1
|
Lin Y, Yang Q, Zeng R. Crosstalk between macrophages and adjacent cells in AKI to CKD transition. Ren Fail 2025; 47:2478482. [PMID: 40110623 PMCID: PMC11926904 DOI: 10.1080/0886022x.2025.2478482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/17/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Acute kidney injury (AKI), triggered by ischemia, sepsis, toxicity, or obstruction, is marked by a rapid impairment of renal function and could lead to the initiation and advancement of chronic kidney disease (CKD). The concept of AKI to CKD transition has gained much interest. Despite a series of studies highlighting the diverse roles of renal macrophages in the immune response following AKI, the intricate mechanisms of macrophage-driven cell-cell communication in AKI to CKD transition remains incompletely understood. In this review, we introduce the dynamic phenotype change of macrophages under the different stages of kidney injury. Importantly, we present novel perspectives on the extensive interaction of renal macrophages with adjacent cells, including tubular epithelial cells, vascular endothelial cells, fibroblasts, and other immune cells via soluble factors, extracellular vesicles, and direct contact, to facilitate the transition from AKI to CKD. Additionally, we summarize the potential therapeutic strategies based on the adverse macrophage-neighboring cell crosstalk.
Collapse
Affiliation(s)
- Yanping Lin
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zeng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Wang Y, Deng B, Pan Y, Ding F. Identification of Potential Targets Associated With Programmed Cell Death for Acute Kidney Injury Based on WGCNA. Cell Biol Int 2025; 49:723-735. [PMID: 40116046 DOI: 10.1002/cbin.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/12/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025]
Abstract
Programmed cell death (PCD) pathways play a crucial role in maintaining normal cell turnover and tissue homeostasis, encompassing apoptosis and regulated necrosis. However, the involvement of PCD in the pathogenesis of acute kidney disease remains unexplored. In this study, we utilized gene expression profiling datasets (GSE139061) obtained from the Gene Expression Omnibus (GEO) database. Through differential gene expression analysis and weighted gene co-expression network analysis (WGCNA), we identified five key genes associated with PCD, namely DPP4, ATF3, KIT, MSX1, and SNAI2 in acute kidney injury (AKI). Subsequently, single sample gene set enrichment analysis (ssGSEA) was employed to demonstrate the correlation between these five hub genes and immune cell infiltration as well as activation of immune pathways. Furthermore, we validated our findings by analyzing gene expression patterns using a mouse model of ischemia-reperfusion injury. In conclusion, our study is the first to propose the concept of PCD in the pathogenesis of AKI. This finding has significant implications for future investigations into pro-inflammatory immune mechanisms mediated by damage-associated molecular patterns (DAMPs) during the stages of AKI. Our findings underscore the necessity for further investigation into these molecules, which may offer new avenues for therapeutic intervention in AKI. These identified genes may serve as promising targets for intervention in cases of acute kidney diseases.
Collapse
Affiliation(s)
- Yu Wang
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bo Deng
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Pan
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Feng Ding
- Division of Nephrology & Unit of Critical Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Hodgin JB, Maity S, Kretzler M, Sharma K. Pathology of Chronic Kidney Disease and Spatial Metabolomics. Semin Nephrol 2025:151579. [PMID: 40335369 DOI: 10.1016/j.semnephrol.2025.151579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Chronic kidney disease (CKD) has diverse etiologies but exhibits common features in presentation and progression. These include glomerular sclerosis, tubular atrophy, and interstitial fibrosis, often with inflammation and vascular rarefaction. Although these pathologic features have been described in CKD for decades, the molecular drivers of the disease process remain poorly understood. In the era of multiomics and spatial biology, the spatial metabolomics platform could well be a critical technology to guide characterization of the shared cellular programs, capturing the important protective and destructive pathways that ultimately culminate in each of these pathologic features. In this review, we discuss the specific approaches and challenges to developing spatial metabolomics signatures for pathologic features in CKD.
Collapse
Affiliation(s)
- Jeffrey B Hodgin
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Soumya Maity
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX; Division of Nephrology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX.
| | - Matthias Kretzler
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI; Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kumar Sharma
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX; Division of Nephrology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX.
| |
Collapse
|
4
|
Qu Z, Chu J, Jin S, Yang C, Zang J, Zhang J, Xu D, Cheng M. Tissue-resident macrophages and renal diseases: landscapes and treatment directions. Front Immunol 2025; 16:1548053. [PMID: 40230850 PMCID: PMC11994677 DOI: 10.3389/fimmu.2025.1548053] [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: 12/19/2024] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Tissue-resident macrophage (TRM) is a specialized subset of macrophage that resides within specific tissues and organs. TRMs play crucial roles in resisting pathogen invasion, maintaining the homeostasis of the immune microenvironment, and promoting tissue repair and regeneration. The development and function of TRMs exhibit significant heterogeneity across different tissues. Kidney TRMs (KTRMs) originate from both embryonic yolk sac erythro-myeloid progenitors and the fetal liver, demonstrating the capacity for self-renewal independent of bone marrow hematopoiesis. KTRMs are not only essential for the maintenance of renal homeostasis and the monitoring of microvascular environment, but contribute to renal injury due to inflammation, fibrosis and immune dysfunction in kidneys. In this review, we summarize currently available studies on the regulatory role of KTRMs in processes of renal injury and repair. The altering effects and underlying mechanisms of KTRMs in regulating local tissue cells and immune cells in different renal diseases are reviewed, primarily including lupus nephritis, diabetic nephropathy, renal fibrosis, and renal carcinoma. Understanding the plasticity and immune regulatory functions of KTRMs may offer new insights into the pathogenesis and the exploration of therapeutic strategies of kidney diseases.
Collapse
Affiliation(s)
- Zhuojian Qu
- School of Basic Medicine, Shandong Second Medical University, Weifang, China
| | - Jinjin Chu
- Center of Medical Research, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Shuyu Jin
- School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Chunjuan Yang
- Center of Medical Research, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Jie Zang
- Center of Medical Research, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Jin Zhang
- Department of Rheumatology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Donghua Xu
- Center of Medical Research, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
- Department of Rheumatology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Min Cheng
- School of Basic Medicine, Shandong Second Medical University, Weifang, China
| |
Collapse
|
5
|
Téllez Garcia JM, Steenvoorden T, Bemelman F, Hilhorst M, Tammaro A, Vogt L. Purinoreceptor P2X7 in Extracellular ATP-Mediated Inflammation through the Spectrum of Kidney Diseases and Kidney Transplantation. J Am Soc Nephrol 2025:00001751-990000000-00602. [PMID: 40152923 DOI: 10.1681/asn.0000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/24/2025] [Indexed: 03/30/2025] Open
Abstract
Extracellular purines not only play a critical role in maintaining a balanced inflammatory response but may also trigger disproportionate inflammation in various kidney pathologies. Extracellular ATP is the most well-characterized inflammatory purine, which serves as a potent extracellular danger-associated molecular pattern ( i.e ., danger-associated molecular pattern). It signals through the P2 purinoreceptors during both acute and chronic kidney damage. The purinoreceptor P2X7 (P2X7R) has been extensively studied in kidney disease because of its potent ability to enhance inflammation by activating the nucleotide-binding oligomerization domain, leucine rich repeat family pyrin domain containing 3 inflammasome in both immune and parenchymal tubular cells and potential role in immunometabolic reprogramming. We will explore how, following a primary insult to the kidney, disturbance of purinergic balance characterized by extracellular ATP-mediated P2X7R activation exacerbates AKI. Second, we will describe how persistent purinergic disbalance promotes a P2X7R-mediated protracted inflammatory reaction leading to the progression of CKD of different etiologies. Finally, we will also highlight the relevant and emerging role of P2X7R signaling in both antigen-presenting cells and adaptive immune cells to modulate cellular and humoral immune responses in kidney transplantation and hypertension. This review underscores that ATP-P2X7R axis is a key driver of pathologic purinergic signaling, representing a largely unexplored but highly promising clinical target against a wide spectrum of kidney diseases.
Collapse
Affiliation(s)
- Juan Miguel Téllez Garcia
- Department of Internal Medicine Nephrology Section, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thei Steenvoorden
- Department of Internal Medicine Nephrology Section, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Frederike Bemelman
- Department of Internal Medicine Nephrology Section, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Marc Hilhorst
- Department of Internal Medicine Nephrology Section, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Alessandra Tammaro
- Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Internal Medicine Nephrology Section, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Wang Z, Zhou Z, Zhao Z, Zhang J, Zhang S, Li L, Fan Y, Li Q. A network toxicology and machine learning approach to investigate the mechanism of kidney injury from melamine and cyanuric acid co-exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 293:118029. [PMID: 40088607 DOI: 10.1016/j.ecoenv.2025.118029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Within the past two decades, high-profile cases of melamine (MA) exposure have raised significant toxicological concerns, particularly regarding food adulteration. While widely used as a fundamental organic chemical intermediate in various household products, MA's potential for unexpected toxicological synergy with its homolog, cyanuric acid (CA), remains a concern. This study aimed to investigate the nephrotoxicity of combined melamine and cyanuric acid (MC) exposure and its underlying mechanisms in rats through an integrative approach, combining network toxicology (NT), bioinformatics, and experimental validation. MATERIALS AND METHODS Rats were exposed to MC at doses of 0/0 mg/kg/day (Control) and 63/63 mg/kg/day (MC) for four weeks. Kidney pathology, injury markers, and RNA sequencing (RNA-seq) data were analyzed to identify differentially expressed genes between the two groups. Bioinformatics analysis, including pathway enrichment and immune microenvironment analysis, was conducted to elucidate the underlying mechanisms of MC-induced kidney injury. Potential target proteins were identified using ChEMBL, STITCH, and GeneCards databases, and hub genes were screened using three machine learning algorithms: LASSO regression, Random Forest, and Molecular Complex Detection. Molecular docking simulations were performed to assess the interactions between MC and the identified hub genes. RESULTS MC exposure resulted in severe kidney morphological and histological changes, as well as elevated levels of kidney injury and fibrosis markers. RNA-seq analysis revealed significant enrichment of immuno-inflammatory and apoptosis-related pathways in the MC group. Immune microenvironment analysis confirmed the infiltration of pro-inflammatory immune cells. Network toxicology analysis identified 20 potential targets associated with MC-induced kidney injury. Two hub genes, Ren and Casp3, were identified as key regulators of the renin-angiotensin-aldosterone system (RAAS) activation and apoptosis, respectively. Further experimental validation, including Western blotting and immunofluorescence, confirmed the upregulation of these proteins. Molecular docking simulations demonstrated strong binding affinities between MC and the two hub proteins. CONCLUSION MC exposure induces significant kidney injury and fibrosis. The activation of the RAAS pathway and apoptosis plays a crucial role in MC-mediated nephrotoxicity. However, additional vivo experimental validation is lacking. Future studies should focus on further exploration for the mechanism of MC-induced nephrotoxicity and more rigorous experimental validation.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zihao Zhao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Junjie Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shengli Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Luping Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yingzhong Fan
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
7
|
Xie L, Zhang K, Pan K, Su X, Zhao X, Li R, Wang Y, Pang H, Fu E, Li Z. Engineered extracellular vesicles promote the repair of acute kidney injury by modulating regulatory T cells and the immune microenvironment. J Transl Med 2025; 23:304. [PMID: 40065372 PMCID: PMC11895318 DOI: 10.1186/s12967-025-06268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and severe clinical condition. However, the underlying mechanisms of AKI have not been fully elucidated, and effective treatment options remain limited. Studies have shown that immune cells play a critical role in AKI, with regulatory T cells (Tregs) being one of the most important immunosuppressive lymphocytes. Tregs proliferation can attenuate AKI, whereas depletion exacerbates kidney injury. Given that endothelial cells (ECs) are the initial cells that interact with immune cells when they invade the tissue parenchyma, ECs are closely associated with immune reactions. METHODS AND RESULTS In this study, P-selectin binding peptide-extracellular vesicles (PBP-EVs) that target and repair ECs are engineered. Transcriptome sequencing reveals that PBP-EVs reduce the expression of inflammatory genes in AKI mice. Using high-resolution intravital two-photon microscopy (TPM), an increased recruitment of Tregs in the kidneys of AKI Foxp3-EGFP transgenic mice following PBP-EVs treatment is observed, as well as significant Lgr5+ renal stem cell proliferation in AKI Lgr5-CreERT2; R26mTmG mice. Additionally, PBP-EVs treatment result in reduced infiltration of inflammatory cells, pathological damage and fibrosis of AKI mice. Upon depletion of Tregs in Foxp3-DTR transgenic mice, we observe diminished therapeutic effect of PBP-EVs on AKI. CONCLUSIONS The experimental results indicate that PBP-EVs can promote the repair and regeneration of AKI by mitigating endothelial cell damage and subsequently modulating Tregs and the immune microenvironment. These findings provide novel insights and strategies for the treatment of AKI.
