1
|
Xiong Y, Li W, Jin S, Wan S, Wu S. Inflammation in glomerular diseases. Front Immunol 2025; 16:1526285. [PMID: 40103820 PMCID: PMC11913671 DOI: 10.3389/fimmu.2025.1526285] [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: 11/11/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
The structural and functional integrity of glomerular cells is critical for maintaining normal kidney function. Glomerular diseases, which involve chronic histological damage to the kidney, are related to injury to glomerular cells such as endothelial cells, mesangial cells (MCs), and podocytes. When faced with pathogenic conditions, these cells release pro-inflammatory cytokines such as chemokines, inflammatory factors, and adhesion factors. These substances interact with glomerular cells through specific inflammatory pathways, resulting in damage to the structure and function of the glomeruli, ultimately causing glomerular disease. Although the role of inflammation in chronic kidney diseases is well known, the specific molecular pathways that result in glomerular diseases remain largely unclear. For a long time, it has been believed that only immune cells can secrete inflammatory factors. Therefore, targeted therapies against immune cells were considered the first choice for treating inflammation in glomerular disease. However, emerging research indicates that non-immune cells such as glomerular endothelial cells, MCs, and podocytes can also play a role in renal inflammation by releasing inflammatory factors. Similarly, targeted therapies against glomerular cells should be considered. This review aims to uncover glomerular diseases related to inflammation and pathways in glomerular inflammation, and for the first time summarized that non-immune cells in the glomerulus can participate in glomerular inflammatory damage by secreting inflammatory factors, providing valuable references for future strategies to prevent and treat glomerular diseases. More importantly, we emphasized targeted glomerular cell therapy, which may be a key direction for the future treatment of glomerular diseases.
Collapse
Affiliation(s)
- Yongqing Xiong
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Wei Li
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Songzhi Jin
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shujing Wan
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Suzhen Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
2
|
Wang Z, Gu Y, Qu Y, Huang X, Sun T, Wu W, Hu Q, Chen X, Li Y, Zhao H, Hu Y, Wu B, Xu J. Prevention of Intrauterine Adhesion with Platelet-Rich Plasma Double-Network Hydrogel. Adv Biol (Weinh) 2025; 9:e2400336. [PMID: 39673358 DOI: 10.1002/adbi.202400336] [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: 10/30/2024] [Indexed: 12/16/2024]
Abstract
Intrauterine adhesion (IUA) can negatively impact on pregnancy outcomes, leading to reduced pregnancy rates, secondary infertility, and an increased risk of pregnancy complications. Studies have shown that the application of platelet-rich plasma (PRP) in IUA patients is effective. However, the clinical readhesive rate of IUA after treatment is still high, especially in severe cases. Platelet-rich plasma double-network hydrogel (DN gel) is an engineered PRP double network hydrogel, which is a sodium alginate (SA) based PRP hydrogel with egg carton ion cross-linking and fibrin double network. The results of this study show that intrauterine injection of DN gel has a better effect on promoting endometrial regeneration and enhancing endometrial receptivity than PRP gel. The mechanism is analyzed through single-cell sequencing, which may be achieved by increasing the expression of neutrophils (Neut), natural killer cells (NK), and type I classical dendritic cells (cDC1) in the endometrium and inhibiting the infiltration of M2 macrophages. Overall, based on the good healing efficiency and good biocompatibility of DN gel, it is expected to become a method of treating IUA with better efficacy and faster clinical translation.
Collapse
Affiliation(s)
- Zhuomin Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ying Gu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiyuan Qu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xujia Huang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Tao Sun
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Department of Assisted Reproduction, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| | - Qianyu Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xiao Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yu Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Huafei Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Bingbing Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jian Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Department of Assisted Reproduction, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
3
|
Mangano K, Diamantopoulos A, Vallianou NG, Stratigou T, Panagopoulos F, Kounatidis D, Dalamaga M, Fagone P, Nicoletti F. Serum and urinary levels of MIF, CD74, DDT and CXCR4 among patients with type 1 diabetes mellitus, type 2 diabetes and healthy individuals: Implications for further research. Metabol Open 2024; 24:100320. [PMID: 39323959 PMCID: PMC11422569 DOI: 10.1016/j.metop.2024.100320] [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: 09/03/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) is a highly conserved cytokine with pleiotropic properties, mainly pro-inflammatory. MIF seems to exert its pro-inflammatory features by binding to its transmembrane cellular receptor CD74. MIF also has CXCR4, which acts as a co-receptor in this inflammatory process. Apart from MIF, D-dopachrome tautomerase (DDT) or MIF2, which belongs to the MIF superfamily, also binds to receptor CD74. Therefore, these molecules, MIF, CD74, DDT and CXCR4 are suggested to work together orchestrating an inflammatory process. Diabetes mellitus is characterised by chronic low-grade inflammation. Therefore, the aim of the present study was to evaluate serum and urinary levels of the aforementioned molecules among patients with type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM) and among healthy controls. Methods We enrolled 13 patients with T1DM, 74 patients with T2DM and 25 healthy individuals as controls. Levels of CD74, CXCR4, DDT, and MIF were measured using ELISA Kits according to the manufacturer's instructions. Results We documented increased serum MIF levels together with higher urinary CD74 levels among patients with T1DM, when compared to patients with T2DM and healthy adults. In particular, patients with T1DM showed significantly increased levels of MIF compared to T2DM (p = 0.011) and healthy controls (p = 0.0093). CD74 in urine were significantly higher in patients with T1DM compared to those affected with T2DM (p = 0.0302) and healthy group (p = 0.0099). On the contrary, serum CD74 were similar among the three groups. No statistical differences were identified in CXCR4 levels both in serum and in urine of all groups. Patients with T2DM and overweight/obesity had increased urinary levels of CD74, when compared to lean patients with T2DM. Conclusion The increased serum MIF levels and urinary CD74 levels among patients with T1DM may be attributed to the autoimmune milieu, which characterises patients with T1DM, when compared to patients with T2DM. These two findings merit further attention as they could pave the way for further research regarding the potential beneficial effects of inhibitors of MIF among patients with T1DM, especially in the early stages of T1DM. Finally, the role of inhibitors of MIF could be further explored in the context of obesity among patients with T2DM.
Collapse
Affiliation(s)
- Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Aristidis Diamantopoulos
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece
| | - Natalia G Vallianou
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece
| | - Fotis Panagopoulos
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece
| | - Dimitris Kounatidis
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
4
|
Gonzalez-Vicente A, Crawford DC, Bush WS, Wu Z, Bruggeman LA, Nair V, Eichinger F, Wessely O, Kretzler M, O'Toole JF, Sedor JR. Analysis of Glomerular Transcriptomes from Nephrotic Patients Suggest APOL1 Risk Variants Impact Parietal Epithelial Cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.05.24316766. [PMID: 39830251 PMCID: PMC11741451 DOI: 10.1101/2024.11.05.24316766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The disproportionate risk for idiopathic proteinuric podocytopathies in Black people is explained, in part, by the presence of two risk alleles (G1 or G2) in the APOL1 gene. The pathogenic mechanisms responsible for this genetic association remain incompletely understood. We analyzed glomerular RNASeq transcriptomes from patients with idiopathic nephrotic syndrome of which 72 had inferred African ancestry (AA) and 152 did not (noAA). Using gene coexpression networks we found a significant association between APOL1 risk allele number and the coexpression metamodule 2 (MM2), even after adjustment for eGFR and proteinuria at biopsy. Unadjusted Kaplan-Meier curves showed that unlike noAA, AA with the highest tertile of MM2 gene activation scores were less likely to achieve complete remission (p≤0.014). Characteristic direction (ChDir) identified a signature of 1481 genes, which separated patients with APOL1 risk alleles from those homozygous for reference APOL1 . Only in AA, the tertile with the highest activation scores of these 1481 genes was less likely to achieve complete remission (p≤0.022) and showed a trend to faster progression to the composite event of kidney failure or loss of 40% eGFR (p≤0.099). The MM2 and ChDir genes significantly overlapped and were both enriched for Epithelial Mesenchymal Transition and inflammation terms. Finally, MM2 significantly overlapped with a parietal epithelial cell (PEC)-identity gene signature but not with a podocyte identity signature. Podocytes expressing variant APOL1s may generate inflammatory signals that activate PECs by paracrine mechanisms contributing to APOL1 nephropathy.
Collapse
|
5
|
Ferhat M, Mayer J, Costa LH, Prendecki M, Tarazona AAP, Schinagl A, Kerschbaumer RJ, Tam FWK, Landlinger C, Thiele M. Targeting of oxidized Macrophage Migration Inhibitory Factor (oxMIF) with antibody ON104 attenuates the severity of glomerulonephritis. PLoS One 2024; 19:e0311837. [PMID: 39374239 PMCID: PMC11458038 DOI: 10.1371/journal.pone.0311837] [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: 05/24/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
The oxidized form of Macrophage Migration Inhibitory Factor (oxMIF) has been identified as the disease-related isoform of MIF, exerting pathological functions in inflamed tissue. In this study, we aimed to explore the in vivo effects of the neutralizing anti-oxMIF antibody ON104 in a rat model of crescentic glomerulonephritis (CGN), to better understand its disease modifying activities. WKY rats received a single intravenous injection of a rabbit nephrotoxic serum (NTS), targeting rat glomerular basement membrane to induce CGN. On day 4 and day 6, ON104 was given intraperitoneally (i.p.) and on day 8 urine, blood and kidney tissue were collected. ON104 substantially attenuated the severity of CGN demonstrated by reduced proteinuria, hematuria, as well as lower levels of kidney injury molecule (KIM)-1. ON104 treatment preserved the glomerular morphology and suppressed crescent formation, a hallmark of the disease. On the cellular level, oxMIF neutralization by ON104 strongly reduced the number of macrophages and neutrophils within the inflamed kidneys. In vitro, we identified human neutrophils, but not monocytes, as main producers of oxMIF among total peripheral cells. The present study demonstrates that oxMIF is a pertinent therapeutic target in a model of CGN which mechanistically resembles human immune mediated CGN. In this model, neutralization of oxMIF by ON104 leads to an improvement in both urinary abnormalities and histological pathological characteristics of the disease. ON104, thus has the potential to become a novel disease-modifying drug for the treatment of glomerulonephritis and other inflammatory kidney diseases.
