1
|
Yang H, Li J, Huang XR, Bucala R, Xu A, Lan HY. Macrophage-derived macrophage migration inhibitory factor mediates renal injury in anti-glomerular basement membrane glomerulonephritis. Front Immunol 2024; 15:1361343. [PMID: 38846956 PMCID: PMC11153660 DOI: 10.3389/fimmu.2024.1361343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophages are a rich source of macrophage migration inhibitory factor (MIF). It is well established that macrophages and MIF play a pathogenic role in anti-glomerular basement membrane crescentic glomerulonephritis (anti-GBM CGN). However, whether macrophages mediate anti-GBM CGN via MIF-dependent mechanism remains unexplored, which was investigated in this study by specifically deleting MIF from macrophages in MIFf/f-lysM-cre mice. We found that compared to anti-GBM CGN induced in MIFf/f control mice, conditional ablation of MIF in macrophages significantly suppressed anti-GBM CGN by inhibiting glomerular crescent formation and reducing serum creatinine and proteinuria while improving creatine clearance. Mechanistically, selective MIF depletion in macrophages largely inhibited renal macrophage and T cell recruitment, promoted the polarization of macrophage from M1 towards M2 via the CD74/NF-κB/p38MAPK-dependent mechanism. Unexpectedly, selective depletion of macrophage MIF also significantly promoted Treg while inhibiting Th1 and Th17 immune responses. In summary, MIF produced by macrophages plays a pathogenic role in anti-GBM CGN. Targeting macrophage-derived MIF may represent a novel and promising therapeutic approach for the treatment of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Hui Yang
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen University, SunYat‐sen University, Shenzhen, China
| | - Xiao-ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anping Xu
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Pan J, Ye F, Li H, Yu C, Mao J, Xiao Y, Chen H, Wu J, Li J, Fei L, Wu Y, Meng X, Guo G, Wang Y. Dissecting the immune discrepancies in mouse liver allograft tolerance and heart/kidney allograft rejection. Cell Prolif 2024; 57:e13555. [PMID: 37748771 PMCID: PMC10905343 DOI: 10.1111/cpr.13555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/23/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023] Open
Abstract
The liver is the most tolerogenic of transplanted organs. However, the mechanisms underlying liver transplant tolerance are not well understood. The comparison between liver transplantation tolerance and heart/kidney transplantation rejection will deepen our understanding of tolerance and rejection in solid organs. Here, we built a mouse model of liver, heart and kidney allograft and performed single-cell RNA sequencing of 66,393 cells to describe the cell composition and immune cell interactions at the early stage of tolerance or rejection. We also performed bulk RNA-seq of mouse liver allografts from Day 7 to Day 60 post-transplantation to map the dynamic transcriptional variation in spontaneous tolerance. The transcriptome of lymphocytes and myeloid cells were characterized and compared in three types of organ allografts. Cell-cell interaction networks reveal the coordinated function of Kupffer cells, macrophages and their associated metabolic processes, including insulin receptor signalling and oxidative phosphorylation in tolerance induction. Cd11b+ dendritic cells (DCs) in liver allografts were found to inhibit cytotoxic T cells by secreting anti-inflammatory cytokines such as Il10. In summary, we profiled single-cell transcriptome analysis of mouse solid organ allografts. We characterized the immune microenvironment of mouse organ allografts in the acute rejection state (heart, kidney) and tolerance state (liver).
Collapse
Affiliation(s)
- Jun Pan
- Department of Thyroid Surgery, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Fang Ye
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hui Li
- Key Laboratory of Combined Multiorgan Transplantation, Ministry of Public Health, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Chengxuan Yu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiajia Mao
- Kidney Disease Center, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Junqing Wu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiaqi Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yijun Wu
- Department of Thyroid Surgery, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of PharmacyAnhui Medical University, The Key Laboratory of Anti‐inflammatory of Immune Medicines, Ministry of EducationHefeiChina
| | - Guoji Guo
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative MedicineDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineHangzhouZhejiangChina
| | - Yingying Wang
- Kidney Disease Center, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
3
|
Short S, Lewik G, Issa F. An Immune Atlas of T Cells in Transplant Rejection: Pathways and Therapeutic Opportunities. Transplantation 2023; 107:2341-2352. [PMID: 37026708 PMCID: PMC10593150 DOI: 10.1097/tp.0000000000004572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/10/2023] [Accepted: 01/28/2023] [Indexed: 04/08/2023]
Abstract
Short-term outcomes in allotransplantation are excellent due to technical and pharmacological advances; however, improvement in long-term outcomes has been limited. Recurrent episodes of acute cellular rejection, a primarily T cell-mediated response to transplanted tissue, have been implicated in the development of chronic allograft dysfunction and loss. Although it is well established that acute cellular rejection is primarily a CD4 + and CD8 + T cell mediated response, significant heterogeneity exists within these cell compartments. During immune responses, naïve CD4 + T cells are activated and subsequently differentiate into specific T helper subsets under the influence of the local cytokine milieu. These subsets have distinct phenotypic and functional characteristics, with reported differences in their contribution to rejection responses specifically. Of particular relevance are the regulatory subsets and their potential to promote tolerance of allografts. Unraveling the specific contributions of these cell subsets in the context of transplantation is complex, but may reveal new avenues of therapeutic intervention for the prevention of rejection.
Collapse
Affiliation(s)
- Sarah Short
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Guido Lewik
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, United Kingdom
| |
Collapse
|
4
|
Harris R, Karimi M. Dissecting the regulatory network of transcription factors in T cell phenotype/functioning during GVHD and GVT. Front Immunol 2023; 14:1194984. [PMID: 37441063 PMCID: PMC10333690 DOI: 10.3389/fimmu.2023.1194984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Transcription factors play a major role in regulation and orchestration of immune responses. The immunological context of the response can alter the regulatory networks required for proper functioning. While these networks have been well-studied in canonical immune contexts like infection, the transcription factor landscape during alloactivation remains unclear. This review addresses how transcription factors contribute to the functioning of mature alloactivated T cells. This review will also examine how these factors form a regulatory network to control alloresponses, with a focus specifically on those factors expressed by and controlling activity of T cells of the various subsets involved in graft-versus-host disease (GVHD) and graft-versus-tumor (GVT) responses.
Collapse
Affiliation(s)
- Rebecca Harris
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Mobin Karimi
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
5
|
Wang Z, Jiang S, Li S, Yu W, Chen J, Yu D, Zhao C, Li Y, Kang K, Wang R, Liang M, Xu M, Ou Y, Li P, Leng X, Tian J, R-Porter T. Targeted galectin-7 inhibition with ultrasound microbubble targeted gene therapy as a sole therapy to prevent acute rejection following heart transplantation in a Rodent model. Biomaterials 2020; 263:120366. [PMID: 32950914 DOI: 10.1016/j.biomaterials.2020.120366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Despite significant advances in transplantation, acute cellular rejection (AR) remains a major obstacle that is most prevalent in the first months post heart transplantation (HT). Current treatments require high doses of immunosuppressive drugs followed by maintenance therapies that have systemic side effects including early infection. In this study, we attempted to prevent AR with a myocardial-targeted galectin-7-siRNA delivery method using cationic microbubbles (CMBs) combined with ultrasound targeted microbubble destruction (UTMD) to create local immunosuppression in a rat abdominal heterotopic heart transplantation acute rejection model. METHODS AND RESULTS Galectin-7-siRNA (siGal-7) bound to CMBs were synthesized and effective ultrasound-targeted delivery of siGal-7 into target cells confirmed in vitro. Based on these observations, three transplant rat models were tested:①isograft (ISO); ② Allograft (ALLO) +UTMD; and ③ALLO + PBS. UTMD treatments were administered at 1, 3, 5, 7 days after HT. Galectin 7 expression was reduced by 50% compared to ALLO + PBS (p < 0.005), and this was associated with significant reductions in both galectin 7 and Interleukin-2 protein levels (p < 0.001). The ALLO + UTMD group had Grade II or less inflammatory infiltration and myocyte damage in 11/12 rats using International Society For Heart and Lung Transplantation grading, compared to 0/12 rats with this grading in the ALLO + PBS group at 10 days post HT (p < 0.001). CONCLUSIONS Ultrasound-targeted galectin-7-siRNA knockdown with UTMD can prevent acute cellular rejection in the early period after allograft heart transplantation without the need for systemic immunosuppression. KEY WORDS Microbubble, Acute Rejection, Heart Transplantation, Galectin-7, RNA.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, 150086, China
| | - Shuangquan Jiang
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Shouqiang Li
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, 150086, China
| | - Weidong Yu
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jianfeng Chen
- Laboratory Animal Center, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Dandan Yu
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chen Zhao
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yingjie Li
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Kai Kang
- Department of Cardiac Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ranran Wang
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Mengmeng Liang
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Mingyuan Xu
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yanmei Ou
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Piyu Li
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xiaoping Leng
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, 150086, China.
| | - Jiawei Tian
- Department of Ultrasound Imaging, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, 150086, China.
| | - Thomas R-Porter
- Department of Cardiology, University of Nebraska Medical Center, Omaha, NE, NE 68198, USA
| |
Collapse
|
6
|
Zhu C, Xiang W, Li B, Wang Y, Feng S, Wang C, Chen Y, Xie W, Qu L, Huang H, Annunziata F, Nunna S, Krepelova A, Mohammad M. Rasa S, Neri F, Chen J, Jiang H. DNA methylation modulates allograft survival and acute rejection after renal transplantation by regulating the mTOR pathway. Am J Transplant 2020. [PMCID: PMC7891393 DOI: 10.1111/ajt.16183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
7
|
Li Z, Nie L, Chen L, Sun Y, Guo L. [Rapamycin alleviates inflammation by up-regulating TGF-β/Smad signaling in a mouse model of autoimmune encephalomyelitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:35-42. [PMID: 30692064 DOI: 10.12122/j.issn.1673-4254.2019.01.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the efficacy of rapmycin for treatment of experimental autoimmune encephalomyelitis (EAE) in mice and explore the underlying mechanism. METHODS An EAE model was established in C57BL/6 mice. After immunization, the mice were divided into model group and rapamycin groups treated daily with low-dose (0.3 mg/kg) or high-dose (1 mg/kg) rapamycin. The clinical scores of the mice were observed using Knoz score, the infiltration of IL-17 cells in the central nervous system (CNS) was determined using immunohistochemistry; the differentiation of peripheral Treg cells was analyzed using flow cytometry, and the changes in the levels of cytokines were detected with ELISA; the changes in the expressions of p-Smad2 and p- smad3 were investigated using Western blotting. RESULTS High-dose rapamycin significantly improved the neurological deficits scores of EAE mice. In high-dose rapamycin group, the scores in the onset stage, peak stage and remission stage were 0.14±0.38, 0.43±1.13 and 0.14±0.37, respectively, as compared with 1.14±0.69, 2.14±1.06 and 2.2±0.75 in the model group. The infiltration of inflammatory IL-17 cells was significantly lower in high-dose rapamycin group than in the model group (43±1.83 vs 153.5±7.02). High-dose rapamycin obviously inhibited the production of IL-12, IFN-γ, IL-17 and IL-23 and induced the anti-inflammatory cytokines IL-10 and TGF-β. The percentage of Treg in CD4+ T cells was significantly higher in high- dose rapamycin group than in the model group (10.17 ± 0.68 vs 3.52 ± 0.32). In the in vitro experiment, combined treatments of the lymphocytes isolated from the mice with rapamycin and TGF-β induced a significant increase in the number of Treg cells (13.66±1.89) compared with the treatment with rapamycin (6.23±0.80) or TGF-β (4.87±0.85) alone. Rapamycin also obviously up-regulated the expression of p-Smad2 and p-Smad3 in the lymphocytes. CONCLUSIONS Rapamycin can promote the differentiation of Treg cells by up-regulating the expression of p-Smad2 and p-smad3 to improve neurological deficits in mice with EAE.
Collapse
Affiliation(s)
- Zhenfei Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lingling Nie
- Shijiazhuang Circulating Chemical Park Hospital, Shijiazhuang 050000, China
| | - Liping Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yafei Sun
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Li Guo
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
8
|
Yang C, Huang XR, Fung E, Liu HF, Lan HY. The Regulatory T-cell Transcription Factor Foxp3 Protects against Crescentic Glomerulonephritis. Sci Rep 2017; 7:1481. [PMID: 28469165 PMCID: PMC5431186 DOI: 10.1038/s41598-017-01515-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 12/31/2022] Open
Abstract
Regulatory T cells (Tregs) have been shown to play a protective role in glomerulonephritis (GN) and Foxp3 is a master transcription factor in Treg development. In this study, we examined the functional role and mechanisms of Foxp3 in a mouse model of accelerated anti-glomerular basement membrane (anti-GBM) GN induced in antigen-primed Foxp3 transgenic (Tg) mice. Compared with littermate of wildtype (WT) mice in which induced severe crescentic GN developed with progressive renal dysfunction, Foxp3 Tg mice had reduced crescent formation, urinary protein excretion, plasma creatinine and decline in creatinine clearance. The protective role of Foxp3 in crescentic GN was associated with a markedly suppressed expression of proinflammatory interleukin-1 beta (IL-1β), tumour necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein 1 (MCP-1), and diminished infiltration of the kidneys by CD3+ T cells and F4/80+ macrophages. Moreover, overexpression of Foxp3 resulted in a significant increase in CD4+ Foxp3+ Tregs systemically and in the diseased kidneys, thereby blunting Th1, Th2, and Th17 responses systemically and in the kidneys. In conclusion, Foxp3 protects against kidney injury in crescentic GN through enhancement of Treg numbers and function, and suppression of Th1, Th2 and Th17 immune responses at the systemic and local tissue levels.
Collapse
Affiliation(s)
- Chen Yang
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China.,Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Erik Fung
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Hua-Feng Liu
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|