Collapse
Affiliation(s)
- Lulu Xie
- School of Medicine, Nankai University, Tianjin, 300071, China
| | | | - Kai Pan
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaomin Su
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaotong Zhao
- Institute for Cardiovascular Science, Soochow University, Suzhou, 215006, China
| | - Rui Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yixin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Haotian Pang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Enze Fu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300052, China.
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, 450016, China.
- National Key Laboratory of Kidney Diseases Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
8
|
Zhang Y, Liu Y, Luo S, Liang H, Guo C, Du Y, Li H, Wang L, Wang X, Tang C, Zhou Y. An adoptive cell therapy with TREM2-overexpressing macrophages mitigates the transition from acute kidney injury to chronic kidney disease. Clin Transl Med 2025; 15:e70252. [PMID: 40000418 PMCID: PMC11859120 DOI: 10.1002/ctm2.70252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Macrophages have been shown to contribute to renal injury and fibrosis as well as repair. Recently, Triggering Receptor Expressed on Myeloid Cells 2 (TREM2)-positive macrophages have been shown to play important roles in regulating tissue inflammation and repair. However, it remains unclear whether they can mitigate the transition from acute kidney injury to chronic kidney disease (the AKI-CKD transition). METHODS The AKI-CKD transition was generated by unilateral ischaemia-reperfusion injury (UIRI) in wild-type (WT) and Trem2 knockout mice. F4/80 magnetic beads were used to isolate renal macrophages. Flow cytometry was used to determine the levels of TREM2 and CD11b levels. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting and histological staining were performed to determine the expression of cytokines and fibrotic markers. RNA-seq was used to investigate transcriptomic changes between WT and Trem2 knockout bone marrow-derived macrophages (BMDMs). TREM2-overexpressing macrophages were generated using lentivirus and transferred intravenously to UIRI mice. RESULTS TREM2 macrophages exhibited a strong renal protective effect on the AKI-CKD transition. Genetic deletion of Trem2 resulted in increased renal inflammation and exacerbated renal injury and fibrosis in UIRI mice. Interestingly, we found that hypoxia could increase TREM2 expression in macrophages via HIF-1α. Upregulated TREM2 expression enhanced macrophage phagocytosis and suppressed the expression of pro-inflammatory cytokines, resulting in lower levels of apoptosis and fibrosis in tubular epithelial cells. Using RNA-seq analysis, we showed that the regulatory effects of TREM2 were orchestrated by the PI3K-AKT pathway. Pharmacological regulation of the PI3K-AKT pathway could modulate the macrophage-mediated inflammation and phagocytosis. In addition, an adoptive cell therapy using TREM2-overexpressing macrophages effectively reduced the immune cell infiltration, renal injury and fibrosis in UIRI mice. CONCLUSION Our study not only provides valuable mechanistic insights into the role of Trem2 in the AKI-CKD transition but also offers a new avenue for TREM2-overexpressing macrophage-based adoptive cell therapy to treat kidney diseases. KEY POINTS TREM2 knockout worsens kidney injury and accelerates AKI-CKD transition. TREM2 is upregulated by hypoxia via HIF1α in AKI-CKD transition. An adoptive cell therapy using TREM2-overexpressing macrophages reduces kidney inflammation and fibrosis.
Collapse
Affiliation(s)
- Yating Zhang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yu Liu
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Siweier Luo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hanzhi Liang
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Chipeng Guo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yufei Du
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hongyu Li
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Le Wang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaohua Wang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Chun Tang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yiming Zhou
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
9
|
Zhang B, Wu Y, Wang Z, Gao S, Liu H, Lin Y, Yu P. Unveiling macrophage dynamics and efferocytosis-related targets in diabetic kidney disease: insights from single-cell and bulk RNA-sequencing. Front Immunol 2025; 16:1521554. [PMID: 40046045 PMCID: PMC11879818 DOI: 10.3389/fimmu.2025.1521554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/28/2025] [Indexed: 05/13/2025] Open
Abstract
Background Chronic inflammation and immune imbalance mediated by macrophages are considered pivotal in diabetic kidney disease (DKD). The study aims to clarify the macrophage heterogeneity and phenotype dynamics, and pinpoint critical targets within efferocytosis in DKD. Methods Utilizing early human DKD sequencing data, we computed the potential communication between leukocytes and renal intrinsic cells. Subsequently, we scrutinized the single-cell RNA sequencing (scRNA-seq) data from CD45-enriched immune cells, concentrating on the macrophage subsets in DKD. Pseudotime trajectory analysis was conducted to explore cell development. Differential expression genes (DEGs) from macrophage subgroups and bulk RNA-sequencing were used to identify shared hub genes. The NephroseqV5 platform was employed to evaluate the clinical significance, and the expression of key molecules was validated in DKD tissues. Results Macrophage infiltration rose in DKD, causing inflammation through the release of chemokines. As time progressed, the number of resident macrophages substantially dropped, with diminishing M1-like and increasing M2-like phenotypes relative to early stages. Further analysis pointed to the most enrichment of macrophage function is the phagosome. We overlapped the DEGs with efferocytosis-related genes and identified key genes, including CD36, ITGAM, and CX3CR1, which exhibited significant correlations with macrophages and T cells. The Nephroseq database revealed that they are associated with proteinuria and renal function. Consistent with the validation set, in vivo experiments verified elevated expression levels of key molecules. Conclusions In essence, our research elucidated the dynamics in macrophage subtype transitions. It emphasized three pivotal genes as critical modulators of macrophage efferocytosis in DKD, indicating their potential as innovative biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Binshan Zhang
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Yunqi Wu
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Zhongli Wang
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Suhua Gao
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Hongyan Liu
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Yao Lin
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
| | - Pei Yu
- National Health Commission (NHC) Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin, China
- Department of Nephrology & Blood Purification Center, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Liu H, Mei M, Zhong H, Lin S, Luo J, Huang S, Zhou J. Identification of Anoikis-Related Genes in Chronic Kidney Disease Based on Bioinformatics Analysis Combined with Experimental Validation. J Inflamm Res 2025; 18:973-994. [PMID: 39867944 PMCID: PMC11761547 DOI: 10.2147/jir.s498820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/09/2025] [Indexed: 01/28/2025] Open
Abstract
Background Chronic kidney disease (CKD) is a progressive condition that arises from diverse etiological factors, resulting in structural alterations and functional impairment of the kidneys. We aimed to establish the Anoikis-related gene signature in CKD by bioinformatics analysis. Methods We retrieved 3 datasets from the Gene Expression Omnibus (GEO) database to obtain differentially expressed genes (DEGs), followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) of them, which were intersected with Anoikis-related genes (ARGs) to derive Anoikis-related differentially expressed genes (ARDEGs). Besides, we conducted weighted gene co-expression network analysis (WGCNA) to identify hub genes. And then, we adopted the quantitative real-time PCR (RT-qPCR) assay to validate the hub genes among several CKD animal models. Furthermore, we constructed a competitive endogenous RNA (ceRNA) network for the hub genes utilizing the ENCORI and miRDB databases, while also calculating Spearman correlation coefficients. Ultimately, we applied the CIBERSORTx algorithm to conduct immune infiltration analysis, classifying immune characteristics based on the abundance of 22 immune cell types. Results To summarize, we identified 13 ARDEGs. WGCNA yielded 6 hub genes, all of which demonstrated significant diagnostic potential in univariate logistic regression analysis (P<0.05). The principal pathways enriched were involved in cell cycle progression Toxoplasmosis, Cell adhesion molecules, Influenza A, Pathogenic Escherichia coli infection, Small cell lung cancer, Amoebiasis, TNF signaling pathway, and Leukocyte transendothelial migration. Notably, 6 immune cell types exhibited significant differences (P<0.05) across subgroups with distinct immune characteristics. Moreover, 2 hub genes showed significant variations (P<0.05) across these immune characteristic subtypes. Among the 4 types of CKD mouse models, the mRNA expression levels of LAMB3 and CDH3 were significantly (P<0.05) up-regulated in the model group. Conclusion We identified 6 hub genes that may serve as potential key biomarkers of Anoikis-related progression in CKD.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Manxue Mei
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Hua Zhong
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People’s Republic of China
| | - Shuyin Lin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Jiahui Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Sirong Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
11
|
Song X, Zhu Y, Geng W, Jiao J, Liu H, Chen R, He Q, Wang L, Sun X, Qin W, Geng J, Chen Z. Spatial and single-cell transcriptomics reveal cellular heterogeneity and a novel cancer-promoting Treg cell subset in human clear-cell renal cell carcinoma. J Immunother Cancer 2025; 13:e010183. [PMID: 39755578 PMCID: PMC11748785 DOI: 10.1136/jitc-2024-010183] [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: 07/29/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common histologic type of RCC. However, the spatial and functional heterogeneity of immunosuppressive cells and the mechanisms by which their interactions promote immunosuppression in the ccRCC have not been thoroughly investigated. METHODS To further investigate the cellular and regional heterogeneity of ccRCC, we analyzed single-cell and spatial transcriptome RNA sequencing data from four patients, which were obtained from samples from multiple regions, including the tumor core, tumor-normal interface, and distal normal tissue. On the basis, the findings were investigated in vitro using tissue and blood samples from 15 patients with ccRCC and validated in the broader samples on tissue microarrays. RESULTS In this study, we revealed previously unreported subsets of both stromal and immune cells, as well as mapped their spatial location at finer resolution. In addition, we validated the clusters of tumor cells after removing batch effects according to six characterized gene sets, including epithelial-mesenchymal transitionhigh clusters, metastatic clusters and proximal tubulehigh clusters. Importantly, we identified a special regulatory T (Treg) cell subpopulation that has the molecular characteristics of terminal effector Treg cells but expresses multiple cytokines, such as interleukin (IL)-1β and IL-18. This group of Treg cells has stronger immunosuppressive function and was associated with a worse prognosis in ccRCC cohorts. They were colocalized with MRC1 + FOLR2 + tumor-associated macrophages (TAMs) at the tumor-normal interface to form a positive feedback loop, maintaining a synergistic procarcinogenic effect. In addition, we traced the origin of IL-1β+ Treg cells and revealed that IL-18 can induce the expression of IL-1β in Treg cells via the ERK/NF-κB pathway. CONCLUSIONS We demonstrated a novel cancer-promoting Treg cell subset and its interactions with MRC1 + FOLR2 +TAMs, which provides new insight into Treg cell heterogeneity and potential therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Xiyu Song
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Zhu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenwen Geng
- Department of Breast Surgery, Shandong University, Jinan, Shandong, China
| | - Jianhua Jiao
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Urology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongjiao Liu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ruo Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qian He
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lijuan Wang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiuxuan Sun
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weijun Qin
- Department of Urology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiejie Geng
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xian, Shaanxi, China
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xian, Shaanxi, China
| |
Collapse
|
12
|
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] [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
|
13
|
Miguel V, Shaw IW, Kramann R. Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nat Rev Nephrol 2025; 21:39-56. [PMID: 39289568 DOI: 10.1038/s41581-024-00889-z] [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: 08/15/2024] [Indexed: 09/19/2024]
Abstract
Chronic kidney disease (CKD), defined as persistent (>3 months) kidney functional loss, has a growing prevalence (>10% worldwide population) and limited treatment options. Fibrosis driven by the aberrant accumulation of extracellular matrix is the final common pathway of nearly all types of chronic repetitive injury in the kidney and is considered a hallmark of CKD. Myofibroblasts are key extracellular matrix-producing cells that are activated by crosstalk between damaged tubules and immune cells. Emerging evidence indicates that metabolic alterations are crucial contributors to the pathogenesis of kidney fibrosis by affecting cellular bioenergetics and metabolite signalling. Immune cell functions are intricately connected to their metabolic characteristics, and kidney cells seem to undergo cell-type-specific metabolic shifts in response to damage, all of which can determine injury and repair responses in CKD. A detailed understanding of the heterogeneity in metabolic reprogramming of different kidney cellular subsets is essential to elucidating communication processes between cell types and to enabling the development of metabolism-based innovative therapeutic strategies against CKD.