Collapse
Affiliation(s)
- Maroua Ferhat
- OncoOne Research & Development GmbH, Vienna, Austria
| | - Julia Mayer
- OncoOne Research & Development GmbH, Vienna, Austria
| | - Lyndon H. Costa
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | - Maria Prendecki
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | | | | | | | - Frederick W. K. Tam
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | | | | |
Collapse
|
6
|
Zheng Y, Zhou Y, Zhu D, Fu X, Xie C, Sun S, Qin G, Feng M, Liu C, Zhou Q, Liu F, Chu C, Wang F, Yang D, Wang MW, Gui Y. Single-cell mapping of peripheral blood mononuclear cells reveals key transcriptomic changes favoring coronary artery lesion in IVIG-resistant Kawasaki disease. Heliyon 2024; 10:e37857. [PMID: 39323779 PMCID: PMC11422586 DOI: 10.1016/j.heliyon.2024.e37857] [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: 03/23/2024] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
Background Intravenous immunoglobulin (IVIG)-resistant Kawasaki disease (KD) poses a considerable challenge to patients and their families due to its severe complications. Previous researches have highlighted the critical role of immune disorders in its pathogenesis. However, fragmented studies based on isolated cases hinder a comprehensive understanding of this deadly illness. This study aimed to explore the overall landscape of peripheral blood mononuclear cells (PBMCs) in IVIG-resistant KD patients using single-cell RNA sequencing (scRNA-seq). Methods The scRNA-seq was used to characterize the transcriptomic profiles of IVIG-resistant KD patients, IVIG-responsive KD patients, and healthy controls. Data quality control (QC) and subsequent analysis were conducted using various R packages. These included DoubletFinder and Harmony for QC, Seurat and SingleR for identifying and annotating major cell types, ggpubr for calculating and visualizing the percentages of each cell type, Seurat for characterizing differentially expressed genes (DEGs) between groups, pheatmap for visualizing the DEGs, clusterProfiler for performing Gene Ontology (GO) enrichment analysis of DEGs, scRepertoire for TCR and BCR data analysis, Monocle for assessing cell differentiation trajectories, and CellChat for intercellular interaction evaluation. Results High-quality single-cell transcriptome data from 12 participants were analyzed, including five with IVIG-resistant KD, four with IVIG-responsive KD, and three healthy controls. We identified 10 major cell types and observed that the differentiation of CD8+ effector T cells was impeded in IVIG-resistant KD patients with coronary artery lesion (CAL) according to cell differentiation trajectory analysis. Subsequent cell communication analysis demonstrated that myeloid cluster with high expression of LCN2, S100P, and LTF played a key role, potentially signaling through MIF-CD74/CXCR4 and MIF-CD74/CD44 ligand-receptor pairs. Conclusion Complex immunopathological changes occur during the development of CAL in IVIG-resistant KD. Stunted differentiation of CD8+ effector T cells is noted in KD-CAL. Interactions between myeloid cells and T cells activates multiple inflammatory signaling pathways, with ligand-receptor pairs, including MIF-CD74/CXCR4 and MIF-CD74/CD44, potentially playing crucial roles.
Collapse
Affiliation(s)
- Yuanzheng Zheng
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yan Zhou
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Di Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xing Fu
- Accuramed Technology (Shanghai) Ltd., Shanghai, 200233, China
| | - Cao Xie
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shuna Sun
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Guoyou Qin
- School of Public Health, Fudan University, Shanghai, 200032, China
| | - Mei Feng
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenglong Liu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qingtong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Research Center for Deepsea Bioresources, Sanya, Hainan, 572025, China
| | - Fang Liu
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Chen Chu
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Feng Wang
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Dehua Yang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Research Center for Deepsea Bioresources, Sanya, Hainan, 572025, China
| | - Ming-Wei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Research Center for Deepsea Bioresources, Sanya, Hainan, 572025, China
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yonghao Gui
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| |
Collapse
|
7
|
Li X, Cui J, Wang L, Cao C, Liu H. Integrated multi-omics profiling reveals the ZZZ3/CD70 axis is a super-enhancer-driven regulator of diffuse large B-cell lymphoma cell-natural killer cell interactions. Exp Biol Med (Maywood) 2024; 249:10155. [PMID: 39376717 PMCID: PMC11457841 DOI: 10.3389/ebm.2024.10155] [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: 03/02/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024] Open
Abstract
Tumor immune microenvironment is crucial for diffuse large B-cell lymphoma (DLBCL) development. However, the mechanisms by which super-enhancers (SEs) regulate the interactions between DLBCL cells and tumor-infiltrating immune cells remains largely unknown. This study aimed to investigate the role of SE-controlled genes in regulating the interactions between DLBCL cells and tumor-infiltrating immune cells. Single-cell RNA-seq, bulk RNA-seq and H3K27ac ChIP-seq data were downloaded from the Heidelberg Open Research Data database and Gene Expression Omnibus database. HOMER algorithm and Seurat package in R were used for bioinformatics analysis. Cell proliferation and lactate dehydrogenase (LDH) release was detected by MTS and LDH release assays, respectively. Interaction between B cell cluster and CD8+ T cell and NK cell cluster was most obviously enhanced in DLBCL, with CD70-CD27, MIF-CD74/CXCR2 complex, MIF-CD74/CD44 complex and CCL3-CCR5 interactions were significantly increased. NK cell sub-cluster showed the strongest interaction with B cell cluster. ZZZ3 upregulated the transcription of CD70 by binding to its SE. Silencing CD70 in DOHH2 cells significantly promoted the proliferation of co-cultured NK92 cells and LDH release from DOHH2 cells, which was counteracted by ZZZ3 overexpression in DOHH2 cells. CD70 silencing combined with PD-L1 blockade promoted LDH release from DOHH2 cells co-cultured with NK92 cells. In conclusion, DLBCL cells inhibited the proliferation and killing of infiltrating NK cells by regulating ZZZ3/CD70 axis. Targeting ZZZ3/CD70 axis combined with PD-L1 blockade is expected to be a promising strategy for DLBCL treatment.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Humans
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/immunology
- CD27 Ligand/metabolism
- CD27 Ligand/genetics
- Cell Line, Tumor
- Tumor Microenvironment
- Gene Expression Regulation, Neoplastic
- Cell Proliferation
- Multiomics
Collapse
|
8
|
Liu B, Li F, Wang Y, Gao X, Li Y, Wang Y, Zhou H. APP-CD74 axis mediates endothelial cell-macrophage communication to promote kidney injury and fibrosis. Front Pharmacol 2024; 15:1437113. [PMID: 39351084 PMCID: PMC11439715 DOI: 10.3389/fphar.2024.1437113] [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: 05/23/2024] [Accepted: 07/26/2024] [Indexed: 10/04/2024] Open
Abstract
Background Kidney injuries often carry a grim prognosis, marked by fibrosis development, renal function loss, and macrophage involvement. Despite extensive research on macrophage polarization and its effects on other cells, like fibroblasts, limited attention has been paid to the influence of non-immune cells on macrophages. This study aims to address this gap by shedding light on the intricate dynamics and diversity of macrophages during renal injury and repair. Methods During the initial research phase, the complexity of intercellular communication in the context of kidney injury was revealed using a publicly available single-cell RNA sequencing library of the unilateral ureteral obstruction (UUO) model. Subsequently, we confirmed our findings using an independent dataset from a renal ischemia-reperfusion injury (IRI) model. We treated two different types of endothelial cells with TGF-β and co-cultured their supernatants with macrophages, establishing an endothelial cell and macrophage co-culture system. We also established a UUO and an IRI mouse model. Western blot analysis, flow cytometry, immunohistochemistry and immunofluorescence staining were used to validate our results at multiple levels. Results Our analysis revealed significant changes in the heterogeneity of macrophage subsets during both injury processes. Amyloid β precursor protein (APP)-CD74 axis mediated endothelial-macrophage intercellular communication plays a dominant role. In the in vitro co-culture system, TGF-β triggers endothelial APP expression, which subsequently enhances CD74 expression in macrophages. Flow cytometry corroborated these findings. Additionally, APP and CD74 expression were significantly increased in the UUO and IRI mouse models. Immunofluorescence techniques demonstrated the co-localization of F4/80 and CD74 in vivo. Conclusion Our study unravels a compelling molecular mechanism, elucidating how endothelium-mediated regulation shapes macrophage function during renal repair. The identified APP-CD74 signaling axis emerges as a promising target for optimizing renal recovery post-injury and preventing the progression of chronic kidney disease.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Faping Li
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuxiong Wang
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Gao
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yunkuo Li
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Yang H, Li J, Huang XR, Bucala R, Xu A, Lan HY. Macrophage-derived macrophage migration inhibitory factor mediates renal injury in anti-glomerular basement membrane glomerulonephritis. Front Immunol 2024; 15:1361343. [PMID: 38846956 PMCID: PMC11153660 DOI: 10.3389/fimmu.2024.1361343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophages are a rich source of macrophage migration inhibitory factor (MIF). It is well established that macrophages and MIF play a pathogenic role in anti-glomerular basement membrane crescentic glomerulonephritis (anti-GBM CGN). However, whether macrophages mediate anti-GBM CGN via MIF-dependent mechanism remains unexplored, which was investigated in this study by specifically deleting MIF from macrophages in MIFf/f-lysM-cre mice. We found that compared to anti-GBM CGN induced in MIFf/f control mice, conditional ablation of MIF in macrophages significantly suppressed anti-GBM CGN by inhibiting glomerular crescent formation and reducing serum creatinine and proteinuria while improving creatine clearance. Mechanistically, selective MIF depletion in macrophages largely inhibited renal macrophage and T cell recruitment, promoted the polarization of macrophage from M1 towards M2 via the CD74/NF-κB/p38MAPK-dependent mechanism. Unexpectedly, selective depletion of macrophage MIF also significantly promoted Treg while inhibiting Th1 and Th17 immune responses. In summary, MIF produced by macrophages plays a pathogenic role in anti-GBM CGN. Targeting macrophage-derived MIF may represent a novel and promising therapeutic approach for the treatment of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Hui Yang
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen University, SunYat‐sen University, Shenzhen, China
| | - Xiao-ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anping Xu
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Gujarati NA, Chow AK, Mallipattu SK. Central role of podocytes in mediating cellular cross talk in glomerular health and disease. Am J Physiol Renal Physiol 2024; 326:F313-F325. [PMID: 38205544 PMCID: PMC11207540 DOI: 10.1152/ajprenal.00328.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes are highly specialized epithelial cells that surround the capillaries of the glomeruli in the kidney. Together with the glomerular endothelial cells, these postmitotic cells are responsible for regulating filtrate from the circulating blood with their organized network of interdigitating foot processes that wrap around the glomerular basement membrane. Although podocyte injury and subsequent loss is the hallmark of many glomerular diseases, recent evidence suggests that the cell-cell communication between podocytes and other glomerular and nonglomerular cells is critical for the development and progression of kidney disease. In this review, we highlight these key cellular pathways of communication and how they might be a potential target for therapy in glomerular disease. We also postulate that podocytes might serve as a central hub for communication in the kidney under basal conditions and in response to cellular stress, which may have implications for the development and progression of glomerular diseases.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Andrew K Chow
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Renal Section, Northport Veterans Affairs Medical Center, Northport, New York, United States
| |
Collapse
|
11
|
Zhu JQ, Zhu Y, Qi M, Zeng Y, Liu ZJ, Ding C, Zhang T, Li XL, Han DD, He Q. Granzyme B+ B cells detected by single-cell sequencing are associated with prognosis in patients with intrahepatic cholangiocarcinoma following liver transplantation. Cancer Immunol Immunother 2024; 73:58. [PMID: 38386050 PMCID: PMC10884120 DOI: 10.1007/s00262-023-03609-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 02/23/2024]
Abstract
B cells possess anti-tumor functions mediated by granzyme B, in addition to their role in antigen presentation and antibody production. However, the variations in granzyme B+ B cells between tumor and non-tumor tissues have been largely unexplored. Therefore, we integrated 25 samples from the Gene Expression Omnibus database and analyzed the tumor immune microenvironment. The findings uncovered significant inter- and intra-tumoral heterogeneity. Notably, single-cell data showed higher proportions of granzyme B+ B cells in tumor samples compared to control samples, and these levels were positively associated with disease-free survival. The elevated levels of granzyme B+ B cells in tumor samples resulted from tumor cell chemotaxis through the MIF- (CD74 + CXCR4) signaling pathway. Furthermore, the anti-tumor function of granzyme B+ B cells in tumor samples was adversely affected, potentially providing an explanation for tumor progression. These findings regarding granzyme B+ B cells were further validated in an independent clinic cohort of 40 liver transplant recipients with intrahepatic cholangiocarcinoma. Our study unveils an interaction between granzyme B+ B cells and intrahepatic cholangiocarcinoma, opening up potential avenues for the development of novel therapeutic strategies against this disease.
Collapse
Affiliation(s)
- Ji-Qiao Zhu
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Ying Zhu
- Department of Clinical Psychology, Mental Hospital of Jianqu Administration Bureau of Jiangsu Province, Nanjing, 210031, Jiangsu, People's Republic of China
| | - Man Qi
- Pathology Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Ye Zeng
- Clinical Lab, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Huazhong University of Science & Technology, Wuhan, 430070, Hubei, People's Republic of China
| | - Zhen-Jia Liu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Cheng Ding
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Tao Zhang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Xian-Liang Li
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Dong-Dong Han
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China.
| | - Qiang He
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China.
| |
Collapse
|
12
|
Yuan T, Zhang J, Zhao Y, Guo Y, Fan S. Single-cell RNA sequencing of intestinal crypts reveals vital events in damage repair and the double-edged sword effect of the Wnt3/β-catenin pathway in irradiated mice. Redox Biol 2023; 68:102942. [PMID: 37918127 PMCID: PMC10638071 DOI: 10.1016/j.redox.2023.102942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
In this study, we executed single-cell RNA sequencing of intestinal crypts. We analyzed the differentially expressed genes (DEGs) at different time points (the first, third, and fifth days) after 13 Gy and 15 Gy abdominal body radiation (ABR) exposure and then executed gene ontology (GO) enrichment analysis, RNA velocity analysis, cell communication analysis, and ligand‒receptor interaction analysis to explore the vital events in damage repair and the multiple effects of the Wnt3/β-catenin pathway on irradiated mice. Results from bioinformatics analysis were confirmed by a series of biological experiments. Results showed that the antibacterial response is a vital event during the damage response process after 13 Gy ABR exposure; ionizing radiation (IR) induced high heterogeneity in the transient amplification (TA) cluster, which may differentiate into mature cells and stem cells in irradiated small intestine (SI) crypts. Conducting an enrichment analysis of the DEGs between mice exposed to 13 Gy and 15 Gy ABR, we concluded that the Wnt3/β-catenin and MIF-CD74/CD44 signaling pathways may contribute to 15 Gy ABR-induced mouse death. Wnt3/β-catenin promotes the recovery of irradiated SI stem/progenitor cells, which may trigger macrophage migration inhibitory factor (MIF) release to further repair IR-induced SI injury; however, with the increase in radiation dose, activation of CD44 on macrophages provides the receptor for MIF signal transduction, initiating the inflammatory cascade response and ultimately causing a cytokine release syndrome. In contrast to previous research, we confirmed that inhibition of the Wnt3/β-catenin pathway or blockade of CD44 on the second day after 15 Gy ABR may significantly protect against ABR-induced death. This study indicates that the Wnt3/β-catenin pathway plays multiple roles in damage repair after IR exposure; we also propose a novel point that the interaction between intestinal crypt stem cells (ISCs) and macrophages through the MIF-CD74/CD44 axis may exacerbate SI damage in irradiated mice.
Collapse
Affiliation(s)
- Tong Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China.
| | - Yue Zhao
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, People's Republic of China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
13
|
Liu C, Wang Y, Zhang YH, Yuan Z, Zhang Z, Zeng X, Guan Z, Bahabayi A, Lu S. Elevated Layilin-Positive Monocyte Levels in the Peripheral Blood of Patients with Systemic Lupus Erythematosus Reflect Their Autoimmune Status. Immunol Invest 2023; 52:879-896. [PMID: 37642473 DOI: 10.1080/08820139.2023.2249531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
OBJECTIVE To investigate the expression of layilin (LAYN) in human circulating monocytes and lymphocytes and its clinical significance in systemic lupus erythematosus (SLE). METHODS Blood samples were collected from 51 SLE patients and 50 healthy controls. Flow cytometry was used to analyze LAYN in lymphocytes and monocyte subsets. Functionally characterized molecules including human HLA, CD74 and CD62L were studied in LAYN+ monocytes. A correlation analysis was conducted between LAYN-related subsets and clinical indicators of SLE such as anti-double-stranded DNA and complements levels. ROC curves were used to explore the potential clinical diagnostic value of LAYN in SLE. RESULTS LAYN was significantly higher in monocytes than in lymphocytes and higher in CD14+CD16+ monocytes than in CD14-CD16+ and CD14+CD16- monocytes. CD74 was upregulated and CD62L was downregulated in LAYN+ monocytes compared with LAYN- monocytes. The absolute number of LAYN+ monocytes was increased in SLE patients, and the median fluorescence intensity of HLA was decreased. LAYN+ monocytes were positively correlated with complement C4, while decreased CD62L+ percentages in LAYN+ monocytes were negatively correlated with C4. The ROC analysis revealed that the area under the curve (AUCs) for CD62L+ percentages in LAYN+ monocytes, LAYN+ lymphocyte numbers, and LAYN+ monocyte numbers to distinguish SLE from healthy individuals were 0.6245, 0.6196 and 0.6173, respectively. CONCLUSION LAYN is differentially expressed in monocytes and their subpopulations and has corresponding functional differences. Changes in LAYN expression on monocytes are associated with complement C4 levels in SLE patients. These suggest that LAYN may be involved in the pathogenesis of SLE. ABBREVIATION ANOVA: analysis of variance; anti-dsDNA: anti-double-stranded DNA; anti-ENA: anti-extractable nuclear antigen; anti-SSA: anti-Sjogren syndrome A; anti-SSB: anti-Sjogren syndrome B; anti-U1RNP: anti-U1 ribonucleoprotein; AUC: area under the ROC curve; CBC: complete blood count; CD62L: L-selectin; CD74/Ii: MHC class II invariant chain; CD44/HCAM: homing cell adhesion molecule; cMos: classical monocytes; CRP: C-reactive protein; CXCR2: C-X-C motif chemokine receptor 2; CXCR4: C-X-C motif chemokine receptor 4; ESR: erythrocyte sedimentation rate; HCs: healthy controls; HA: hyaluronan; HLA: human leukocyte antigen; Ig: immunoglobulin; iMos: intermediate monocytes; LAYN: layilin; MFI: median fluorescence intensity; MIF: migration inhibitory factor; ncMos: nonclassical monocytes; PBMCs: peripheral blood mononuclear cells; ROC: receiver operating characteristic curve; SLE: systemic lupus erythematosus; SLEDAI, SLE disease activity index; Treg: regulatory T cells; WBCs: white blood cells.
Collapse
Affiliation(s)
- Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yiying Wang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ya-Hui Zhang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zihang Yuan
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhonghui Zhang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Zhao Guan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Songsong Lu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
14
|
Bronstein R, Pace J, Gowthaman Y, Salant DJ, Mallipattu SK. Podocyte-Parietal Epithelial Cell Interdependence in Glomerular Development and Disease. J Am Soc Nephrol 2023; 34:737-750. [PMID: 36800545 PMCID: PMC10125654 DOI: 10.1681/asn.0000000000000104] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/04/2023] [Indexed: 02/19/2023] Open
Abstract
Podocytes and parietal epithelial cells (PECs) are among the few principal cell types within the kidney glomerulus, the former serving as a crucial constituent of the kidney filtration barrier and the latter representing a supporting epithelial layer that adorns the inner wall of Bowman's capsule. Podocytes and PECs share a circumscript developmental lineage that only begins to diverge during the S-shaped body stage of nephron formation-occurring immediately before the emergence of the fully mature nephron. These two cell types, therefore, share a highly conserved gene expression program, evidenced by recently discovered intermediate cell types occupying a distinct spatiotemporal gene expression zone between podocytes and PECs. In addition to their homeostatic functions, podocytes and PECs also have roles in kidney pathogenesis. Rapid podocyte loss in diseases, such as rapidly progressive GN and collapsing and cellular subtypes of FSGS, is closely allied with PEC proliferation and migration toward the capillary tuft, resulting in the formation of crescents and pseudocrescents. PECs are thought to contribute to disease progression and severity, and the interdependence between these two cell types during development and in various manifestations of kidney pathology is the primary focus of this review.
Collapse
Affiliation(s)
- Robert Bronstein
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Jesse Pace
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Yogesh Gowthaman
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - David J. Salant
- Division of Nephrology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Sandeep K. Mallipattu
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
- Renal Section, Northport VA Medical Center, Northport, New York
| |
Collapse
|
15
|
Ermert K, Buhl EM, Klinkhammer BM, Floege J, Boor P. Reduction of Endothelial Glycocalyx on Peritubular Capillaries in Chronic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:138-147. [PMID: 36414084 DOI: 10.1016/j.ajpath.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/07/2022] [Accepted: 11/03/2022] [Indexed: 11/20/2022]
Abstract
In chronic kidney disease (CKD), peritubular capillaries undergo anatomic and functional alterations, such as rarefaction and increased permeability. The endothelial glycocalyx (EG) is a carbohydrate-rich gel-like mesh, which covers the luminal surface of endothelial cells. It is involved in many regulatory functions of the endothelium, including vascular permeability. Herein, we investigated ultrastructural alterations of the EG in different murine CKD models. Fluorescence staining using different lectins with high affinity to components of the renal glycocalyx revealed a reduced binding to the endothelium in CKD in the animal models, and there were similar finding in human kidney specimens. Lanthanum Dysprosium Glycosamino Glycan adhesion staining technique was used to visualize the ultrastructure of the glycocalyx in transmission electron microscopy. This also enabled quantitative analyses, showing a significant reduction of the EG thickness and density. In addition, mRNA expression of proteins involved in glycocalyx biology, synthesis, and turnover (ie, syndecan 1 and glypican 1), which are main components of the glycocalyx, and exostosin 2, involved in the synthesis of the glycocalyx, were significantly up-regulated in endothelial cells isolated from murine CKD models. Visualization of glycocalyx using specific transmission electron microscopy analyses allows qualitative and quantitative analyses and revealed significant pathologic alterations in peritubular capillaries in CKD.
Collapse
Affiliation(s)
- Katja Ermert
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Eva M Buhl
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Barbara M Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
16
|
Jia T, Xu T, Smeets B, Buhl EM, Moeller MJ, Floege J, Klinkhammer BM, Boor P. The Role of Platelet-Derived Growth Factor in Focal Segmental Glomerulosclerosis. J Am Soc Nephrol 2023; 34:241-257. [PMID: 36351762 PMCID: PMC10103089 DOI: 10.1681/asn.2022040491] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND FSGS is the final common pathway to nephron loss in most forms of severe or progressive glomerular injury. Although podocyte injury initiates FSGS, parietal epithelial cells (PECs) are the main effectors. Because PDGF takes part in fibrotic processes, we hypothesized that the ligand PDGF-B and its receptor PDGFR- β participate in the origin and progression of FSGS. METHODS We challenged Thy1.1 transgenic mice, which express Thy1.1 in the podocytes, with anti-Thy1.1 antibody to study the progression of FSGS. We investigated the role of PDGF in FSGS using challenged Thy1.1 mice, 5/6 nephrectomized mice, Col4 -/- (Alport) mice, patient kidney biopsies, and primary murine PECs, and challenged Thy1.1 mice treated with neutralizing anti-PDGF-B antibody therapy. RESULTS The unchallenged Thy1.1 mice developed only mild spontaneous FSGS, whereas challenged mice developed progressive FSGS accompanied by a decline in kidney function. PEC activation, proliferation, and profibrotic phenotypic switch drove the FSGS. During disease, PDGF-B was upregulated in podocytes, whereas PDGFR- β was upregulated in PECs from both mice and patients with FSGS. Short- and long-term treatment with PDGF-B neutralizing antibody improved kidney function and reduced FSGS, PEC proliferation, and profibrotic activation. In vitro , stimulation of primary murine PECs with PDGF-B recapitulated in vivo findings with PEC activation and proliferation, which was inhibited by PDGF-B antibody or imatinib. CONCLUSION PDGF-B-PDGFR- β molecular crosstalk between podocytes and PECs drives glomerulosclerosis and the progression of FSGS. PODCAST This article contains a podcast at.
Collapse
Affiliation(s)
- Ting Jia
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tong Xu
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Eva Miriam Buhl
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Marcus Johannes Moeller
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
- Heisenberg Chair for Preventive and Translational Nephrology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Barbara Mara Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
17
|
Huang Y, Zhao X, Zhang Q, Yang X, Hou G, Peng C, Jia M, Zhou L, Yamamoto T, Zheng J. Novel therapeutic perspectives for crescentic glomerulonephritis through targeting parietal epithelial cell activation and proliferation. Expert Opin Ther Targets 2023; 27:55-69. [PMID: 36738160 DOI: 10.1080/14728222.2023.2177534] [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: 02/05/2023]
Abstract
INTRODUCTION Kidney injury is clinically classified as crescentic glomerulonephritis (CrGN) when ≥50% of the glomeruli in a biopsy sample contain crescentic lesions. However, current strategies, such as systemic immunosuppressive therapy and plasmapheresis for CrGN, are partially effective, and these drugs have considerable systemic side effects. Hence, targeted therapy to prevent glomerular crescent formation and expansion remains an unmet clinical need. AREAS COVERED Hyperproliferative parietal epithelial cells (PECs) are the main constituent cells of the glomerular crescent with cell-tracing evidence. Crescents obstruct the flow of primary urine, pressure the capillaries, and degenerate the affected nephrons. We reviewed the markers of PEC activation and proliferation, potential therapeutic effects of thrombin and thrombin receptor inhibitors, and how podocytes cross-talk with PECs. These experiments may help identify potential early specific targets for the prevention and treatment of glomerular crescentic injury. EXPERT OPINION Inhibiting PEC activation and proliferation in CrGN can alleviate glomerular crescent progression, which has been supported by preclinical studies with evidence of genetic deletion. Clarifying the outcome of PEC transformation to the podocyte phenotype and suppressing thrombin, thrombin receptors, and PEC hyperproliferation in early therapeutic strategies will be the research goals in the next ten years.
Collapse
Affiliation(s)
- Yanjie Huang
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China.,Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xueru Zhao
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qiushuang Zhang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoqing Yang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Gailing Hou
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Chaoqun Peng
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mengzhen Jia
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Li Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tatsuo Yamamoto
- Department of Nephrology, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda, Japan
| | - Jian Zheng
- Institute of Pediatrics of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| |
Collapse
|
18
|
Du Y, Hao H, Ma H, Liu H. Macrophage migration inhibitory factor in acute kidneyinjury. Front Physiol 2022; 13:945827. [PMID: 36117692 PMCID: PMC9478040 DOI: 10.3389/fphys.2022.945827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Acute kidney injury (AKI) is a complex clinical syndrome with multiple etiologies and pathogenesis, which lacks early biomarkers and targeted therapy. Recently, macrophage migration inhibitory factor (MIF) family protein have received increasing attention owing to its pleiotropic protein molecule character in acute kidney injury, where it performed a dual role in the pathological process. macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 are released into the peripheral circulation when Acute kidney injury occurs and interact with various cellular pathways. On the one hand, macrophage migration inhibitory factor exerts a protective effect in anti-oxidation and macrophage migration inhibitory factor-2 promotes cell proliferation and ameliorates renal fibrosis. On the other hand, macrophage migration inhibitory factor aggravates renal injury as an upstream inflammation factor. Herein, we provide an overview on the biological role and possible mechanisms of macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 in the process of Acute kidney injury and the clinical application prospects of macrophage migration inhibitory factor family proteins as a potential therapeutic target.
Collapse
Affiliation(s)
- Yiwei Du
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Hao Hao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| | - Hongbao Liu
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| |
Collapse
|
19
|
Jorgačević B, Stanković S, Filipović J, Samardžić J, Vučević D, Radosavljević T. Betaine Modulating MIF-Mediated Oxidative Stress, Inflammation and Fibrogenesis in Thioacetamide-Induced Nephrotoxicity. Curr Med Chem 2022; 29:5254-5267. [PMID: 35400322 DOI: 10.2174/0929867329666220408102856] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with chemokine properties released by various immune and non-immune cells. It contributes to the pathogenesis of many inflammatory, autoimmune diseases and malignant tumors. OBJECTIVE Our study aimed to investigate the role of betaine in the modulation of MIF-mediated oxidative stress, inflammation, and fibrogenesis during toxic kidney damage induced by thioacetamide (TAA). METHODS The experiment is performed on wild-type and knockout MIF-/- C57BL/6 mice. They are randomly divided into groups: Control; Bet-group, received betaine (2% wt/v dissolved in drinking water); MIF-/- mice group; MIF-/- + Bet; TAA-group, treated with TAA (200 mg/kg b.w.), intraperitoneally, 3x/week/8 weeks); TAA+Bet; MIF-/-+TAA, and MIF-/- + TAA+Bet group. After eight weeks of treatment, animals are sacrificed and kidney samples are taken to determine oxidative stress parameters, proinflammatory cytokines, profibrogenic factors, and histopathology of renal tissue. RESULTS In MIF-/-mice, TAA decreases malondialdehyde (MDA) concentration, IL-6, tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta 1 (TGF-β1) and platelet-derived growth factor-BB (PDGF-BB) and increases superoxide dismutases (SOD) and catalase (CAT) activities, as well as glutathione (GSH) content in kidneys, compared to TAA group. Betaine alleviates the mechanism of MIF-mediated effects in TAA-induced nephrotoxicity, reducing MDA, IL-6, TNF-α, TGF-β1, and PDGF-BB, and increasing SOD and CAT activity, as well as GSH levels. CONCLUSION MIF mediates TAA-induced nephrotoxicity by increasing oxidative stress, inflammation, and profibrogenic mediators. MIF-targeted therapy could potentially alleviate oxidative stress and inflammation in the kidney, as well as pathohistological changes in renal tissue, but the exact mechanism of its action is not completely clear. Betaine alleviates MIF nephrotoxic effects by increasing the antioxidative capacity of kidney cells, and decreasing lipid peroxidation and cytokine production in the renal tissue. It suggests that betaine can be used for the prevention of kidney damage.
Collapse
Affiliation(s)
- Bojan Jorgačević
- Faculty of Medicine, Institute of Pathophysiology \'\'Ljubodrag Buba Mihailović\'\', University of Belgrade, Belgrade 11000, Serbia
| | - Sanja Stanković
- Centre of Medical Biochemistry, Clinical Centre of Serbia, 11000 Belgrade, Belgrade, Serbia
| | - Jelena Filipović
- Faculty of Medicine, Institute of Pathology \'\'Đorđe Jovanović\'\', University of Belgrade, Belgrade 11000, Serbia
| | - Janko Samardžić
- Faculty of Medicine, Institute of Pharmacology, Clinical Pharmacology and Toxicology, University of Belgrade, 11000 Belgrade, Serbia
| | - Danijela Vučević
- Faculty of Medicine, Institute of Pathophysiology \'\'Ljubodrag Buba Mihailović\'\', University of Belgrade, Belgrade 11000, Serbia
| | - Tatjana Radosavljević
- Faculty of Medicine, Institute of Pathophysiology \'\'Ljubodrag Buba Mihailović\'\', University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
20
|
Raghubar AM, Pham DT, Tan X, Grice LF, Crawford J, Lam PY, Andersen SB, Yoon S, Teoh SM, Matigian NA, Stewart A, Francis L, Ng MSY, Healy HG, Combes AN, Kassianos AJ, Nguyen Q, Mallett AJ. Spatially Resolved Transcriptomes of Mammalian Kidneys Illustrate the Molecular Complexity and Interactions of Functional Nephron Segments. Front Med (Lausanne) 2022; 9:873923. [PMID: 35872784 PMCID: PMC9300864 DOI: 10.3389/fmed.2022.873923] [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: 02/11/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Available transcriptomes of the mammalian kidney provide limited information on the spatial interplay between different functional nephron structures due to the required dissociation of tissue with traditional transcriptome-based methodologies. A deeper understanding of the complexity of functional nephron structures requires a non-dissociative transcriptomics approach, such as spatial transcriptomics sequencing (ST-seq). We hypothesize that the application of ST-seq in normal mammalian kidneys will give transcriptomic insights within and across species of physiology at the functional structure level and cellular communication at the cell level. Here, we applied ST-seq in six mice and four human kidneys that were histologically absent of any overt pathology. We defined the location of specific nephron structures in the captured ST-seq datasets using three lines of evidence: pathologist's annotation, marker gene expression, and integration with public single-cell and/or single-nucleus RNA-sequencing datasets. We compared the mouse and human cortical kidney regions. In the human ST-seq datasets, we further investigated the cellular communication within glomeruli and regions of proximal tubules-peritubular capillaries by screening for co-expression of ligand-receptor gene pairs. Gene expression signatures of distinct nephron structures and microvascular regions were spatially resolved within the mouse and human ST-seq datasets. We identified 7,370 differentially expressed genes (p adj < 0.05) distinguishing species, suggesting changes in energy production and metabolism in mouse cortical regions relative to human kidneys. Hundreds of potential ligand-receptor interactions were identified within glomeruli and regions of proximal tubules-peritubular capillaries, including known and novel interactions relevant to kidney physiology. Our application of ST-seq to normal human and murine kidneys confirms current knowledge and localization of transcripts within the kidney. Furthermore, the generated ST-seq datasets provide a valuable resource for the kidney community that can be used to inform future research into this complex organ.
Collapse
Affiliation(s)
- Arti M. Raghubar
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Duy T. Pham
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Xiao Tan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Laura F. Grice
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Joanna Crawford
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Pui Yeng Lam
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Stacey B. Andersen
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
- UQ Sequencing Facility, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Sohye Yoon
- Genome Innovation Hub, University of Queensland, Brisbane, QLD, Australia
| | - Siok Min Teoh
- UQ Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas A. Matigian
- QCIF Facility for Advanced Bioinformatics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Anne Stewart
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
| | - Leo Francis
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
| | - Monica S. Y. Ng
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Nephrology Department, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Helen G. Healy
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Alexander N. Combes
- Department of Anatomy and Developmental Biology, Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Andrew J. Kassianos
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Mallett
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, Queensland, QLD, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, Queensland, QLD, Australia
| |
Collapse
|
21
|
Macrophage Migration Inhibitory Factor (MIF) as a Stress Molecule in Renal Inflammation. Int J Mol Sci 2022; 23:ijms23094908. [PMID: 35563296 PMCID: PMC9102975 DOI: 10.3390/ijms23094908] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Renal inflammation is an initial pathological process during progressive renal injury regardless of the initial cause. Macrophage migration inhibitory factor (MIF) is a truly proinflammatory stress mediator that is highly expressed in a variety of both inflammatory cells and intrinsic kidney cells. MIF is released from the diseased kidney immediately upon stimulation to trigger renal inflammation by activating macrophages and T cells, and promoting the production of proinflammatory cytokines, chemokines, and stress molecules via signaling pathways involving the CD74/CD44 and chemokine receptors CXCR2, CXCR4, and CXCR7 signaling. In addition, MIF can function as a stress molecule to counter-regulate the immunosuppressive effect of glucocorticoid in renal inflammation. Given the critical position of MIF in the upstream inflammatory cascade, this review focuses on the regulatory role and molecular mechanisms of MIF in kidney diseases. The therapeutic potential of targeting MIF signaling to treat kidney diseases is also discussed.
Collapse
|
22
|
Loss of CLDN5 in podocytes deregulates WIF1 to activate WNT signaling and contributes to kidney disease. Nat Commun 2022; 13:1600. [PMID: 35332151 PMCID: PMC8948304 DOI: 10.1038/s41467-022-29277-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Although mature podocytes lack tight junctions, tight junction integral membrane protein claudin-5 (CLDN5) is predominantly expressed on plasma membranes of podocytes under normal conditions. Using podocyte-specific Cldn5 knockout mice, we identify CLDN5 as a crucial regulator of podocyte function and reveal that Cldn5 deletion exacerbates podocyte injury and proteinuria in a diabetic nephropathy mouse model. Mechanistically, CLDN5 deletion reduces ZO1 expression and induces nuclear translocation of ZONAB, followed by transcriptional downregulation of WNT inhibitory factor-1 (WIF1) expression, which leads to activation of WNT signaling pathway. Podocyte-derived WIF1 also plays paracrine roles in tubular epithelial cells, as evidenced by the finding that animals with podocyte-specific deletion of Cldn5 or Wif1 have worse kidney fibrosis after unilateral ureteral obstruction than littermate controls. Systemic delivery of WIF1 suppresses the progression of diabetic nephropathy and ureteral obstruction-induced renal fibrosis. These findings establish a function for podocyte CLDN5 in restricting WNT signaling in kidney. Claudin-5 is a tight junction integral membrane protein, but it is also expressed in mature podocytes which lack tight junctions. Here the authors report that podocyte claudin-5 regulates WNT signaling activity by modulating WIF1 expression, and its downregulation contributes to kidney disease progression in mice.
Collapse
|
23
|
Li ZH, Guo XY, Quan XY, Yang C, Liu ZJ, Su HY, An N, Liu HF. The Role of Parietal Epithelial Cells in the Pathogenesis of Podocytopathy. Front Physiol 2022; 13:832772. [PMID: 35360248 PMCID: PMC8963495 DOI: 10.3389/fphys.2022.832772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 02/05/2023] Open
Abstract
Podocytopathy is the most common feature of glomerular disorder characterized by podocyte injury- or dysfunction-induced excessive proteinuria, which ultimately develops into glomerulosclerosis and results in persistent loss of renal function. Due to the lack of self-renewal ability of podocytes, mild podocyte depletion triggers replacement and repair processes mostly driven by stem cells or resident parietal epithelial cells (PECs). In contrast, when podocyte recovery fails, activated PECs contribute to the establishment of glomerular lesions. Increasing evidence suggests that PECs, more than just bystanders, have a crucial role in various podocytopathies, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, diabetic nephropathy, IgA nephropathy, and lupus podocytopathy. In this review, we attempt to dissect the diverse role of PECs in the pathogenesis of podocytopathy based on currently available information.
Collapse
|
24
|
Moeller MJ, Kramann R, Lammers T, Hoppe B, Latz E, Ludwig-Portugall I, Boor P, Floege J, Kurts C, Weiskirchen R, Ostendorf T. New Aspects of Kidney Fibrosis-From Mechanisms of Injury to Modulation of Disease. Front Med (Lausanne) 2022; 8:814497. [PMID: 35096904 PMCID: PMC8790098 DOI: 10.3389/fmed.2021.814497] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 02/02/2023] Open
Abstract
Organ fibrogenesis is characterized by a common pathophysiological final pathway independent of the underlying progressive disease of the respective organ. This makes it particularly suitable as a therapeutic target. The Transregional Collaborative Research Center “Organ Fibrosis: From Mechanisms of Injury to Modulation of Disease” (referred to as SFB/TRR57) was hosted from 2009 to 2021 by the Medical Faculties of RWTH Aachen University and the University of Bonn. This consortium had the ultimate goal of discovering new common but also different fibrosis pathways in the liver and kidneys. It finally successfully identified new mechanisms and established novel therapeutic approaches to interfere with hepatic and renal fibrosis. This review covers the consortium's key kidney-related findings, where three overarching questions were addressed: (i) What are new relevant mechanisms and signaling pathways triggering renal fibrosis? (ii) What are new immunological mechanisms, cells and molecules that contribute to renal fibrosis?, and finally (iii) How can renal fibrosis be modulated?
Collapse
Affiliation(s)
- Marcus J Moeller
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Heisenberg Chair for Preventive and Translational Nephrology, Aachen, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Faculty of Medicine, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Bernd Hoppe
- Division of Pediatric Nephrology and Kidney Transplantation, University Hospital of Bonn, Bonn, Germany.,German Hyperoxaluria Center, Pediatric Kidney Care Center, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital of Bonn, Bonn, Germany
| | - Isis Ludwig-Portugall
- Institute for Molecular Medicine and Experimental Immunology, University Hospital of Bonn, Bonn, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Kurts
- Institute for Molecular Medicine and Experimental Immunology, University Hospital of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital RWTH Aachen, Aachen, Germany
| | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
25
|
Pace JA, Bronstein R, Guo Y, Yang Y, Estrada CC, Gujarati N, Salant DJ, Haley J, Bialkowska AB, Yang VW, He JC, Mallipattu SK. Podocyte-specific KLF4 is required to maintain parietal epithelial cell quiescence in the kidney. SCIENCE ADVANCES 2021; 7:eabg6600. [PMID: 34516901 PMCID: PMC8442927 DOI: 10.1126/sciadv.abg6600] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/13/2021] [Indexed: 06/06/2023]
Abstract
Podocyte loss triggering aberrant activation and proliferation of parietal epithelial cells (PECs) is a central pathogenic event in proliferative glomerulopathies. Podocyte-specific Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor, is essential for maintaining podocyte homeostasis and PEC quiescence. Using mice with podocyte-specific knockdown of Klf4, we conducted glomerular RNA-sequencing, tandem mass spectrometry, and single-nucleus RNA-sequencing to identify cell-specific transcriptional changes that trigger PEC activation due to podocyte loss. Integration with in silico chromatin immunoprecipitation identified key ligand-receptor interactions, such as fibronectin 1 (FN1)–αVβ6, between podocytes and PECs dependent on KLF4 and downstream signal transducer and activator of transcription 3 (STAT3) signaling. Knockdown of Itgb6 in PECs attenuated PEC activation. Additionally, podocyte-specific induction of human KLF4 or pharmacological inhibition of downstream STAT3 activation reduced FN1 and integrin β 6 (ITGB6) expression and mitigated podocyte loss and PEC activation in mice. Targeting podocyte-PEC crosstalk might be a critical therapeutic strategy in proliferative glomerulopathies.
Collapse
Affiliation(s)
- Jesse A. Pace
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Robert Bronstein
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Yiqing Guo
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Yaqi Yang
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Chelsea C. Estrada
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Nehaben Gujarati
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - David J. Salant
- Division of Nephrology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John Haley
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Agnieszka B. Bialkowska
- Division of Gastroenterology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Vincent W. Yang
- Division of Gastroenterology, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - John C. He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandeep K. Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
- Renal Section, Northport VA Medical Center, Northport, NY, USA
| |
Collapse
|
26
|
Parietal epithelial cell dysfunction in crescentic glomerulonephritis. Cell Tissue Res 2021; 385:345-354. [PMID: 34453566 PMCID: PMC8523405 DOI: 10.1007/s00441-021-03513-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Crescentic glomerulonephritis represents a group of kidney diseases characterized by rapid loss of kidney function and the formation of glomerular crescents. While the role of the immune system has been extensively studied in relation to the development of crescents, recent findings show that parietal epithelial cells play a key role in the pathophysiology of crescent formation, even in the absence of immune modulation. This review highlights our current understanding of parietal epithelial cell biology and the reported physiological and pathological roles that these cells play in glomerular lesion formation, especially in the context of crescentic glomerulonephritis.
Collapse
|
27
|
Zhao Y, Li Z, Zhu Y, Fu J, Zhao X, Zhang Y, Wang S, Wu J, Wang K, Wu R, Sui C, Shen S, Wu X, Wang H, Gao D, Chen L. Single-Cell Transcriptome Analysis Uncovers Intratumoral Heterogeneity and Underlying Mechanisms for Drug Resistance in Hepatobiliary Tumor Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003897. [PMID: 34105295 PMCID: PMC8188185 DOI: 10.1002/advs.202003897] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/29/2020] [Indexed: 05/30/2023]
Abstract
Molecular heterogeneity of hepatobiliary tumor including intertumoral and intratumoral disparity always leads to drug resistance. Here, seven hepatobiliary tumor organoids are generated to explore heterogeneity and evolution via single-cell RNA sequencing. HCC272 with high status of epithelia-mesenchymal transition proves broad-spectrum drug resistance. By examining the expression pattern of cancer stem cells markers (e.g., PROM1, CD44, and EPCAM), it is found that CD44 positive population may render drug resistance in HCC272. UMAP and pseudo-time analysis identify the intratumoral heterogeneity and distinct evolutionary trajectories, of which catenin beta-1 (CTNNB1), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and nuclear paraspeckle assembly transcript 1 (NEAT1) advantage expression clusters are commonly shared across hepatobiliary organoids. CellphoneDB analysis further implies that metabolism advantage organoids with enrichment of hypoxia signal upregulate NEAT1 expression in CD44 subgroup and mediate drug resistance that relies on Jak-STAT pathway. Moreover, metabolism advantage clusters shared in several organoids have similar characteristic genes (GAPDH, NDRG1 (N-Myc downstream regulated 1), ALDOA, and CA9). The combination of GAPDH and NDRG1 is an independent risk factor and predictor for patient survival. This study delineates heterogeneity of hepatobiliary tumor organoids and proposes that the collaboration of intratumoral heterogenic subpopulations renders malignant phenotypes and drug resistance.
Collapse
Affiliation(s)
- Yan Zhao
- School of Life Sciences and Institute of Metabolism and Integrative BiologyFudan UniversityShanghai200438China
| | - Zhi‐Xuan Li
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Yan‐Jing Zhu
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Jing Fu
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Xiao‐Fang Zhao
- Fudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Ya‐Ni Zhang
- Institute of Metabolism and Integrative BiologyFudan UniversityShanghai200438China
| | - Shan Wang
- Fudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jian‐Min Wu
- School of Life Sciences and Institute of Metabolism and Integrative BiologyFudan UniversityShanghai200438China
| | - Kai‐Ting Wang
- School of Life Sciences and Institute of Metabolism and Integrative BiologyFudan UniversityShanghai200438China
| | - Rui Wu
- Eastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Cheng‐Jun Sui
- Eastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Si‐Yun Shen
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
| | - Xuan Wu
- Department of Laboratory MedicineThe Tenth People's Hospital of ShanghaiTongji UniversityShanghai200072China
| | - Hong‐Yang Wang
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
- Fudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Dong Gao
- The State Key Laboratory of Cell BiologyShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Lei Chen
- National Center for Liver CancerShanghai200441China
- The International Cooperation Laboratory on Signal TransductionEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghai200438China
- Fudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
28
|
Parietal epithelial cells role in repair versus scarring after glomerular injury. Curr Opin Nephrol Hypertens 2021; 29:293-301. [PMID: 32235272 DOI: 10.1097/mnh.0000000000000600] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW The recent years have been marked by the publication of several articles highlighting the pathophysiological role of glomerular parietal epithelial cells (PEC) and refining their phenotypic heterogeneity. RECENT FINDINGS The present review synthetizes recent findings on (i) the potential regenerative role of PEC in glomerular diseases, and (ii) the mechanisms and signaling of leading to PEC pathogenic involvement in crescentic glomerulonephritis (CGN) and focal segmental glomerulosclerosis (FSGS). SUMMARY The debate is still open regarding the podocyte regenerative properties of PEC in glomerular disease, whereas the pathogenic involvement of PEC activation in glomerular disease is increasingly admitted. Recent highlights on the podocyte regenerative role of PEC, on one hand, and on their pathological function, on the other hand, for sure will feed the debate in the kidney community for the next years. Nevertheless, from a therapeutic perspective, the two options, boosting cellular regeneration and blocking PECs pathogenicity, should not be seen as antagonistic but, rather, complementary.
Collapse
|
29
|
Heinrichs D, Brandt EF, Fischer P, Köhncke J, Wirtz TH, Guldiken N, Djudjaj S, Boor P, Kroy D, Weiskirchen R, Bucala R, Wasmuth HE, Strnad P, Trautwein C, Bernhagen J, Berres ML. Unexpected Pro-Fibrotic Effect of MIF in Non-Alcoholic Steatohepatitis Is Linked to a Shift in NKT Cell Populations. Cells 2021; 10:252. [PMID: 33525493 PMCID: PMC7918903 DOI: 10.3390/cells10020252] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory cytokine with anti-fibrotic properties in toxic liver injury models and anti-steatotic functions in non-alcoholic fatty liver disease (NAFLD) attributed to the CD74/AMPK signaling pathway. As NAFLD progression is associated with fibrosis, we studied MIF function during NAFLD-associated liver fibrogenesis in mice and men by molecular, histological and immunological methods in vitro and in vivo. After NASH diet feeding, hepatic Mif expression was strongly induced, an effect which was absent in Mif∆hep mice. In contrast to hepatotoxic fibrosis models, NASH diet-induced fibrogenesis was significantly abrogated in Mif-/- and Mif∆hep mice associated with a reduced accumulation of the pro-fibrotic type-I NKT cell subpopulation. In vitro, MIF skewed the differentiation of NKT cells towards the type-I subtype. In line with the murine results, expression of fibrosis markers strongly correlated with MIF, its receptors, and markers of NKT type-I cells in NASH patients. We conclude that MIF expression is induced during chronic metabolic injury in mice and men with hepatocytes representing the major source. In NAFLD progression, MIF contributes to liver fibrogenesis skewing NKT cell polarization toward a pro-fibrotic phenotype highlighting the complex, context-dependent role of MIF during chronic liver injury.
Collapse
Affiliation(s)
- Daniel Heinrichs
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Elisa F. Brandt
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Petra Fischer
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Janine Köhncke
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Theresa H. Wirtz
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Nurdan Guldiken
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Sonja Djudjaj
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.D.); (P.B.)
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.D.); (P.B.)
| | - Daniela Kroy
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, 52074 Aachen, Germany;
| | - Richard Bucala
- Rheumatology Section of the Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8031, USA;
| | - Hermann E. Wasmuth
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Pavel Strnad
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute of Stroke and Dementia Research, LMU Klinikum, Lud-wig-Maximilian-University (LMU), 81377 Munich, Germany;
- Munich Cluster for Systems Neurology, 81377 Munich, Germany
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (D.H.); (E.F.B.); (P.F.); (J.K.); (T.H.W.); (N.G.); (D.K.); (H.E.W.); (P.S.); (C.T.)
| |
Collapse
|
30
|
Bouteldja N, Klinkhammer BM, Bülow RD, Droste P, Otten SW, Freifrau von Stillfried S, Moellmann J, Sheehan SM, Korstanje R, Menzel S, Bankhead P, Mietsch M, Drummer C, Lehrke M, Kramann R, Floege J, Boor P, Merhof D. Deep Learning-Based Segmentation and Quantification in Experimental Kidney Histopathology. J Am Soc Nephrol 2021; 32:52-68. [PMID: 33154175 PMCID: PMC7894663 DOI: 10.1681/asn.2020050597] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/09/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Nephropathologic analyses provide important outcomes-related data in experiments with the animal models that are essential for understanding kidney disease pathophysiology. Precision medicine increases the demand for quantitative, unbiased, reproducible, and efficient histopathologic analyses, which will require novel high-throughput tools. A deep learning technique, the convolutional neural network, is increasingly applied in pathology because of its high performance in tasks like histology segmentation. METHODS We investigated use of a convolutional neural network architecture for accurate segmentation of periodic acid-Schiff-stained kidney tissue from healthy mice and five murine disease models and from other species used in preclinical research. We trained the convolutional neural network to segment six major renal structures: glomerular tuft, glomerulus including Bowman's capsule, tubules, arteries, arterial lumina, and veins. To achieve high accuracy, we performed a large number of expert-based annotations, 72,722 in total. RESULTS Multiclass segmentation performance was very high in all disease models. The convolutional neural network allowed high-throughput and large-scale, quantitative and comparative analyses of various models. In disease models, computational feature extraction revealed interstitial expansion, tubular dilation and atrophy, and glomerular size variability. Validation showed a high correlation of findings with current standard morphometric analysis. The convolutional neural network also showed high performance in other species used in research-including rats, pigs, bears, and marmosets-as well as in humans, providing a translational bridge between preclinical and clinical studies. CONCLUSIONS We developed a deep learning algorithm for accurate multiclass segmentation of digital whole-slide images of periodic acid-Schiff-stained kidneys from various species and renal disease models. This enables reproducible quantitative histopathologic analyses in preclinical models that also might be applicable to clinical studies.
Collapse
Affiliation(s)
- Nassim Bouteldja
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Barbara M. Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany,Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Roman D. Bülow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Patrick Droste
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Simon W. Otten
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Julia Moellmann
- Department of Cardiology and Vascular Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | | | | | - Sylvia Menzel
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Bankhead
- Edinburgh Pathology, University of Edinburgh, Edinburgh, United Kingdom,Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthias Mietsch
- Laboratory Animal Science Unit, German Primate Center, Goettingen, Germany
| | - Charis Drummer
- Platform Degenerative Diseases, German Primate Center, Goettingen, Germany
| | - Michael Lehrke
- Department of Cardiology and Vascular Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany,Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Dorit Merhof
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany,Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| |
Collapse
|
31
|
Valiño-Rivas L, Cuarental L, Nuñez G, Sanz AB, Ortiz A, Sanchez-Niño MD. Loss of NLRP6 expression increases the severity of acute kidney injury. Nephrol Dial Transplant 2020; 35:587-598. [PMID: 31504777 DOI: 10.1093/ndt/gfz169] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Nlrp6 is a nucleotide-binding oligomerization domain-like receptor (NLR) that forms atypical inflammasomes. Nlrp6 modulates the gut epithelium interaction with the microbiota. However, the expression and function of Nlrp6 in the kidney, a sterile environment, have not been characterized. We explored the role of Nlrp6 in acute kidney injury (AKI). METHODS In a transcriptomics array of murine nephrotoxic AKI, Nlrp6 and Naip3 were the only significantly downregulated NLR genes. The functional implications of Nlrp6 downregulation were explored in mice and in cultured murine tubular cells. RESULTS Nlrp6 was expressed by healthy murine and human kidney tubular epithelium, and expression was reduced during human kidney injury or murine nephrotoxic AKI induced by cisplatin or a folic acid overdose. Genetic Nlrp6 deficiency resulted in upregulation of kidney extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK) phosphorylation and more severe AKI and kidney inflammation. In cultured tubular cells, Nlrp6 downregulation induced by specific small interfering RNA resulted in upregulation of ERK1/2 and p38 phosphorylation and chemokine messenger RNA expression and downregulation of the nephroprotective gene Klotho. MAPK inhibition prevented the inflammatory response in Nlrp6-deficient cells. CONCLUSION Nlrp6 dampens sterile inflammation and has a nephroprotective role during nephrotoxic kidney injury through suppression of MAP kinase activation.
Collapse
Affiliation(s)
- Lara Valiño-Rivas
- Nephrology and Hypertension Laboratory, IIS-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain.,Nephrology and Hypertension Laboratory, REDINREN, Madrid, Spain
| | - Leticia Cuarental
- Nephrology and Hypertension Laboratory, IIS-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - Gabriel Nuñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ana B Sanz
- Nephrology and Hypertension Laboratory, IIS-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain.,Nephrology and Hypertension Laboratory, REDINREN, Madrid, Spain
| | - Alberto Ortiz
- Nephrology and Hypertension Laboratory, IIS-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain.,Nephrology and Hypertension Laboratory, REDINREN, Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Nephrology and Hypertension Laboratory, IIS-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain.,Nephrology and Hypertension Laboratory, REDINREN, Madrid, Spain
| |
Collapse
|
32
|
Al Abdulmonem W, Rasheed Z, Al Ssadh H, Alkhamiss A, Aljohani AS, Fernández N. Bacterial lipopolysaccharide induces the intracellular expression of trophoblastic specific CD74 isoform in human first trimester trophoblast cells: Correlation with unsuccessful early pregnancy. J Reprod Immunol 2020; 141:103152. [PMID: 32521377 DOI: 10.1016/j.jri.2020.103152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 10/24/2022]
Abstract
OBJECTIVE During first trimester of human pregnancy, the maternal system develops immunity against infection and to provide protection of allogeneic foetus from abortion. This study was undertaken to determine the role of trophoblast specific CD74 isoforms in first trimester trophoblast derived cells under normal and lipopolysaccharide (LPS) stimulated conditions. METHODS Gene and protein of CD74 were determined in first trimester trophoblast derived cells, JEG-3 and ACH-3 P and also in human placenta by PCR, western blotting and immunoprecipitation. Effect of LPS mediated infection on the regulation of CD74 isoforms was studied intracellularly and also on the cells surface by flow cytometry. RESULTS Data demonstrated that JEG-3 and ACH-3 P cells under normal conditions have not expressed CD74 isoforms neither intracellularly or nor on the surface. These results were further validated directly in human placenta. However, treatment of these trophoblast cells with a bacterial LPS, significantly upregulated CD74 mRNA expression (p < 0.05). Furthermore, expression of CD74 on the surface was not detected even after stimulation with LPS. Interestingly, CD74 isoform at 35 kDa was significantly detected intracellularly upon stimulation with LPS (p < 0.05). These results were further confirmed by western blotting followed by immunoprecipitation. CONCLUSIONS To the best of our knowledge, this is the first study concluded that the bacterial LPS induce infection in the first trimester trophoblasts via intracellular upregulation of CD74. Data indicated that the lack of cell surface expression of trophoblastic specific isoforms of CD74 may provide protection for human pregnancy in the first trimester.
Collapse
Affiliation(s)
- Waleed Al Abdulmonem
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom; Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Qassim University, Buraidah, Saudi Arabia.
| | - Hussain Al Ssadh
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Abdullah Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Abdullah Sm Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Nelson Fernández
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| |
Collapse
|
33
|
Buhl EM, Djudjaj S, Klinkhammer BM, Ermert K, Puelles VG, Lindenmeyer MT, Cohen CD, He C, Borkham‐Kamphorst E, Weiskirchen R, Denecke B, Trairatphisan P, Saez‐Rodriguez J, Huber TB, Olson LE, Floege J, Boor P. Dysregulated mesenchymal PDGFR-β drives kidney fibrosis. EMBO Mol Med 2020; 12:e11021. [PMID: 31943786 PMCID: PMC7059015 DOI: 10.15252/emmm.201911021] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Kidney fibrosis is characterized by expansion and activation of platelet-derived growth factor receptor-β (PDGFR-β)-positive mesenchymal cells. To study the consequences of PDGFR-β activation, we developed a model of primary renal fibrosis using transgenic mice with PDGFR-β activation specifically in renal mesenchymal cells, driving their pathological proliferation and phenotypic switch toward myofibroblasts. This resulted in progressive mesangioproliferative glomerulonephritis, mesangial sclerosis, and interstitial fibrosis with progressive anemia due to loss of erythropoietin production by fibroblasts. Fibrosis induced secondary tubular epithelial injury at later stages, coinciding with microinflammation, and aggravated the progression of hypertensive and obstructive nephropathy. Inhibition of PDGFR activation reversed fibrosis more effectively in the tubulointerstitium compared to glomeruli. Gene expression signatures in mice with PDGFR-β activation resembled those found in patients. In conclusion, PDGFR-β activation alone is sufficient to induce progressive renal fibrosis and failure, mimicking key aspects of chronic kidney disease in humans. Our data provide direct proof that fibrosis per se can drive chronic organ damage and establish a model of primary fibrosis allowing specific studies targeting fibrosis progression and regression.
Collapse
Affiliation(s)
- Eva M Buhl
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
- Electron Microscopy FacilityRWTH University of AachenAachenGermany
| | - Sonja Djudjaj
- Institute of PathologyRWTH University of AachenAachenGermany
| | | | - Katja Ermert
- Institute of PathologyRWTH University of AachenAachenGermany
| | - Victor G Puelles
- Division of NephrologyRWTH University of AachenAachenGermany
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of NephrologyMonash Health, and Center for Inflammatory DiseasesMonash UniversityMelbourneVic.Australia
| | - Maja T Lindenmeyer
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Clemens D Cohen
- Nephrological CenterMedical Clinic and Policlinic IVUniversity of MunichMunichGermany
| | - Chaoyong He
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Erawan Borkham‐Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research (IZKF)RWTH University of AachenAachenGermany
| | - Panuwat Trairatphisan
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Julio Saez‐Rodriguez
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Tobias B Huber
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Lorin E Olson
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Jürgen Floege
- Division of NephrologyRWTH University of AachenAachenGermany
| | - Peter Boor
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
| |
Collapse
|
34
|
Klinkhammer BM, Djudjaj S, Kunter U, Palsson R, Edvardsson VO, Wiech T, Thorsteinsdottir M, Hardarson S, Foresto-Neto O, Mulay SR, Moeller MJ, Jahnen-Dechent W, Floege J, Anders HJ, Boor P. Cellular and Molecular Mechanisms of Kidney Injury in 2,8-Dihydroxyadenine Nephropathy. J Am Soc Nephrol 2020; 31:799-816. [PMID: 32086278 DOI: 10.1681/asn.2019080827] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hereditary deficiency of adenine phosphoribosyltransferase causes 2,8-dihydroxyadenine (2,8-DHA) nephropathy, a rare condition characterized by formation of 2,8-DHA crystals within renal tubules. Clinical relevance of rodent models of 2,8-DHA crystal nephropathy induced by excessive adenine intake is unknown. METHODS Using animal models and patient kidney biopsies, we assessed the pathogenic sequelae of 2,8-DHA crystal-induced kidney damage. We also used knockout mice to investigate the role of TNF receptors 1 and 2 (TNFR1 and TNFR2), CD44, or alpha2-HS glycoprotein (AHSG), all of which are involved in the pathogenesis of other types of crystal-induced nephropathies. RESULTS Adenine-enriched diet in mice induced 2,8-DHA nephropathy, leading to progressive kidney disease, characterized by crystal deposits, tubular injury, inflammation, and fibrosis. Kidney injury depended on crystal size. The smallest crystals were endocytosed by tubular epithelial cells. Crystals of variable size were excreted in urine. Large crystals obstructed whole tubules. Medium-sized crystals induced a particular reparative process that we term extratubulation. In this process, tubular cells, in coordination with macrophages, overgrew and translocated crystals into the interstitium, restoring the tubular luminal patency; this was followed by degradation of interstitial crystals by granulomatous inflammation. Patients with adenine phosphoribosyltransferase deficiency showed similar histopathological findings regarding crystal morphology, crystal clearance, and renal injury. In mice, deletion of Tnfr1 significantly reduced tubular CD44 and annexin two expression, as well as inflammation, thereby ameliorating the disease course. In contrast, genetic deletion of Tnfr2, Cd44, or Ahsg had no effect on the manifestations of 2,8-DHA nephropathy. CONCLUSIONS Rodent models of the cellular and molecular mechanisms of 2,8-DHA nephropathy and crystal clearance have clinical relevance and offer insight into potential future targets for therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Thorsten Wiech
- Institute of Pathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; and
| | | | - Sverrir Hardarson
- Department of Pathology Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Shrikant R Mulay
- Division of Nephrology, Klinikum der Universität, LMU München, Munich, Germany
| | | | | | | | - Hans-Joachim Anders
- Division of Nephrology, Klinikum der Universität, LMU München, Munich, Germany
| | - Peter Boor
- Institute of Pathology, .,Division of Nephrology and Immunology.,Department of Electron Microscopy, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
35
|
Ito N, Sakamoto K, Hikichi C, Matsusaka T, Nagata M. Biphasic MIF and SDF1 expression during podocyte injury promote CD44-mediated glomerular parietal cell migration in focal segmental glomerulosclerosis. Am J Physiol Renal Physiol 2020; 318:F741-F753. [PMID: 32068458 DOI: 10.1152/ajprenal.00414.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glomerular parietal epithelial cell (PEC) activation, as revealed by de novo expression of CD44 and cell migration toward the injured filtration barrier, is a hallmark of podocyte injury-driven focal segmental glomerulosclerosis (FSGS). However, the signaling pathway that mediates activation of PECs in response to podocyte injury is unknown. The present study focused on CD44 signaling, particularly the roles of two CD44-related chemokines, migration inhibitory factor (MIF) and stromal cell-derived factor 1 (SDF1), and their common receptor, chemokine (C-X-C motif) receptor 4 (CXCR4), in the NEP25/LMB2 mouse podocyte-toxin model of FSGS. In the early phase of the disease, CD44-positive PECs were locally evident on the opposite side of the intact glomerular tuft and subsequently increased in the vicinity of synechiae with podocyte loss. Expression of MIF and SDF1 was first increased in injured podocytes and subsequently transferred to activated PECs expressing CD44 and CXCR4. In an immortalized mouse PEC (mPEC) line, recombinant MIF and SDF1 (rMIF and rSDF1, respectively) individually increased CD44 and CXCR4 mRNA and protein levels. rMIF and rSDF1 stimulated endogenous MIF and SDF1 production. rMIF- and rSDF1-induced mPEC migration was suppressed by CD44 siRNA. However, MIF and SDF1 inhibitors failed to show any impact on proteinuria, podocyte number, and CD44 expression in NEP25/LMB2 mice. Our data suggest that injured podocytes upregulate MIF and SDF1 that stimulate CD44 expression and CD44-mediated migration, which is enhanced by endogenous MIF and SDF1 in PECs. This biphasic expression pattern of the chemokine-CD44 axis in podocytes and PECs may be a novel mechanism of "podocyte-PEC cross-talk" signaling underlying podocyte injury-driven FSGS.
Collapse
Affiliation(s)
- Naoko Ito
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazuo Sakamoto
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Chihiro Hikichi
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Taiji Matsusaka
- Department of Basic Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Michio Nagata
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
36
|
Chen CA, Chang JM, Yang YL, Chang EE, Chen HC. Macrophage migration inhibitory factor regulates integrin-β1 and cyclin D1 expression via ERK pathway in podocytes. Biomed Pharmacother 2020; 124:109892. [PMID: 31986415 DOI: 10.1016/j.biopha.2020.109892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS Macrophage migration inhibitory factor (MIF) is found to increase in proliferative glomerulonephritis. MIF binds to the MIF receptor (CD74) that activates MAP kinase (ERK and p38). Integrins and cyclinD1 regulate cell proliferation, differentiation and adhesion. This study evaluates whether MIF can regulate integrin-β1/cyclin D1 expression and cell adhesion of podocytes. MAIN METHODS Expression of integrin-β1 mRNA/protein and cyclin D1 mRNA under stimulation of MIF was evaluated by real-time PCR and Western blotting. MIF receptor (CD74) and MAP kinase under MIF treatment were examined to determine which pathway regulated integrin-β1 and cyclin D1 expression. Cell adhesion was evaluated under MIF treatment and/or anti-integrin-β1 antibody by cell adhesion assay. KEY FINDINGS Protein levels of integrin-β1 were up-regulated under MIF treatment in a dosage-dependent manner. CD74 protein levels were not changed after MIF treatment. Integrin-β1 and cyclin D1 mRNA levels were up-regulated after MIF 100 ng/ml treatment. ERK inhibitor U0126 reduced MIF-induced the increase in integrin-β1 mRNA and protein expression following MIF stimulation. However, p38 inhibitor SB 203580 did not inhibit MIF-induced increase in integrin-β1 mRNA and protein expression following MIF stimulation. MIF-induced increase in cyclin D1 mRNA level also was inhibited only by U0126 following MIF stimulation. Podocyte adhesion was increased after MIF treatment, but, anti-integrin-β1 antibody decreased MIF-enhanced podocyte adhesion. SIGNIFICANCE MIF increases integrin-β1 and cyclin D1 expression through the ERK pathway in podocytes, and the up-regulated expression of integrin-β1 increases podocyte adhesion. These results provide further understanding for the role of MIF in developing proliferative glomerulonephritis.
Collapse
Affiliation(s)
- Chien-An Chen
- Department of Nephrology, Tainan Sinlau Hospital, Tainan, 701, Taiwan; Department of Health Care Administration, Chang Jung Christian University, Tainan, 711, Taiwan.
| | - Jer-Ming Chang
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Yu-Lin Yang
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, 703, Taiwan
| | - Eddy-Essen Chang
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hung-Chun Chen
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| |
Collapse
|
37
|
Matrix-assisted laser desorption/ionization mass spectrometry imaging to uncover protein alterations associated with the progression of IgA nephropathy. Virchows Arch 2019; 476:903-914. [PMID: 31838587 DOI: 10.1007/s00428-019-02705-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/27/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023]
Abstract
IgA nephropathy (IgAN) is one of the most diffuse glomerulonephrites worldwide, and many issues still remain regarding our understanding of its pathogenesis. The disease is diagnosed by renal biopsy examination, but potential pitfalls still persist with regard to discriminating its primary origin and, as a result, determining patient outcome remains challenging. In this pilot study, matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) was performed on renal biopsies obtained from patients with IgAN (n = 11) and other mesangioproliferative glomerulonephrites (MesPGN, n = 6) in order to enlighten proteomic alterations that may be associated with the progression of IgAN. Differences in the proteomic profiles of IgAN and MesPGN tissue could clearly be detected using this approach and, furthermore, 14 signals (AUC ≥ 0.8) were observed to have an altered intensity among the different CKD stages within the IgAN group. In particular, large increases in the intensity of these signals could be observed at CKD stages II and above. These signals primarily corresponded to proteins involved in either inflammatory and healing pathways and their increased intensity was localized within regions of tissue with large amounts of inflammatory cells or sclerosis. Despite much work in recent years, our molecular understanding of IgAN progression remains incomplete. This pilot study represents a promising starting point in the search for novel protein markers that can assist clinicians in better understanding the pathogenesis of IgAN and highlighting those patients who may progress to end-stage renal disease.
Collapse
|
38
|
Smeets B, Miesen L, Shankland SJ. CD9 Is a Novel Target in Glomerular Diseases Typified by Parietal Epithelial Cell Activation. Am J Kidney Dis 2019; 75:812-814. [PMID: 31668876 DOI: 10.1053/j.ajkd.2019.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 08/29/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Bart Smeets
- Department of Pathology, RIMLS, Radboudumc, Nijmegen, the Netherlands
| | - Laura Miesen
- Department of Pathology, RIMLS, Radboudumc, Nijmegen, the Netherlands
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
39
|
Majo J, Klinkhammer BM, Boor P, Tiniakos D. Pathology and natural history of organ fibrosis. Curr Opin Pharmacol 2019; 49:82-89. [PMID: 31671319 DOI: 10.1016/j.coph.2019.09.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/24/2019] [Indexed: 02/08/2023]
Abstract
Histopathological assessment of fibrosis focusing on morphological patterns provides important information for the management of patients with chronic diseases of the kidney, liver and the lung. This review summarizes key histopathological features of pulmonary, renal and hepatic fibrosis and discusses advances in the understanding of the pathogenesis of pulmonary fibrosis and pathogenetic insights with translational implications for renal fibrosis. The review also tackles new staging approaches based on liver fibrosis dynamics and evaluation of fibrosis regression, digital pathology and second harmonic generation microscopy methods for hepatic fibrosis assessment and critical appraisal of non-invasive tests for liver and renal fibrosis evaluation.
Collapse
Affiliation(s)
- Joaquim Majo
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Barbara Mara Klinkhammer
- Institute of Pathology & Department of Nephrology, University Clinic of RWTH, Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology & Department of Nephrology, University Clinic of RWTH, Aachen, Aachen, Germany
| | - Dina Tiniakos
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom; Department of Pathology, Aretaieion Hospital, Medical School, National & Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
40
|
Tu Y, Guo R, Li J, Wang S, Leng L, Deng J, Bucala R, Lu L. MiRNA Regulation of MIF in SLE and Attenuation of Murine Lupus Nephritis With miR-654. Front Immunol 2019; 10:2229. [PMID: 31608058 PMCID: PMC6761280 DOI: 10.3389/fimmu.2019.02229] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Objective: Macrophage Migration Inhibitory Factor (MIF) is involved in the pathogenesis of systemic lupus erythematosus (SLE) and lupus nephritis (LN). MicroRNAs (miRNAs) play important roles in LN but whether specific miRNAs regulate the expression of MIF in LN is unknown. We explore specific miRNAs that can regulate MIF expression, and investigate miR-654 for the treatment of experimentally-induced murine lupus nephritis. Methods: Sera samples from 24 SLE patients and 24 controls were collected to measure the MIF concentration and its correlation with disease activity. A luciferase reporter assay was used to explore the target of miR-654. ELISA was used to detect the downstream cytokines regulated by miR-654 and MIF. Western blot was applied to measure the impact of miR-654 inhibition on downstream MIF signaling. The therapeutic efficacy of miR-654 was tested in the pristine-induced lupus mouse model. We further measured miR-654 expression and analyzed its relationship with MIF expression in SLE patients. Results: The serum MIF level was increased in SLE patients (p < 0.001) and positively correlated with the SLEDAI score (r = 0.5473; p = 0.0056). MiR-654 inhibited MIF and downstream inflammatory cytokine production by selectively inhibiting the phosphorylation of ERK and AKT. Activation of miR-654 reduced IL-1β, IL-6, IL-8, and TNF-α production, reduced gomerulonephritis, and decreased MIF, IgG, and C3 expression in murine lupus glomeruli. Furthermore, MIF was negatively correlated with miR-654 expression (r = −0.4644; p = 0.0222) in SLE patients. Conclusion: MiR-654 negatively correlated with MIF and disease activity in patients with SLE. MiR-654 inhibits MIF expression via binding to MIF 3'UTR, selectively suppresses the phosphorylation of ERK and AKT, and reduces downstream inflammatory cytokine production. In vivo miR-654 treatment decreases MIF and downstream cytokine production and ameliorates murine lupus nephritis.
Collapse
Affiliation(s)
- Yang Tu
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ruru Guo
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Li
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Suli Wang
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Leng
- Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, United States
| | - Jun Deng
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard Bucala
- Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, United States
| | - Liangjing Lu
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
41
|
Kang I, Bucala R. The immunobiology of MIF: function, genetics and prospects for precision medicine. Nat Rev Rheumatol 2019; 15:427-437. [DOI: 10.1038/s41584-019-0238-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 01/01/2023]
|
42
|
Jankauskas SS, Wong DW, Bucala R, Djudjaj S, Boor P. Evolving complexity of MIF signaling. Cell Signal 2019; 57:76-88. [DOI: 10.1016/j.cellsig.2019.01.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 01/27/2023]
|
43
|
Breda PC, Wiech T, Meyer-Schwesinger C, Grahammer F, Huber T, Panzer U, Tiegs G, Neumann K. Renal proximal tubular epithelial cells exert immunomodulatory function by driving inflammatory CD4 + T cell responses. Am J Physiol Renal Physiol 2019; 317:F77-F89. [PMID: 31017008 DOI: 10.1152/ajprenal.00427.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In immune-mediated glomerular diseases like crescentic glomerulonephritis (cGN), inflammatory CD4+ T cells accumulate within the tubulointerstitial compartment in close contact to proximal and distal tubular epithelial cells and drive renal inflammation and tissue damage. However, whether renal epithelial cell populations play a role in the pathogenesis of cGN by modulating CD4+ T cell responses is less clear. In the present study, we aimed to investigate the potential of renal epithelial cells to function as antigen-presenting cells, thereby stimulating CD4+ T cell responses. Using a FACS-based protocol that allowed comparative analysis of cortical epithelial cell populations, we showed that particularly proximal tubular epithelial cells (PTECs) express molecules linked with antigen-presenting cell function, including major histocompatibility complex class II (MHCII), CD74, CD80, and CD86 in homeostasis and nephrotoxic nephritis, a murine model of cGN. Protein expression was visualized at the PTEC single cell level by imaging flow cytometry. Interestingly, we found inflammation-dependent regulation of epithelium-expressed CD74, CD80, and CD86, whereas MHCII expression was not altered. Antigen-specific stimulation of CD4+ T cells by PTECs in vitro supported CD4+ T cell survival and induced CD4+ T cell activation, proliferation, and inflammatory cytokine production. In patients with antineutrophil cytoplasmic antibody-associated glomerulonephritis, MHCII and CD74 were expressed by both proximal and distal tubules, whereas CD86 was predominantly expressed by proximal tubules. Thus, particularly PTECs have the potential to induce an inflammatory phenotype in CD4+ T cells in vitro, which might also play a role in the pathology of immune-mediated kidney disease.
Collapse
Affiliation(s)
- Philippe Christophe Breda
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Thorsten Wiech
- Institute of Pathology, University Hospital Eppendorf , Hamburg , Germany
| | | | - Florian Grahammer
- III, Medical Clinic University Hospital Eppendorf , Hamburg , Germany
| | - Tobias Huber
- III, Medical Clinic University Hospital Eppendorf , Hamburg , Germany
| | - Ulf Panzer
- III, Medical Clinic University Hospital Eppendorf , Hamburg , Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
44
|
Li JH, Tang Y, Lv J, Wang XH, Yang H, Tang PMK, Huang XR, He ZJ, Zhou ZJ, Huang QY, Klug J, Meinhardt A, Fingerle-Rowson G, Xu AP, Zheng ZH, Lan HY. Macrophage migration inhibitory factor promotes renal injury induced by ischemic reperfusion. J Cell Mol Med 2019; 23:3867-3877. [PMID: 30968541 PMCID: PMC6533527 DOI: 10.1111/jcmm.14234] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/27/2018] [Accepted: 12/16/2018] [Indexed: 11/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is pleiotropic cytokine that has multiple effects in many inflammatory and immune diseases. This study reveals a potential role of MIF in acute kidney injury (AKI) in patients and in kidney ischemic reperfusion injury (IRI) mouse model in MIF wild‐type (WT) and MIF knockout (KO) mice. Clinically, plasma and urinary MIF levels were largely elevated at the onset of AKI, declined to normal levels when AKI was resolved and correlated tightly with serum creatinine independent of disease causes. Experimentally, MIF levels in plasma and urine were rapidly elevated after IRI‐AKI and associated with the elevation of serum creatinine and the severity of tubular necrosis, which were suppressed in MIF KO mice. It was possible that MIF may mediate AKI via CD74/TLR4‐NF‐κB signalling as mice lacking MIF were protected from AKI by largely suppressing CD74/TLR‐4‐NF‐κB associated renal inflammation, including the expression of MCP‐1, TNF‐α, IL‐1β, IL‐6, iNOS, CXCL15(IL‐8 in human) and infiltration of macrophages, neutrophil, and T cells. In conclusion, our study suggests that MIF may be pathogenic in AKI and levels of plasma and urinary MIF may correlate with the progression and regression of AKI.
Collapse
Affiliation(s)
- Jin H Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.,Department of Medicine and Therapeutics, Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Ying Tang
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun Lv
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao H Wang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Hui Yang
- Department of Medicine and Therapeutics, Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China.,Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Patrick M K Tang
- Department of Medicine and Therapeutics, Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiao R Huang
- Department of Medicine and Therapeutics, