatotic donor liver. miR-124-3p in HM-exos downregulates Steap3 expression to inhibit ferroptosis, thereby attenuating graft IRI, which might be a promising strategy to treat IRI in steatotic grafts. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01407-8.
Collapse
Affiliation(s)
- Longlong Wu
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xuan Tian
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.,NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, People's Republic of China
| | - Xiang Li
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Mengshu Yuan
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xiaorong Tian
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China. .,Tianjin Key Laboratory of Organ Transplantation, Tianjin, People's Republic of China.
| |
Collapse
|
48
|
Nucleic Acid Nanotechnology for Diagnostics and Therapeutics in Acute Kidney Injury. Int J Mol Sci 2022; 23:ijms23063093. [PMID: 35328515 PMCID: PMC8953740 DOI: 10.3390/ijms23063093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
Acute kidney injury (AKI) has impacted a heavy burden on global healthcare system with a high morbidity and mortality in both hospitalized and critically ill patients. However, there are still some shortcomings in clinical approaches for the disease to date, appealing for an earlier recognition and specific intervention to improve long-term outcomes. In the past decades, owing to the predictable base-pairing rule and highly modifiable characteristics, nucleic acids have already become significant biomaterials for nanostructure and nanodevice fabrication, which is known as nucleic acid nanotechnology. In particular, its excellent programmability and biocompatibility have further promoted its intersection with medical challenges. Lately, there have been an influx of research connecting nucleic acid nanotechnology with the clinical needs for renal diseases, especially AKI. In this review, we begin with the diagnostics of AKI based on nucleic acid nanotechnology with a highlight on aptamer- and probe-functionalized detection. Then, recently developed nanoscale nucleic acid therapeutics towards AKI will be fully elucidated. Furthermore, the strengths and limitations will be summarized, envisioning a wiser and wider application of nucleic acid nanotechnology in the future of AKI.
Collapse
|
49
|
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
|
50
|
Huang J, Fu L, Huang J, Zhao J, Zhang X, Wang W, Liu Y, Sun B, Qiu J, Hu X, Liu Z, Guo X. Group 3 Innate Lymphoid Cells Protect the Host from the Uropathogenic Escherichia coli Infection in the Bladder. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103303. [PMID: 35018740 PMCID: PMC8867143 DOI: 10.1002/advs.202103303] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/01/2021] [Indexed: 05/09/2023]
Abstract
Innate lymphoid cells (ILCs) are crucial in orchestrating immunity and maintaining tissue homeostasis in various barrier tissues, but whether ILCs influence immune responses in the urinary tract remains poorly understood. Here, bladder-resident ILCs are comprehensively explored and identified their unique phenotypic and developmental characteristics. Notably, bladder-resident ILCs rapidly respond to uropathogenic Escherichia coli (UPEC) infection. It is found that ILC3 is necessary for early protection against UPEC infection in the bladder. Mechanistically, UPEC infection leads to interleukin (IL)-1β production in the bladder via a MyD88-dependent pathway, which promotes ILC3 activation. ILC3-expressed IL-17A further recruits neutrophils and controls UPEC infection in the bladder. Together, these results demonstrate a critical role for bladder ILCs in the host defense against UPEC infection.
Collapse
Affiliation(s)
- Jiaoyan Huang
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Liuhui Fu
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Jida Huang
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Jie Zhao
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Xin Zhang
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Wenyan Wang
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Yeyang Liu
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Bowen Sun
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthChinese Academy of SciencesShanghai200031China
| | - Xiaoyu Hu
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| | - Zhihua Liu
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
| | - Xiaohuan Guo
- Institute for ImmunologyTsinghua UniversityBeijing100084China
- Department of Basic Medical SciencesSchool of MedicineTsinghua UniversityBeijing100084China
- Beijing Key Lab for Immunological Research on Chronic DiseasesTsinghua UniversityBeijing100084China
| |
Collapse
|