Collapse
Affiliation(s)
- Verónica Miguel
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Isaac W Shaw
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Rafael Kramann
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany.
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Jin R, Dai Y, Wang Z, Hu Q, Zhang C, Gao H, Yan Q. Unraveling Ferroptosis: A New Frontier in Combating Renal Fibrosis and CKD Progression. BIOLOGY 2024; 14:12. [PMID: 39857243 PMCID: PMC11763183 DOI: 10.3390/biology14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Chronic kidney disease (CKD) is a global health concern caused by conditions such as hypertension, diabetes, hyperlipidemia, and chronic nephritis, leading to structural and functional kidney injury. Kidney fibrosis is a common outcome of CKD progression, with abnormal fatty acid oxidation (FAO) disrupting renal energy homeostasis and leading to functional impairments. This results in maladaptive repair mechanisms and the secretion of profibrotic factors, and exacerbates renal fibrosis. Understanding the molecular mechanisms of renal fibrosis is crucial for delaying CKD progression. Ferroptosis is a type of discovered an iron-dependent lipid peroxidation-regulated cell death. Notably, Ferroptosis contributes to tissue and organ fibrosis, which is correlated with the degree of renal fibrosis. This study aims to clarify the complex mechanisms of ferroptosis in renal parenchymal cells and explore how ferroptosis intervention may help alleviate renal fibrosis, particularly by addressing the gap in CKD mechanisms related to abnormal lipid metabolism under the ferroptosis context. The goal is to provide a new theoretical basis for clinically delaying CKD progression.
Collapse
Affiliation(s)
- Rui Jin
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Dai
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qinyang Hu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongyu Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Enshi 445000, China
| |
Collapse
|
15
|
Zhang L, Xu F, Hou L. IL-6 and diabetic kidney disease. Front Immunol 2024; 15:1465625. [PMID: 39749325 PMCID: PMC11693507 DOI: 10.3389/fimmu.2024.1465625] [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: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes associated with high mortality and disability rates. Inflammation has emerged as a key pathological mechanism in DKD, prompting interest in novel therapeutic approaches targeting inflammatory pathways. Interleukin-6 (IL-6), a well-established inflammatory cytokine known for mediating various inflammatory responses, has attracted great attention in the DKD field. Although multiple in vivo and in vitro studies highlight the potential of targeting IL-6 in DKD treatment, its exact roles in the disease remains unclear. This review presents the roles of IL-6 in the pathogenesis of DKD, including immunoinflammation, metabolism, hemodynamics, and ferroptosis. In addition, we summarize the current status of IL-6 inhibitors in DKD-related clinical trials and discuss the potential of targeting IL-6 for treating DKD in the clinic.
Collapse
Affiliation(s)
- Lei Zhang
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Futian Xu
- Logistics Management Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Liyan Hou
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| |
Collapse
|
16
|
Liu Z, Dai B, Bao J, Pan Y. T cell metabolism in kidney immune homeostasis. Front Immunol 2024; 15:1498808. [PMID: 39737193 PMCID: PMC11684269 DOI: 10.3389/fimmu.2024.1498808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Kidney immune homeostasis is intricately linked to T cells. Inappropriate differentiation, activation, and effector functions of T cells lead to a spectrum of kidney disease. While executing immune functions, T cells undergo a series of metabolic rewiring to meet the rapid energy demand. The key enzymes and metabolites involved in T cell metabolism metabolically and epigenetically modulate T cells' differentiation, activation, and effector functions, thereby being capable of modulating kidney immune homeostasis. In this review, we first summarize the latest advancements in T cell immunometabolism. Second, we outline the alterations in the renal microenvironment under certain kidney disease conditions. Ultimately, we highlight the metabolic modulation of T cells within kidney immune homeostasis, which may shed light on new strategies for treating kidney disease.
Collapse
Affiliation(s)
- Zikang Liu
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Binbin Dai
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jiwen Bao
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yangbin Pan
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
17
|
do Carmo JM, Hall JE, Furukawa LNS, Woronik V, Dai X, Ladnier E, Wang Z, Omoto ACM, Mouton A, Li X, Luna-Suarez EM, da Silva AA. Chronic central nervous system leptin administration attenuates kidney dysfunction and injury in a model of ischemia/reperfusion-induced acute kidney injury. Am J Physiol Renal Physiol 2024; 327:F957-F966. [PMID: 39361725 PMCID: PMC11687842 DOI: 10.1152/ajprenal.00158.2024] [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: 05/28/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024] Open
Abstract
In the present study, we examined whether chronic intracerebroventricular (ICV) leptin administration protects against ischemia/reperfusion (I/R)-induced acute kidney injury (AKI). Twelve-week-old male rats were implanted with an ICV cannula into the right lateral ventricle, and 8-10 days after surgery, leptin (0.021 µg/h, n = 8) or saline vehicle (0.5 µL/h, n = 8) was infused via osmotic minipump connected to the ICV cannula for 12 days. On day 8 of leptin or vehicle infusion, rats were submitted to unilateral ischemia/reperfusion (UIR) by clamping the left pedicle for 30 min. To control for leptin-induced reductions in food intake, the vehicle-treated group was pair-fed (UIR-PF) to match the same amount of food consumed by leptin-treated (UIR-Leptin) rats. On the 12th day of leptin or vehicle infusion (fourth day after AKI), single-left kidney glomerular filtration rate (GFR) was measured, blood samples were collected to quantify white blood cells, and kidneys were collected for histological assessment of injury. UIR-Leptin-treated rats showed reduced right and left kidney weights (right: 1,040 ± 24 vs. 1,281 ± 36 mg; left: 1,127 ± 71 vs. 1,707 ± 45 mg, for UIR-Leptin and UIR-PF, respectively). ICV leptin infusion improved GFR (0.50 ± 0.06 vs. 0.13 ± 0.03 mL/min/g kidney wt) and reduced kidney injury scores. ICV leptin treatment also attenuated the reduction in circulating adiponectin levels that was observed in UIR-PF rats and increased the circulating white blood cells count compared with UIR-PF rats (16.3 ± 1.3 vs. 9.8 ± 0.6 k/µL). Therefore, we show that leptin, via its actions on the central nervous system, confers significant protection against major kidney dysfunction and injury in a model of ischemia/reperfusion-induced AKI.NEW & NOTEWORTHY A major new finding of this study is that chronic activation of leptin receptors in the CNS markedly attenuates acute kidney injury and protects against severe renal dysfunction after ischemia/reperfusion, independently of leptin's anorexic effects.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - John E Hall
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Luzia N S Furukawa
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Viktoria Woronik
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Xuemei Dai
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emily Ladnier
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Zhen Wang
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Ana C M Omoto
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alan Mouton
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Xuan Li
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emilio M Luna-Suarez
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
18
|
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
|
19
|
Tahvildari M, Me R, Setia M, Gao N, Suvas P, McClellan SA, Suvas S. Foxp3 + regulatory T cells reside within the corneal epithelium and co-localize with limbal stem cells. Exp Eye Res 2024; 249:110123. [PMID: 39396695 PMCID: PMC11622170 DOI: 10.1016/j.exer.2024.110123] [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: 06/16/2024] [Revised: 09/08/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
In this study we investigated the presence of resident Foxp3+ regulatory T cells (Tregs) within the cornea and assessed the role of resident Tregs in corneal epithelial wound healing. Using a mouse model, we showed that in the steady state Foxp3+Tregs are either in close proximity or co-localize with ABCG2+ limbal stem cells. We also showed that these Tregs reside within the epithelial layer and not the corneal stroma. In addition, using a mouse model of mechanical injury, we demonstrated that depletion of Tregs from the cornea prior to corneal mechanical injury, using subconjunctival injection of anti-CD25, was associated with delayed epithelial healing. These results suggest a role for cornea resident Tregs in corneal epithelial cell function and wound healing and opens doors for further exploration of the role of Tregs in limbal stem cell function and survival.
Collapse
Affiliation(s)
- Maryam Tahvildari
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA; Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA.
| | - Rao Me
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mizumi Setia
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nan Gao
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Pratima Suvas
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sharon A McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Susmit Suvas
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
20
|
Noel S, Kapoor R, Rabb H. New approaches to acute kidney injury. Clin Kidney J 2024; 17:65-81. [PMID: 39583139 PMCID: PMC11581771 DOI: 10.1093/ckj/sfae265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Indexed: 11/26/2024] Open
Abstract
Acute kidney injury (AKI) is a common and serious clinical syndrome that involves complex interplay between different cellular, molecular, metabolic and immunologic mechanisms. Elucidating these pathophysiologic mechanisms is crucial to identify novel biomarkers and therapies. Recent innovative methodologies and the advancement of existing technologies has accelerated our understanding of AKI and led to unexpected new therapeutic candidates. The aim of this review is to introduce and update the reader about recent developments applying novel technologies in omics, imaging, nanomedicine and artificial intelligence to AKI research, plus to provide examples where this can be translated to improve patient care.
Collapse
Affiliation(s)
- Sanjeev Noel
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Radhika Kapoor
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
21
|
Humphries TLR, Gobe GC, Urquhart AJ, Ellis RJ, Galloway GJ, Vesey DA, Francis RS. Identifying biochemical changes in the kidney using proton nuclear magnetic resonance in an adenine diet chronic kidney disease mouse model. NMR IN BIOMEDICINE 2024; 37:e5257. [PMID: 39229964 DOI: 10.1002/nbm.5257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
This study aimed to investigate the metabolic changes in the kidneys in a murine adenine-diet model of chronic kidney disease (CKD). Kidney fibrosis is the common pathological manifestation across CKD aetiologies. Sustained inflammation and fibrosis cause changes in preferred energy metabolic pathways in the cells of the kidney. Kidney cortical tissue from mice receiving a control or adenine-supplemented diet for 8 weeks (late inflammation and fibrosis) and 12 weeks (8 weeks of treatment followed by 4 weeks recovery) were analysed by 2D-correlated nuclear magnetic resonance spectroscopy and compared with histopathology and biomarkers of kidney damage. Tissue metabolite and lipid levels were assessed using the MestreNova software. Expression of genes related to inflammation, fibrosis, and metabolism were measured using quantitative polymerase chain reaction. Animals showed indicators of severely impaired kidney function at 8 and 12 weeks. Significantly increased fibrosis was present at 8 weeks but not in the recovery group suggesting some reversal of fibrosis and amelioration of inflammation. At 8 weeks, metabolites associated with glycolysis were increased, while lipid signatures were decreased. Genes involved in fatty acid oxidation were decreased at 8 weeks but not 12 weeks while genes associated with glycolysis were significantly increased at 8 weeks but not at 12 weeks. In this murine model of CKD, kidney fibrosis was associated with the accumulation of triglyceride and free lactate. There was an up-regulation of glycolytic enzymes and down-regulation of lipolytic enzymes. These metabolic changes reflect the energy demands associated with progressive kidney disease where there is a switch from fatty acid oxidation to that of glycolysis.
Collapse
Affiliation(s)
- Tyrone L R Humphries
- The University of Queensland, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Glenda C Gobe
- The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Aaron J Urquhart
- The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Robert J Ellis
- The University of Queensland, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Graham J Galloway
- The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - David A Vesey
- The University of Queensland, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Ross S Francis
- The University of Queensland, Brisbane, Australia
- Princess Alexandra Hospital, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| |
Collapse
|
22
|
Li Y, Zhang J, Qiu X, Zhang Y, Wu J, Bi Q, Sun Z, Wang W. Diverse regulated cell death patterns and immune traits in kidney allograft with fibrosis: a prediction of renal allograft failure based on machine learning, single-nucleus RNA sequencing and molecular docking. Ren Fail 2024; 46:2435487. [PMID: 39632251 PMCID: PMC11619039 DOI: 10.1080/0886022x.2024.2435487] [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: 05/26/2024] [Revised: 11/02/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives: Post-transplant allograft fibrosis remains a challenge in prolonging allograft survival. Regulated cell death has been widely implicated in various kidney diseases, including renal fibrosis. However, the role of different regulated cell death (RCD) pathways in post-transplant allograft fibrosis remains unclear. Methods and Results: Microarray transcriptome profiling and single-nuclei sequencing data of post-transplant fibrotic and normal grafts were obtained and used to identify RCD-related differentially expressed genes. The enrichment activity of nine RCD modalities in tissue and cells was examined using single-sample gene set enrichment analysis, and their relations with immune infiltration in renal allograft samples were also assessed. Parenchymal and non-parenchymal cells displayed heterogeneity in RCD activation. Additionally, cell-cell communication analysis was also conducted in fibrotic samples. Subsequently, weighted gene co-expression network analysis and seven machine learning algorithms were employed to identify RCD-related hub genes for renal fibrosis. A 9-gene signature, termed RCD risk score (RCDI), was constructed using the least absolute shrinkage and selection operator and multivariate Cox regression algorithms. This signature showed robust accuracy in predicting 1-, 2-, and 3-year allograft survival status (area under the curve for 1-, 2-, and 3-year were 0.900, 0.877, 0.858, respectively). Immune infiltration analysis showed a strong correlation with RCDI and the nine model genes. Finally, molecular docking simulation suggested rapamycin, tacrolimus and mycophenolate mofetil exhibit strong interactions with core RCD-related receptors. Conclusions: In summary, this study explored the activation of nine RCD pathways and their relationships with immune traits, identified potential RCD-related hub genes associated with renal fibrosis, and highlighted potential therapeutic targets for renal allograft fibrosis.
Collapse
Affiliation(s)
- Yuqing Li
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Xuemeng Qiu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Yifei Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Qing Bi
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Lu X, Ma K, Ren J, Peng H, Wang J, Wang X, Nasser MI, Liu C. The immune regulatory role of lymphangiogenesis in kidney disease. J Transl Med 2024; 22:1053. [PMID: 39578812 PMCID: PMC11583545 DOI: 10.1186/s12967-024-05859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
The renal lymphatic system is critical for maintaining kidney homeostasis and regulating the immune response inside the kidney. In various kidney pathological situations, the renal lymphatic network experiences lymphangiogenesis, which is defined as the creation of new lymphatic vessels. Kidney lymphangiogenesis controls immunological response inside the kidney by controlling lymphatic flow, immune cell trafficking, and immune cell regulation. Ongoing study reveals lymphangiogenesis's different architecture and functions in numerous tissues and organs. New research suggests that lymphangiogenesis in kidney disorders may regulate the renal immune response in various ways. The flexibility of lymphatic endothelial cells (LECs) improves the kidney's immunological regulatory function of lymphangiogenesis. Furthermore, current research has shown disparate findings regarding its impact on distinct renal diseases, resulting in contradictory outcomes even within the same kidney condition. The fundamental causes of the various effects of lymphangiogenesis on renal disorders remain unknown. In this thorough review, we explore the dual impacts of renal lymphangiogenesis on several kidney pathologies, with a particular emphasis on existing empirical data and new developments in understanding its immunological regulatory function in kidney disease. An improved understanding of the immunological regulatory function of lymphangiogenesis in kidney diseases might help design novel medicines targeting lymphatics to treat kidney pathologies.
Collapse
Affiliation(s)
- Xiangheng Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jia Wang
- General Practice Center, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Moussa Ide Nasser
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, 510100, Guangdong, China.
| | - Chi Liu
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, China.
| |
Collapse
|
24
|
Tasca P, van den Berg BM, Rabelink TJ, Wang G, Heijs B, van Kooten C, de Vries APJ, Kers J. Application of spatial-omics to the classification of kidney biopsy samples in transplantation. Nat Rev Nephrol 2024; 20:755-766. [PMID: 38965417 DOI: 10.1038/s41581-024-00861-x] [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: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Improvement of long-term outcomes through targeted treatment is a primary concern in kidney transplant medicine. Currently, the validation of a rejection diagnosis and subsequent treatment depends on the histological assessment of allograft biopsy samples, according to the Banff classification system. However, the lack of (early) disease-specific tissue markers hinders accurate diagnosis and thus timely intervention. This challenge mainly results from an incomplete understanding of the pathophysiological processes underlying late allograft failure. Integration of large-scale multimodal approaches for investigating allograft biopsy samples might offer new insights into this pathophysiology, which are necessary for the identification of novel therapeutic targets and the development of tailored immunotherapeutic interventions. Several omics technologies - including transcriptomic, proteomic, lipidomic and metabolomic tools (and multimodal data analysis strategies) - can be applied to allograft biopsy investigation. However, despite their successful application in research settings and their potential clinical value, several barriers limit the broad implementation of many of these tools into clinical practice. Among spatial-omics technologies, mass spectrometry imaging, which is under-represented in the transplant field, has the potential to enable multi-omics investigations that might expand the insights gained with current clinical analysis technologies.
Collapse
Affiliation(s)
- Paola Tasca
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (Renew), Leiden University Medical Center, Leiden, the Netherlands
| | - Gangqi Wang
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (Renew), Leiden University Medical Center, Leiden, the Netherlands
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
- Bruker Daltonics GmbH & Co. KG, Bremen, Germany
| | - Cees van Kooten
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Aiko P J de Vries
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands.
- Department of Internal Medicine, Division of Nephrology, Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Jesper Kers
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Center for Analytical Sciences Amsterdam, Van't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Liu M, Chen X. Human Umbilical Cord-Derived Mesenchymal Stem Cells-Exosomes-Delivered miR-375 Targets HDAC4 to Promote Autophagy and Suppress T Cell Apoptosis in Sepsis-Associated Acute Kidney Injury. Appl Biochem Biotechnol 2024; 196:7954-7973. [PMID: 38668845 DOI: 10.1007/s12010-024-04963-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 12/14/2024]
Abstract
This study sought to elucidate the mechanism of human umbilical cord-derived mesenchymal stem cells (HUCMSCs)-exosomes (Exos) in sepsis-associated acute kidney injury (SAKI). Exos were isolated from HUCMSCs and co-cultured with CD4+ T cells exposed to lipopolysaccharide to detect the effects of HUCMSCs-Exos on CD4+ T cell apoptosis and autophagy. miR-375 expression in CD4+ T cells and HUCMSCs-Exos was examined. The relationship between miR-375 and HDAC4 was analyzed. A mouse model of SAKI was established and injected with HUCMSCs-Exos to verify the function of HUCMSCs-Exos in vivo. HUCMSCs-Exos inhibited lipopolysaccharide-induced apoptosis of CD4+ T cells and promoted autophagy. miR-375 expression was noted to be elevated in the HUCMSCs-Exos. Importantly, HUCMSCs-Exos could deliver miR-375 into CD4+ T cells where miR-375 targeted HDAC4 and negatively regulated its expression. By this mechanism, HUCMSCs-Exos decreased CD4+ T cell apoptosis and augmented autophagy. This finding was further confirmed in an in vivo SAKI model. Collectively, HUCMSCs-Exos can protect against SAKI via delivering miR-375 that promotes autophagy and arrests T cell apoptosis through HDAC4 downregulation. These findings suggest a promising therapeutic potential for HUCMSCs-Exos in the context of SAKI.
Collapse
Affiliation(s)
- Min Liu
- Department of Intensive Care, the First Hospital of Changsha, No. 311 Yingpan Road, Changsha, Hunan, 410005, People's Republic of China
| | - Xiyun Chen
- Department of Gynecology, the First Hospital of Changsha, No. 311 Yingpan Road, Changsha, Hunan, 410005, People's Republic of China.
| |
Collapse
|
26
|
Zhou J, Liu W, Liu X, Wu J, Chen Y. Independent and joint influence of depression and advanced lung cancer inflammation index on mortality among individuals with chronic kidney disease. Front Nutr 2024; 11:1453062. [PMID: 39507908 PMCID: PMC11539836 DOI: 10.3389/fnut.2024.1453062] [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: 06/22/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024] Open
Abstract
Background The combined effect of depression and nutritional-inflammatory status on mortality in the chronic kidney disease (CKD) population is unclear. Methods We prospectively analyzed 3,934 (weighted population: 22,611,423) CKD participants from the National Health and Nutrition Examination Survey (2007-2018). Depression and nutritional-inflammatory status were assessed with Patient Health Questionnaire 9 (PHQ-9) and Advanced Lung Cancer Inflammation Index (ALI), respectively. Weighted multivariate COX regression models, restricted cubic splines (RCS) models, and stratified analyses were used to investigate the association of PHQ-9 scores and ALI with all-cause mortality. Results During a median follow-up of 5.8 years (interquartile range 3.4-8.6 years), a total of 985 patients died (25.0%). Each point increase in a patient's PHQ-9 score increased the risk of all-cause mortality by 4% (HR, 1.04; 95% CI, 1.02-1.06; p < 0.001), in the full adjusted model. However, an increase in ALI levels was associated with a decreased risk. HRs (95% CI) of 0.76 (0.65-0.90), 0.70 (0.57-0.86), and 0.51 (0.41-0.64) in the Q2, Q3, and Q4 of ALI compared with the Q1 of ALI, respectively. In addition, the joint analysis showed that CKD patients without depression and with higher ALI were associated with a reduced risk of all-cause mortality. Namely, patients in the highest ALI group (Q4) without depression had the lowest risk (HR, 0.32; 95% CI, 0.21-0.48). Furthermore, this combined effect was consistent across all subgroups, and no significant interaction was found (p > 0.05 for interaction). Conclusion In a nationally representative sample of US patients with CKD, coexisting depression and poorer nutrition-inflammation were associated with a significantly increased risk of all-cause mortality.
Collapse
Affiliation(s)
- Jie Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Liu
- Department of Vascular Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoxin Liu
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jijun Wu
- Department of Interventional Radiology, Zhongshan Torch Development Zone People's Hospital, Zhongshan, China
- Third Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Ying Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
Xu C, Zhang Y, Zhou J, Zhang J, Dong H, Chen X, Tian Y, Wu Y. Integrated temporal transcriptional and epigenetic single-cell analysis reveals the intrarenal immune characteristics in an early-stage model of IgA nephropathy during its acute injury. Front Immunol 2024; 15:1405748. [PMID: 39493754 PMCID: PMC11528150 DOI: 10.3389/fimmu.2024.1405748] [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: 03/23/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Rationale Kidney inflammation plays a crucial role in the pathogenesis of IgA nephropathy (IgAN), yet the specific phenotypes of immune cells involved in disease progression remain incompletely understood. Utilizing joint profiling through longitudinal single-cell RNA-sequencing (scRNAseq) and single-cell assay for transposase-accessible chromatin sequencing (scATACseq) can provide a comprehensive framework for elucidating the development of cell subset diversity and how chromatin accessibility regulates transcription. Objective We aimed to characterize the dynamic immune cellular landscape at a high resolution in an early IgAN mouse model with acute kidney injury (AKI). Methods and results A murine model was utilized to mimic 3 immunological states -"immune stability (IS), immune activation (IA) and immune remission (IR)" in early human IgAN-associated glomerulopathy during AKI, achieved through lipopolysaccharide (LPS) injection. Urinary albumin to creatinine ratio (UACR) was measured to further validate the exacerbation and resolution of kidney inflammation during this course. Paired scRNAseq and scATACseq analysis was performed on CD45+ immune cells isolated from kidney tissues obtained from CTRL (healthy vehicle), IS, IA and IR (4 or 5 mice each). The analyses revealed 7 major cell types and 24 clusters based on 72304 single-cell transcriptomes, allowing for the identification and characterization of various immune cell types within each cluster. Our data offer an impartial depiction of the immunological characteristics, as the proportions of immune cell types fluctuated throughout different stages of the disease. Specifically, these analyses also revealed novel subpopulations, such as a macrophage subset (Nlrp1b Mac) with distinct epigenetic features and a unique transcription factor motif profile, potentially exerting immunoregulatory effects, as well as an early subset of Tex distinguished by their effector and cytolytic potential (CX3CR1-transTeff). Furthermore, in order to investigate the potential interaction between immune cells and renal resident cells, we conducted single-cell RNA sequencing on kidney cells obtained from a separate cohort of IS and IA mice without isolating immune cells. These findings underscored the diverse roles played by macrophages and CD8+ T cells in maintaining homeostasis of endothelial cells (ECs) under stress. Conclusions This study presents a comprehensive analysis of the dynamic changes in immune cell profiles in a model of IgAN, identifying key cell types and their roles and interactions. These findings significantly contribute to the understanding of the pathogenesis of IgAN and may provide potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Chen Xu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiwei Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Zhou
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiangnan Zhang
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Dong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese People's Liberation Army (PLA) General Hospital, Chinese People's Liberation Army (PLA) Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yi Tian
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
28
|
Chen Q, Zhang X, Yang H, Luo G, Zhou X, Xu Z, Xu A. CD8 + CD103 + iTregs protect against ischemia-reperfusion-induced acute kidney Injury by inhibiting pyroptosis. Apoptosis 2024; 29:1709-1722. [PMID: 39068624 DOI: 10.1007/s10495-024-02001-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] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
The occurrence of acute kidney injury (AKI) is elevated, one of the main causes is ischemia-reperfusion (I/R). However, no specific therapy is currently available to treat I/R-induced AKI (I/R-AKI). Treg cells have been demonstrated to perform an anti-inflammatory role in a range of autoimmune and inflammatory illnesses. However, there is limited available information about the possible functions of CD8 + CD103 + iTregs in I/R-AKI. We utilized renal tubular epithelial cells (RTECs) subjected to hypoxia-reoxygenation (H/R) and I/R-AKI mouse model to investigate whether CD8 + CD103 + iTregs could attenuate AKI and the underlying mechanism. In vitro, co-cultured with CD8 + CD103 + iTregs alleviated H/R-induced cell injury. After treatment of CD8 + CD103 + iTregs rather than control cells, a significant improvement of I/R-AKI was observed in vivo, including decreased serum creatinine (sCr) and blood urea nitrogen (BUN) levels, reduced renal pathological injury, lowered tubular apoptosis and inhibition of the transition from AKI to chronic kidney disease (CKD). Mechanically, CD8 + CD103 + iTregs alleviated H/R-induced cell injury and I/R-AKI partly by suppressing RTECs pyroptosis via inhibiting the NLRP3/Caspase-1 axis. Our study provides a novel perspective on the possibility of CD8 + CD103 + iTregs for the treatment of I/R-AKI.
Collapse
Affiliation(s)
- Qiuju Chen
- Department of Nephrology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Xiao Zhang
- Department of Nephrology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510120, China
| | - Hui Yang
- Department of Nephrology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Guangxuan Luo
- Department of Nephrology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Xin Zhou
- Department of Nephrology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhenjian Xu
- Department of Nephrology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
| | - Anping Xu
- Department of Medicine, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Nephrology, PengPai Memorial Hospital, Shanwei, 516400, China.
| |
Collapse
|
29
|
Tang M, Shen L, Tang M, Liu L, Rao Z, Wang Z, Wang Y, Yin S, Li S, Xu G, Zhang K. Human mesenchymal stromal cells ameliorate cisplatin-induced acute and chronic kidney injury via TSG-6. Stem Cells 2024; 42:848-859. [PMID: 38804841 DOI: 10.1093/stmcls/sxae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Cisplatin is widely used in tumor chemotherapy, but nephrotoxicity is an unavoidable side effect of cisplatin. Several studies have demonstrated that mesenchymal stromal cells (MSCs) ameliorate cisplatin-induced kidney injury, but the underlying mechanisms are unknown. In this study, the cisplatin-induced kidney injury mouse model was established by subjecting a single intraperitoneal injection with cisplatin. One hour before cisplatin injection, the mice received human bone marrow MSCs (hBM-MSCs) with or without siRNA-transfection, recombinant human tumor necrosis factor-α-stimulated gene/protein 6 (rhTSG-6), or PBS through the tail vein. In addition, cisplatin-stimulated HK-2 cells were treated with hBM-MSCs or rhTSG-6. Human BM-MSCs treatment remarkably ameliorated cisplatin-induced acute and chronic kidney injury, as evidenced by significant reductions in serum creatinine (Scr), blood urea nitrogen, tubular injury, collagen deposition, α-smooth muscle actin accumulation, as well as inflammatory responses, and by remarkable increased anti-inflammatory factor expression and Treg cells infiltration in renal tissues. Furthermore, we found that only a few hBM-MSCs engrafted into damaged kidney and that the level of human TSG-6 in the serum of mice increased significantly following hBM-MSCs administration. Moreover, hBM-MSCs significantly increased the viability of damaged HK-2 cells and decreased the levels of inflammatory cytokines in the culture supernatant. However, the knockdown of the TSG-6 gene in hBM-MSCs significantly attenuated their beneficial effects in vivo and in vitro. On the contrary, treated with rhTSG-6 achieved similar beneficial effects of hBM-MSCs. Our results indicate that systemic administration of hBM-MSCs alleviates cisplatin-induced acute and chronic kidney injury in part by paracrine TSG-6 secretion.
Collapse
Affiliation(s)
- Ming Tang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Linguo Shen
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Maozhi Tang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Ling Liu
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Zhengsheng Rao
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Zhilin Wang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Yadi Wang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Supei Yin
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Shujing Li
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| | - Guilian Xu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, People's Republic of China
| | - Keqin Zhang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400065, People's Republic of China
| |
Collapse
|
30
|
Wu H, Qiu Z, Wang L, Li W. Renal Fibrosis: SIRT1 Still of Value. Biomedicines 2024; 12:1942. [PMID: 39335456 PMCID: PMC11428497 DOI: 10.3390/biomedicines12091942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern. Renal fibrosis, a prevalent outcome regardless of the initial cause, ultimately leads to end-stage renal disease. Glomerulosclerosis and renal interstitial fibrosis are the primary pathological features. Preventing and slowing renal fibrosis are considered effective strategies for delaying CKD progression. However, effective treatments are lacking. Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase belonging to class III histone deacetylases, is implicated in the physiological regulation and protection of the kidney and is susceptible to a diverse array of pathological influences, as demonstrated in previous studies. Interestingly, controversial conclusions have emerged as research has progressed. This review provides a comprehensive summary of the current understanding and advancements in the field; specifically, the biological roles and mechanisms of SIRT1 in regulating renal fibrosis progression. These include aspects such as lipid metabolism, epithelial-mesenchymal transition, oxidative stress, aging, inflammation, and autophagy. This manuscript explores the potential of SIRT1 as a therapeutic target for renal fibrosis and offers new perspectives on treatment approaches and prognostic assessments.
Collapse
Affiliation(s)
- Huailiang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| | - Liyan Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.W.); (Z.Q.)
| |
Collapse
|
31
|
Bao Y, Shan Q, Lu K, Yang Q, Liang Y, Kuang H, Wang L, Hao M, Peng M, Zhang S, Cao G. Renal tubular epithelial cell quality control mechanisms as therapeutic targets in renal fibrosis. J Pharm Anal 2024; 14:100933. [PMID: 39247486 PMCID: PMC11377145 DOI: 10.1016/j.jpha.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 09/10/2024] Open
Abstract
Renal fibrosis is a devastating consequence of progressive chronic kidney disease, representing a major public health challenge worldwide. The underlying mechanisms in the pathogenesis of renal fibrosis remain unclear, and effective treatments are still lacking. Renal tubular epithelial cells (RTECs) maintain kidney function, and their dysfunction has emerged as a critical contributor to renal fibrosis. Cellular quality control comprises several components, including telomere homeostasis, ubiquitin-proteasome system (UPS), autophagy, mitochondrial homeostasis (mitophagy and mitochondrial metabolism), endoplasmic reticulum (ER, unfolded protein response), and lysosomes. Failures in the cellular quality control of RTECs, including DNA, protein, and organelle damage, exert profibrotic functions by leading to senescence, defective autophagy, ER stress, mitochondrial and lysosomal dysfunction, apoptosis, fibroblast activation, and immune cell recruitment. In this review, we summarize recent advances in understanding the role of quality control components and intercellular crosstalk networks in RTECs, within the context of renal fibrosis.
Collapse
Affiliation(s)
- Yini Bao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiyuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ying Liang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haodan Kuang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuosheng Zhang
- College of Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030600, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310009, China
| |
Collapse
|
32
|
Zhou T, Fang YL, Tian TT, Wang GX. Pathological mechanism of immune disorders in diabetic kidney disease and intervention strategies. World J Diabetes 2024; 15:1111-1121. [PMID: 38983817 PMCID: PMC11229953 DOI: 10.4239/wjd.v15.i6.1111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/29/2024] [Accepted: 04/15/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetic kidney disease is one of the most severe chronic microvascular complications of diabetes and a primary cause of end-stage renal disease. Clinical studies have shown that renal inflammation is a key factor determining kidney damage during diabetes. With the development of immunological technology, many studies have shown that diabetic nephropathy is an immune complex disease, and that most patients have immune dysfunction. However, the immune response associated with diabetic nephropathy and autoimmune kidney disease, or caused by ischemia or infection with acute renal injury, is different, and has a com-plicated pathological mechanism. In this review, we discuss the pathogenesis of diabetic nephropathy in immune disorders and the intervention mechanism, to provide guidance and advice for early intervention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun 130021, Jilin Province, China
| | - Yi-Lin Fang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun 130021, Jilin Province, China
| | - Tian-Tian Tian
- School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Gui-Xia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
33
|
Lee K, Gharaie S, Kurzhagen JT, Newman-Rivera AM, Arend LJ, Noel S, Rabb H. Double-negative T cells have a reparative role after experimental severe ischemic acute kidney injury. Am J Physiol Renal Physiol 2024; 326:F942-F956. [PMID: 38634135 PMCID: PMC11386976 DOI: 10.1152/ajprenal.00376.2023] [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: 11/27/2023] [Revised: 03/25/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024] Open
Abstract
T cells mediate organ injury and repair. A proportion of unconventional kidney T cells called double-negative (DN) T cells (TCR+ CD4- CD8-), with anti-inflammatory properties, were previously demonstrated to protect from early injury in moderate experimental acute kidney injury (AKI). However, their role in repair after AKI has not been studied. We hypothesized that DN T cells mediate repair after severe AKI. C57B6 mice underwent severe (40 min) unilateral ischemia-reperfusion injury (IRI). Kidney DN T cells were studied by flow cytometry and compared with gold-standard anti-inflammatory CD4+ regulatory T cells (Tregs). In vitro effects of DN T cells and Tregs on renal tubular epithelial cell (RTEC) repair after injury were quantified with live-cell analysis. DN T cells, Tregs, CD4, or vehicle were adoptively transferred after severe AKI. Glomerular filtration rate (GFR) was measured using fluorescein isothiocyanate (FITC)-sinistrin. Fibrosis was assessed with Masson's trichrome staining. Profibrotic genes were measured with qRT-PCR. Percentages and the numbers of DN T cells substantially decreased during repair phase after severe AKI, as well as their activation and proliferation. Both DN T cells and Tregs accelerated RTEC cell repair in vitro. Post-AKI transfer of DN T cells reduced kidney fibrosis and improved GFR, as did Treg transfer. DN T cell transfer lowered transforming growth factor (TGF)β1 and α-smooth muscle actin (αSMA) expression. DN T cells reduced effector-memory CD4+ T cells and IL-17 expression. DN T cells undergo quantitative and phenotypical changes after severe AKI, accelerate RTEC repair in vitro as well as improve GFR and renal fibrosis in vivo. DN T cells have potential as immunotherapy to accelerate repair after AKI.NEW & NOTEWORTHY Double-negative (DN) T cells (CD4- CD8-) are unconventional kidney T cells with regulatory abilities. Their role in repair from acute kidney injury (AKI) is unknown. Kidney DN T cell population decreased during repair after ischemic AKI, in contrast to regulatory T cells (Tregs) which increased. DN T cell administration accelerated tubular repair in vitro, while after severe in vivo ischemic injury reduced kidney fibrosis and increased glomerular filtration rate (GFR). DN T cell infusion is a potential therapeutic agent to improve outcome from severe AKI.
Collapse
Affiliation(s)
- Kyungho Lee
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sepideh Gharaie
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Lois J Arend
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
34
|
Gritter M, Wei KY, Wouda RD, Musterd-Bhaggoe UM, Dijkstra KL, Kers J, Ramakers C, Vogt L, de Borst MH, Danser AHJ, Hoorn EJ, Rotmans JI. Chronic kidney disease increases the susceptibility to negative effects of low and high potassium intake. Nephrol Dial Transplant 2024; 39:795-807. [PMID: 37813819 PMCID: PMC11045281 DOI: 10.1093/ndt/gfad220] [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: 07/09/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Dietary potassium (K+) has emerged as a modifiable factor for cardiovascular and kidney health in the general population, but its role in people with chronic kidney disease (CKD) is unclear. Here, we hypothesize that CKD increases the susceptibility to the negative effects of low and high K+ diets. METHODS We compared the effects of low, normal and high KChloride (KCl) diets and a high KCitrate diet for 4 weeks in male rats with normal kidney function and in male rats with CKD using the 5/6th nephrectomy model (5/6Nx). RESULTS Compared with rats with normal kidney function, 5/6Nx rats on the low KCl diet developed more severe extracellular and intracellular K+ depletion and more severe kidney injury, characterized by nephromegaly, infiltration of T cells and macrophages, decreased estimated glomerular filtration rate and increased albuminuria. The high KCl diet caused hyperkalemia, hyperaldosteronism, hyperchloremic metabolic acidosis and severe hypertension in 5/6Nx but not in sham rats. The high KCitrate diet caused hypochloremic metabolic alkalosis but attenuated hypertension despite higher abundance of the phosphorylated sodium chloride cotransporter (pNCC) and similar levels of plasma aldosterone and epithelial sodium channel abundance. All 5/6Nx groups had more collagen deposition than the sham groups and this effect was most pronounced in the high KCitrate group. Plasma aldosterone correlated strongly with kidney collagen deposition. CONCLUSIONS CKD increases the susceptibility to negative effects of low and high K+ diets in male rats, although the injury patterns are different. The low K+ diet caused inflammation, nephromegaly and kidney function decline, whereas the high K+ diet caused hypertension, hyperaldosteronism and kidney fibrosis. High KCitrate attenuated the hypertensive but not the pro-fibrotic effect of high KCl, which may be attributable to K+-induced aldosterone secretion. Our data suggest that especially in people with CKD it is important to identify the optimal threshold of dietary K+ intake.
Collapse
Affiliation(s)
- Martin Gritter
- Department of Internel Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Kuang-Yu Wei
- Department of Internel Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Rosa D Wouda
- Department of Internal Medicine, Section of Nephrology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Usha M Musterd-Bhaggoe
- Department of Internel Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kyra L Dijkstra
- Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesper Kers
- Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Van ‘t Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Martin H de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander H J Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Department of Internel Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
35
|
Tan ZY, Lou Y, Qin YC, Lin W, Liang BB, Sooranna SR, Ma YL, Zhou SF. Novel kinase 1 regulates CD8+T cells as a potential therapeutic mechanism for idiopathic pulmonary fibrosis. Int J Med Sci 2024; 21:1079-1090. [PMID: 38774751 PMCID: PMC11103402 DOI: 10.7150/ijms.93510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare, chronic and progressively worsening lung disease that poses a significant threat to patient prognosis, with a mortality rate exceeding that of some common malignancies. Effective methods for early diagnosis and treatment remain for this condition are elusive. In our study, we used the GEO database to access second-generation sequencing data and associated clinical information from IPF patients. By utilizing bioinformatics techniques, we identified crucial disease-related genes and their biological functions, and characterized their expression patterns. Furthermore, we mapped out the immune landscape of IPF, which revealed potential roles for novel kinase 1 and CD8+T cells in disease progression and outcome. These findings can aid the development of new strategies for the clinical diagnosis and treatment of IPF.
Collapse
Affiliation(s)
- Zhen-Yuan Tan
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuan Lou
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530021, China
| | - Yu-Cui Qin
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Wei Lin
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bin-Bin Liang
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Suren R. Sooranna
- Department of Metabolism, Digestion and Reproduction Faculty of Medicine Imperial College London Chelsea & Westminster Hospital, London SW10 9NH, UK
- Life Science and Clinical Research Center, Youjiang Medical University for Nationalities,18 Zhongshan Road II, Baise 533000, Guangxi, China
| | - Yi-Li Ma
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Su-Fang Zhou
- School of Basic Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
36
|
Tsokos GC, Boulougoura A, Kasinath V, Endo Y, Abdi R, Li H. The immunoregulatory roles of non-haematopoietic cells in the kidney. Nat Rev Nephrol 2024; 20:206-217. [PMID: 37985868 PMCID: PMC11005998 DOI: 10.1038/s41581-023-00786-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
The deposition of immune complexes, activation of complement and infiltration of the kidney by cells of the adaptive and innate immune systems have long been considered responsible for the induction of kidney damage in autoimmune, alloimmune and other inflammatory kidney diseases. However, emerging findings have highlighted the contribution of resident immune cells and of immune molecules expressed by kidney-resident parenchymal cells to disease processes. Several types of kidney parenchymal cells seem to express a variety of immune molecules with a distinct topographic distribution, which may reflect the exposure of these cells to different pathogenic threats or microenvironments. A growing body of literature suggests that these cells can stimulate the infiltration of immune cells that provide protection against infections or contribute to inflammation - a process that is also regulated by draining kidney lymph nodes. Moreover, components of the immune system, such as autoantibodies, cytokines and immune cells, can influence the metabolic profile of kidney parenchymal cells in the kidney, highlighting the importance of crosstalk in pathogenic processes. The development of targeted nanomedicine approaches that modulate the immune response or control inflammation and damage directly within the kidney has the potential to eliminate the need for systemically acting drugs.
Collapse
Affiliation(s)
- George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | | | - Vivek Kasinath
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Reza Abdi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
37
|
Chew C, Brand OJ, Yamamura T, Lawless C, Morais MRPT, Zeef L, Lin IH, Howell G, Lui S, Lausecker F, Jagger C, Shaw TN, Krishnan S, McClure FA, Bridgeman H, Wemyss K, Konkel JE, Hussell T, Lennon R. Kidney resident macrophages have distinct subsets and multifunctional roles. Matrix Biol 2024; 127:23-37. [PMID: 38331051 DOI: 10.1016/j.matbio.2024.02.002] [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: 06/15/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND The kidney contains distinct glomerular and tubulointerstitial compartments with diverse cell types and extracellular matrix components. The role of immune cells in glomerular environment is crucial for dampening inflammation and maintaining homeostasis. Macrophages are innate immune cells that are influenced by their tissue microenvironment. However, the multifunctional role of kidney macrophages remains unclear. METHODS Flow and imaging cytometry were used to determine the relative expression of CD81 and CX3CR1 (C-X3-C motif chemokine receptor 1) in kidney macrophages. Monocyte replenishment was assessed in Cx3cr1CreER X R26-yfp-reporter and shielded chimeric mice. Bulk RNA-sequencing and mass spectrometry-based proteomics were performed on isolated kidney macrophages from wild type and Col4a5-/- (Alport) mice. RNAscope was used to visualize transcripts and macrophage purity in bulk RNA assessed by CIBERSORTx analyses. RESULTS In wild type mice we identified three distinct kidney macrophage subsets using CD81 and CX3CR1 and these subsets showed dependence on monocyte replenishment. In addition to their immune function, bulk RNA-sequencing of macrophages showed enrichment of biological processes associated with extracellular matrix. Proteomics identified collagen IV and laminins in kidney macrophages from wild type mice whilst other extracellular matrix proteins including cathepsins, ANXA2 and LAMP2 were enriched in Col4a5-/- (Alport) mice. A subset of kidney macrophages co-expressed matrix and macrophage transcripts. CONCLUSIONS We identified CD81 and CX3CR1 positive kidney macrophage subsets with distinct dependence for monocyte replenishment. Multiomic analysis demonstrated that these cells have diverse functions that underscore the importance of macrophages in kidney health and disease.
Collapse
Affiliation(s)
- Christine Chew
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom; Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom
| | - Oliver J Brand
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Tomohiko Yamamura
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom
| | - Craig Lawless
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom
| | - Mychel Raony Paiva Teixeira Morais
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom
| | - Leo Zeef
- Bioinformatics Core Facility, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - I-Hsuan Lin
- Bioinformatics Core Facility, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Gareth Howell
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Sylvia Lui
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom
| | - Christopher Jagger
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Tovah N Shaw
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh EH9 3FL, United Kingdom
| | - Siddharth Krishnan
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Flora A McClure
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Hayley Bridgeman
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Kelly Wemyss
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Joanne E Konkel
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Tracy Hussell
- Lydia Becker Institute for Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, United Kingdom; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, United Kingdom.
| |
Collapse
|
38
|
Rao SN, Zahm M, Casemayou A, Buleon M, Faguer S, Feuillet G, Iacovoni JS, Joffre OP, Gonzalez-Fuentes I, Lhuillier E, Martins F, Riant E, Zakaroff-Girard A, Schanstra JP, Saulnier-Blache JS, Belliere J. Single-cell RNA sequencing identifies senescence as therapeutic target in rhabdomyolysis-induced acute kidney injury. Nephrol Dial Transplant 2024; 39:496-509. [PMID: 37697719 DOI: 10.1093/ndt/gfad199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND The role of macrophages in the development of rhabdomyolysis-induced acute kidney injury (RM-AKI) has been established, but an in-depth understanding of the changes in the immune landscape could help to improve targeted strategies. Whereas senescence is usually associated with chronic kidney processes, we also wished to explore whether senescence could also occur in AKI and whether senolytics could act on immune cells. METHODS Single-cell RNA sequencing was used in the murine glycerol-induced RM-AKI model to dissect the transcriptomic characteristics of CD45+ live cells sorted from kidneys 2 days after injury. Public datasets from murine AKI models were reanalysed to explore cellular senescence signature in tubular epithelial cells (TECs). A combination of senolytics (dasatinib and quercetin, DQ) was administered to mice exposed or not to RM-AKI. RESULTS Unsupervised clustering of nearly 17 000 single-cell transcriptomes identified seven known immune cell clusters. Sub-clustering of the mononuclear phagocyte cells revealed nine distinct cell sub-populations differently modified with RM. One macrophage cluster was particularly interesting since it behaved as a critical node in a trajectory connecting one major histocompatibility complex class IIhigh (MHCIIhigh) cluster only present in Control to two MHCIIlow clusters only present in RM-AKI. This critical cluster expressed a senescence gene signature, that was very different from that of the TECs. Senolytic DQ treatment blocked the switch from a F4/80highCD11blow to F4/80lowCD11bhigh phenotype, which correlated with prolonged nephroprotection in RM-AKI. CONCLUSIONS Single-cell RNA sequencing unmasked novel transitional macrophage subpopulation associated with RM-AKI characterized by the activation of cellular senescence processes. This work provides a proof-of-concept that senolytics nephroprotective effects may rely, at least in part, on subtle immune modulation.
Collapse
Affiliation(s)
- Snigdha N Rao
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Margot Zahm
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291-CNRS UMR5051-Université Paul Sabatier (UPS), Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Marie Buleon
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
- Department of Nephrology and Organ Transplantation, Referral Centre for Rare Kidney Diseases, French Intensive Care Renal Network, University Hospital of Toulouse, Toulouse, France
| | - Guylène Feuillet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jason S Iacovoni
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Olivier P Joffre
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291-CNRS UMR5051-Université Paul Sabatier (UPS), Toulouse, France
| | - Ignacio Gonzalez-Fuentes
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Emeline Lhuillier
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Frédéric Martins
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Elodie Riant
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Alexia Zakaroff-Girard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jean Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Julie Belliere
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
- Department of Nephrology and Organ Transplantation, Referral Centre for Rare Kidney Diseases, French Intensive Care Renal Network, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
39
|
Liu X, Li X, Chen Y, Liu X, Liu Y, Wei H, Li N. Systemic immune-inflammation Index is associated with chronic kidney disease in the U.S. population: insights from NHANES 2007-2018. Front Immunol 2024; 15:1331610. [PMID: 38449859 PMCID: PMC10915063 DOI: 10.3389/fimmu.2024.1331610] [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: 11/01/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Objectives The systemic immune-inflammation index (SII), a novel and systematic inflammatory biomarker that is associated with chronic kidney disease (CKD), has not received much attention. This study aimed to investigate the relationship between SII and CKD in the United States (U.S.) population. Methods Our study ultimately included a nationally representative sample of 10,787 adults who participated in the 2007-2018 National Health and Nutrition Examination Survey. Weighted multivariate logistic regression was used to assess the correlation between SII and CKD, and a restricted cubic spline (RCS) model was subsequently used to explore the non-linear relationship between SII and CKD. Subgroup analyses were performed to further the effects of other covariates on the relationship between SII and CKD. Results Following confounder adjustment, a higher SII was related to the incidence of CKD (OR =1.36; 95% CI, 1.07-1.73; p =0.01), as validated by multivariable logistic regression. The RCS curve revealed a non-linear positive correlation between SII/1000 and CKD incidence (p for non-linear =0.0206). Additionally, subgroup analysis confirmed a stronger correlation for male participants (OR =2.628; 95% CI, 1.829-3.776) than for female participants (OR =1.733; 95% CI, 1.379-2.178) (p for interaction =0.046). Conclusions SII is positively associated with the incidence of CKD among U.S. adults, especially in males. However, further studies are needed to confirm our findings and explore the causal factors that can contribute to the prevention and treatment of CKD.
Collapse
Affiliation(s)
- Xiaoxin Liu
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinyu Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yulin Chen
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyu Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanyan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haotian Wei
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ningxu Li
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
40
|
Zhao Y, Nicholson L, Wang H, Qian YW, Hawthorne WJ, Jimenez-Vera E, Gloss BS, Lai J, Thomas A, Chew YV, Burns H, Zhang GY, Wang YM, Rogers NM, Zheng G, Yi S, Alexander SI, O’Connell PJ, Hu M. Intragraft memory-like CD127hiCD4+Foxp3+ Tregs maintain transplant tolerance. JCI Insight 2024; 9:e169119. [PMID: 38516885 PMCID: PMC11063946 DOI: 10.1172/jci.insight.169119] [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: 02/14/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) play an essential role in suppressing transplant rejection, but their role within the graft and heterogeneity in tolerance are poorly understood. Here, we compared phenotypic and transcriptomic characteristics of Treg populations within lymphoid organs and grafts in an islet xenotransplant model of tolerance. We showed Tregs were essential for tolerance induction and maintenance. Tregs demonstrated heterogeneity within the graft and lymphoid organs of tolerant mice. A subpopulation of CD127hi Tregs with memory features were found in lymphoid organs, presented in high proportions within long-surviving islet grafts, and had a transcriptomic and phenotypic profile similar to tissue Tregs. Importantly, these memory-like CD127hi Tregs were better able to prevent rejection by effector T cells, after adoptive transfer into secondary Rag-/- hosts, than naive Tregs or unselected Tregs from tolerant mice. Administration of IL-7 to the CD127hi Treg subset was associated with a strong activation of phosphorylation of STAT5. We proposed that memory-like CD127hi Tregs developed within the draining lymph node and underwent further genetic reprogramming within the graft toward a phenotype that had shared characteristics with other tissue or tumor Tregs. These findings suggested that engineering Tregs with these characteristics either in vivo or for adoptive transfer could enhance transplant tolerance.
Collapse
Affiliation(s)
| | | | - Hannah Wang
- Centre for Transplant and Renal Research and
| | - Yi Wen Qian
- Centre for Transplant and Renal Research and
| | | | | | - Brian S. Gloss
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Joey Lai
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | | | - Yi Vee Chew
- Centre for Transplant and Renal Research and
| | | | - Geoff Y. Zhang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research and
- Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | | | - Shounan Yi
- Centre for Transplant and Renal Research and
| | - Stephen I. Alexander
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | | | - Min Hu
- Centre for Transplant and Renal Research and
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
41
|
Bai F, Han L, Yang J, Liu Y, Li X, Wang Y, Jiang R, Zeng Z, Gao Y, Zhang H. Integrated analysis reveals crosstalk between pyroptosis and immune regulation in renal fibrosis. Front Immunol 2024; 15:1247382. [PMID: 38343546 PMCID: PMC10853448 DOI: 10.3389/fimmu.2024.1247382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/09/2024] [Indexed: 02/15/2024] Open
Abstract
PURPOSE The pathogenesis of renal fibrosis (RF) involves intricate interactions between profibrotic processes and immune responses. This study aimed to explore the potential involvement of the pyroptosis signaling pathway in immune microenvironment regulation within the context of RF. Through comprehensive bioinformatics analysis and experimental validation, we investigated the influence of pyroptosis on the immune landscape in RF. METHODS We obtained RNA-seq datasets from Gene Expression Omnibus (GEO) databases and identified Pyroptosis-Associated Regulators (PARs) through literature reviews. Systematic evaluation of alterations in 27 PARs was performed in RF and normal kidney samples, followed by relevant functional analyses. Unsupervised cluster analysis revealed distinct pyroptosis modification patterns. Using single-sample gene set enrichment analysis (ssGSEA), we examined the correlation between pyroptosis and immune infiltration. Hub regulators were identified via weighted gene coexpression network analysis (WGCNA) and further validated in a single-cell RNA-seq dataset. We also established a unilateral ureteral obstruction-induced RF mouse model to verify the expression of key regulators at the mRNA and protein levels. RESULTS Our comprehensive analysis revealed altered expression of 19 PARs in RF samples compared to normal samples. Five hub regulators, namely PYCARD, CASP1, AIM2, NOD2, and CASP9, exhibited potential as biomarkers for RF. Based on these regulators, a classifier capable of distinguishing normal samples from RF samples was developed. Furthermore, we identified correlations between immune features and PARs expression, with PYCARD positively associated with regulatory T cells abundance in fibrotic tissues. Unsupervised clustering of RF samples yielded two distinct subtypes (Subtype A and Subtype B), with Subtype B characterized by active immune responses against RF. Subsequent WGCNA analysis identified PYCARD, CASP1, and NOD2 as hub PARs in the pyroptosis modification patterns. Single-cell level validation confirmed PYCARD expression in myofibroblasts, implicating its significance in the stress response of myofibroblasts to injury. In vivo experimental validation further demonstrated elevated PYCARD expression in RF, accompanied by infiltration of Foxp3+ regulatory T cells. CONCLUSIONS Our findings suggest that pyroptosis plays a pivotal role in orchestrating the immune microenvironment of RF. This study provides valuable insights into the pathogenesis of RF and highlights potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Fengxia Bai
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Longchao Han
- Department of Gastrointestinal Oncology, Affiliated Xingtai People's Hospital of Hebei Medical University, Xingtai, China
| | - Jifeng Yang
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Yuxiu Liu
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiangmeng Li
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Yaqin Wang
- Department of Critical Care Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ruijian Jiang
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhaomu Zeng
- Department of Neurosurgery, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yan Gao
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| | - Haisong Zhang
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
42
|
Ma K, Luo L, Yang M, Meng Y. The suppression of sepsis-induced kidney injury via the knockout of T lymphocytes. Heliyon 2024; 10:e23311. [PMID: 38283245 PMCID: PMC10818183 DOI: 10.1016/j.heliyon.2023.e23311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 01/30/2024] Open
Abstract
Patients with sepsis always have a high mortality rate, and acute kidney injury (AKI) is the main cause of death. It seems obvious that the immune response is involved in this process, but the specific mechanism is unknown, especially the pathogenic role of T cells and B cells needs to be further clarified. Acute kidney injury models induced by lipopolysaccharide were established using T-cell, B-cell, and T&B cell knockout mice to elucidate the role of immune cells in sepsis. Flow cytometry was used to validate the mouse models, and the pathology can confirm renal tubular injury. LPS-induced sepsis caused significant renal pathological damage, Second-generation gene sequencing showed T cells-associated pathway was enriched in sepsis. The renal tubular injury was significantly reduced in T cell and T&B cell knockout mice (BALB/c-nu, Rag1-/-), especially in BALB/c-nu mice, with a decrease in the secretion of inflammatory cytokines in the renal tissue after LPS injection. LPS injection did not produce the same effect after the knockout of B cells. We found that blocking T cells could alleviate inflammation and renal injury caused by sepsis, providing a promising strategy for controlling renal injury.
Collapse
Affiliation(s)
- Ke Ma
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Liang Luo
- The Biomedical Translational Research Institute, Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University), Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Meixiang Yang
- The Biomedical Translational Research Institute, Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University), Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Yu Meng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
- Department of Nephrology, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Heyuan, 517000, China
| |
Collapse
|
43
|
Yamashita N, Kramann R. Mechanisms of kidney fibrosis and routes towards therapy. Trends Endocrinol Metab 2024; 35:31-48. [PMID: 37775469 DOI: 10.1016/j.tem.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023]
Abstract
Kidney fibrosis is the final common pathway of virtually all chronic kidney diseases (CKDs) and is therefore considered to be a promising therapeutic target for these conditions. However, despite great progress in recent years, no targeted antifibrotic therapies for the kidney have been approved, likely because the complex mechanisms that initiate and drive fibrosis are not yet completely understood. Recent single-cell genomic approaches have allowed novel insights into kidney fibrosis mechanisms in mouse and human, particularly the heterogeneity and differentiation processes of myofibroblasts, the role of injured epithelial cells and immune cells, and their crosstalk mechanisms. In this review we summarize the key mechanisms that drive kidney fibrosis, including recent advances in understanding the mechanisms, as well as potential routes for developing novel targeted antifibrotic therapeutics.
Collapse
Affiliation(s)
- Noriyuki Yamashita
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
44
|
Di X, Chen J, Li Y, Wang M, Wei J, Li T, Liao B, Luo D. Crosstalk between fibroblasts and immunocytes in fibrosis: From molecular mechanisms to clinical trials. Clin Transl Med 2024; 14:e1545. [PMID: 38264932 PMCID: PMC10807359 DOI: 10.1002/ctm2.1545] [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: 07/17/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND The impact of fibroblasts on the immune system provides insight into the function of fibroblasts. In various tissue microenvironments, multiple fibroblast subtypes interact with immunocytes by secreting growth factors, cytokines, and chemokines, leading to wound healing, fibrosis, and escape of cancer immune surveillance. However, the specific mechanisms involved in the fibroblast-immunocyte interaction network have not yet been fully elucidated. MAIN BODY AND CONCLUSION Therefore, we systematically reviewed the molecular mechanisms of fibroblast-immunocyte interactions in fibrosis, from the history of cellular evolution and cell subtype divisions to the regulatory networks between fibroblasts and immunocytes. We also discuss how these communications function in different tissue and organ statuses, as well as potential therapies targeting the reciprocal fibroblast-immunocyte interplay in fibrosis. A comprehensive understanding of these functional cells under pathophysiological conditions and the mechanisms by which they communicate may lead to the development of effective and specific therapies targeting fibrosis.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jiawei Chen
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Menghua Wang
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Deyi Luo
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
45
|
Luo L, Wang S, Hu Y, Wang L, Jiang X, Zhang J, Liu X, Guo X, Luo Z, Zhu C, Xie M, Li Y, You J, Yang F. Precisely Regulating M2 Subtype Macrophages for Renal Fibrosis Resolution. ACS NANO 2023; 17:22508-22526. [PMID: 37948096 DOI: 10.1021/acsnano.3c05998] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Macrophages are central to the pathogenesis of kidney disease and serve as an effective therapeutic target for kidney injury and fibrosis. Among them, M2-type macrophages have double-edged effects regarding anti-inflammatory effects and tissue repair. Depending on the polarization of the M2 subtypes (M2a or M2c) in the diseased microenvironment, they can either mediate normal tissue repair or drive tissue fibrosis. In renal fibrosis, M2a promotes disease progression through macrophage-to-myofibroblast transition (MMT) cells, while M2c possesses potent anti-inflammatory functions and promotes tissue repair, and is inhibited. The mechanisms underlying this differentiation are complex and are currently not well understood. Therefore, in this study, we first confirmed that M2a-derived MMT cells are responsible for the development of renal fibrosis and demonstrated that the intensity of TGF-β signaling is a major factor determining the differential polarization of M2a and M2c. Under excessive TGF-β stimulation, M2a undergoes a process known as MMT cells, whereas moderate TGF-β stimulation favors the polarization of M2c phenotype macrophages. Based on these findings, we employed targeted nanotechnology to codeliver endoplasmic reticulum stress (ERS) inhibitor (Ceapin 7, Cea or C) and conventional glucocorticoids (Dexamethasone, Dex or D), precisely modulating the ATF6/TGF-β/Smad3 signaling axis within macrophages. This approach calibrated the level of TGF-β stimulation on macrophages, promoting their polarization toward the M2c phenotype and suppressing excessive MMT polarization. The study indicates that the combination of ERS inhibitor and a first-line anti-inflammatory drug holds promise as an effective therapeutic approach for renal fibrosis resolution.
Collapse
Affiliation(s)
- Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Hangzhou 310058, Zhejiang, China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Yilong Hu
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Litong Wang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Xindong Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Chunqi Zhu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Miaomiao Xie
- The Second Affiliated Hospital of Shenzhen University, 118 Longjinger Road, Baoan District, Shenzhen 518101, Guangdong, China
| | - Yeqing Li
- The People's Hospital of Baoan Shenzhen, 118 Longjinger Road, Baoan District, Shenzhen 518101, Guangdong, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou 310058, Zhejiang, China
| | - Fuchun Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| |
Collapse
|
46
|
Gharaie S, Lee K, Noller K, Lo EK, Miller B, Jung HJ, Newman-Rivera AM, Kurzhagen JT, Singla N, Welling PA, Fan J, Cahan P, Noel S, Rabb H. Single cell and spatial transcriptomics analysis of kidney double negative T lymphocytes in normal and ischemic mouse kidneys. Sci Rep 2023; 13:20888. [PMID: 38017015 PMCID: PMC10684868 DOI: 10.1038/s41598-023-48213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
T cells are important in the pathogenesis of acute kidney injury (AKI), and TCR+CD4-CD8- (double negative-DN) are T cells that have regulatory properties. However, there is limited information on DN T cells compared to traditional CD4+ and CD8+ cells. To elucidate the molecular signature and spatial dynamics of DN T cells during AKI, we performed single-cell RNA sequencing (scRNA-seq) on sorted murine DN, CD4+, and CD8+ cells combined with spatial transcriptomic profiling of normal and post AKI mouse kidneys. scRNA-seq revealed distinct transcriptional profiles for DN, CD4+, and CD8+ T cells of mouse kidneys with enrichment of Kcnq5, Klrb1c, Fcer1g, and Klre1 expression in DN T cells compared to CD4+ and CD8+ T cells in normal kidney tissue. We validated the expression of these four genes in mouse kidney DN, CD4+ and CD8+ T cells using RT-PCR and Kcnq5, Klrb1, and Fcer1g genes with the NIH human kidney precision medicine project (KPMP). Spatial transcriptomics in normal and ischemic mouse kidney tissue showed a localized cluster of T cells in the outer medulla expressing DN T cell genes including Fcer1g. These results provide a template for future studies in DN T as well as CD4+ and CD8+ cells in normal and diseased kidneys.
Collapse
Affiliation(s)
- Sepideh Gharaie
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kyungho Lee
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kathleen Noller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Emily K Lo
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Brendan Miller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Hyun Jun Jung
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Nirmish Singla
- Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
47
|
Dong Z, Chen F, Peng S, Liu X, Liu X, Guo L, Wang E, Chen X. Identification of the key immune-related genes and immune cell infiltration changes in renal interstitial fibrosis. Front Endocrinol (Lausanne) 2023; 14:1207444. [PMID: 38027143 PMCID: PMC10663291 DOI: 10.3389/fendo.2023.1207444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Chronic kidney disease (CKD) is the third-leading cause of premature mortality worldwide. It is characterized by rapid deterioration due to renal interstitial fibrosis (RIF) via excessive inflammatory infiltration. The aim of this study was to discover key immune-related genes (IRGs) to provide valuable insights and therapeutic targets for RIF in CKD. Materials and methods We screened differentially expressed genes (DEGs) between RIF samples from CKD patients and healthy controls from a public database. Least absolute shrinkage and selection operator regression analysis and receiver operating characteristic curve analysis were applied to identify significant key biomarkers. The single-sample Gene Set Enrichment Analysis (ssGSEA) algorithm was used to analyze the infiltration of immune cells between the RIF and control samples. The correlation between biomarkers and immune cell composition was assessed. Results A total of 928 DEGs between CKD and control samples from six microarray datasets were found, 17 overlapping immune-correlated DEGs were identified by integration with the ImmPort database, and six IRGs were finally identified in the model: apolipoprotein H (APOH), epidermal growth factor (EGF), lactotransferrin (LTF), lysozyme (LYZ), phospholipid transfer protein (PLTP), and secretory leukocyte peptidase inhibitor (SLPI). Two additional datasets and in vivo experiments indicated that the expression levels of APOH and EGF in the fibrosis group were significantly lower than those in the control group, while the expression levels of LTF, LYZ, PLTP, and SLPI were higher (all P < 0.05). These IRGs also showed a significant correlation with renal function impairment. Moreover, four upregulated IRGs were positively associated with various T cell populations, which were enriched in RIF tissues, whereas two downregulated IRGs had opposite results. Several signaling pathways, such as the "T cell receptor signaling pathway" and "positive regulation of NF-κB signaling pathway", were discovered to be associated not only with immune cell infiltration, but also with the expression levels of six IRGs. Conclusion In summary, six IRGs were identified as key biomarkers for RIF, and exhibited a strong correlation with various T cells and with the NF-κB signaling pathway. All these IRGs and their signaling pathways may evolve as valuable therapeutic targets for RIF in CKD.
Collapse
Affiliation(s)
- Zhitao Dong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Peng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiongfei Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingyang Liu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Lao P, Chen J, Tang L, Zhang J, Chen Y, Fang Y, Fan X. Regulatory T cells in lung disease and transplantation. Biosci Rep 2023; 43:BSR20231331. [PMID: 37795866 PMCID: PMC10611924 DOI: 10.1042/bsr20231331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/06/2023] Open
Abstract
Pulmonary disease can refer to the disease of the lung itself or the pulmonary manifestations of systemic diseases, which are often connected to the malfunction of the immune system. Regulatory T (Treg) cells have been shown to be important in maintaining immune homeostasis and preventing inflammatory damage, including lung diseases. Given the increasing amount of evidence linking Treg cells to various pulmonary conditions, Treg cells might serve as a therapeutic strategy for the treatment of lung diseases and potentially promote lung transplant tolerance. The most potent and well-defined Treg cells are Foxp3-expressing CD4+ Treg cells, which contribute to the prevention of autoimmune lung diseases and the promotion of lung transplant rejection. The protective mechanisms of Treg cells in lung disease and transplantation involve multiple immune suppression mechanisms. This review summarizes the development, phenotype and function of CD4+Foxp3+ Treg cells. Then, we focus on the therapeutic potential of Treg cells in preventing lung disease and limiting lung transplant rejection. Furthermore, we discussed the possibility of Treg cell utilization in clinical applications. This will provide an overview of current research advances in Treg cells and their relevant application in clinics.
Collapse
Affiliation(s)
- Peizhen Lao
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jingyi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Longqian Tang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jiwen Zhang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuxi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuyin Fang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Xingliang Fan
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| |
Collapse
|
49
|
Kim TM, Lee KW, Kim HD, Hong SO, Cho HJ, Yang JH, Kim SJ, Park JB. Evaluation of Selected Markers in Kidneys of Cynomolgus Monkey ( Macaca fascicularis) with Induced Diabetes during Renal Ischemia-reperfusion Injury. Comp Med 2023; 73:357-372. [PMID: 38087409 PMCID: PMC10702167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/26/2023] [Accepted: 05/04/2023] [Indexed: 12/18/2023]
Abstract
We previously reported that induced type 1 diabetes mellitus (DM) increases the susceptibility of acute kidney injury in- duced by ischemia-reperfusion injury (IRI) in cynomolgus monkeys. In this follow-up study, we compared the expression of selected markers in the renal tissues of monkeys subjected to bilateral renal IRI with and without diabetes. All tissues were obtained from the original study. Renal biopsies were obtained before and 24 and 48 h after ischemia and were examined for expression of KI-67 (tubular proliferation), Na+ /K+ ATPase (sodium-potassium pump), TNF-α(tumor necrosis factor-α, inflammation), CD31 (microvessels), CD3 (T-cells), 2 fibrotic markers (fibroblast specific protein-1, FSP-1;α-smooth muscle actin,α -SMA), and cleaved caspase 3 (apoptosis). Generally, the expression of these markers differed in monkeys with and without DM. As compared with non-DM monkeys, DM monkeys had more cells that expressed KI-67 during progression of acute kidney injury (AKI). Na+ /K+ ATPase expression was clearly present at baseline in the basolateral tubular areas only in the non-DM monkeys. At 48 h, its expression in the basolateral area was not visible in DM monkeys, but was still present in intercellular junctions of non-DM monkeys. The expression of TNF-αwas higher in DM before and 48 h after ischemia. Before and 24 h after ischemia, the number of CD31-positive capillaries was not different between 2 groups, although more collapsed vessels were found at in DM at 24 h. At 48 h, the number of capillaries was less in DM compared with those from non-DM animals. DM monkeys had more interstitial CD3-positive cells than did non-DM monkeys at 24 and 48 h after ischemia. Finally, FSP-1-stained cells were more abundant in DM than non-DM at 24 and 48 h. Our results show that DM aggravates the recovery of renal ischemia/reperfusion injury by affecting tubular proliferation, capillary density, T cell infil- tration and by altering protein and mRNA expression of various genes involved in ion channel, inflammation, and fibrotic change. The results from this observational study demonstrate that DM aggravates the recovery of renal ischemia/reperfusion injury by affecting multiple events including tubular necrosis, proliferation, function, inflammation and by inducing capillary rarefaction in cynomolgus monkeys.
Collapse
Affiliation(s)
- Tae M Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea
| | - Kyo W Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea
| | - Hong D Kim
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea
| | - Sung O Hong
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea
| | - Hye J Cho
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea
| | - Je H Yang
- Laboratory Animal Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea
| | - Sung J Kim
- GenNBio Inc., 80 Deurimsandan 2-ro, Cheongbuk-myeon, Pyeongtaek-si, Gyeonggi-do 17796, South Korea
| | - Jae B Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea
| |
Collapse
|
50
|
Jeyamogan S, Leventhal JR, Mathew JM, Zhang ZJ. CD4 +CD25 +FOXP3 + regulatory T cells: a potential "armor" to shield "transplanted allografts" in the war against ischemia reperfusion injury. Front Immunol 2023; 14:1270300. [PMID: 37868962 PMCID: PMC10587564 DOI: 10.3389/fimmu.2023.1270300] [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: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Despite the advances in therapeutic interventions, solid organ transplantation (SOT) remains the "gold standard" treatment for patients with end-stage organ failure. Recently, vascularized composite allotransplantation (VCA) has reemerged as a feasible treatment option for patients with complex composite tissue defects. In both SOT and VCA, ischemia reperfusion injury (IRI) is inevitable and is a predominant factor that can adversely affect transplant outcome by potentiating early graft dysfunction and/or graft rejection. Restoration of oxygenated blood supply to an organ which was previously hypoxic or ischemic for a period of time triggers cellular oxidative stress, production of both, pro-inflammatory cytokines and chemokines, infiltration of innate immune cells and amplifies adaptive alloimmune responses in the affected allograft. Currently, Food and Drug Administration (FDA) approved drugs for the treatment of IRI are unavailable, therefore an efficacious therapeutic modality to prevent, reduce and/or alleviate allograft damages caused by IRI induced inflammation is warranted to achieve the best-possible transplant outcome among recipients. The tolerogenic capacity of CD4+CD25+FOXP3+ regulatory T cells (Tregs), have been extensively studied in the context of transplant rejection, autoimmunity, and cancer. It was not until recently that Tregs have been recognized as a potential cell therapeutic candidate to be exploited for the prevention and/or treatment of IRI, owing to their immunomodulatory potential. Tregs can mitigate cellular oxidative stress, produce anti-inflammatory cytokines, promote wound healing, and tissue repair and prevent the infiltration of pro-inflammatory immune cells in injured tissues. By using strategic approaches to increase the number of Tregs and to promote targeted delivery, the outcome of SOT and VCA can be improved. This review focuses on two sections: (a) the therapeutic potential of Tregs in preventing and mitigating IRI in the context of SOT and VCA and (b) novel strategies on how Tregs could be utilized for the prevention and/or treatment of IRI.
Collapse
Affiliation(s)
- Shareni Jeyamogan
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Leventhal
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - James M. Mathew
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Zheng Jenny Zhang
- Department of Surgery, Comprehensive Transplant Center Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Microsurgery and Pre-Clinical Research Core, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|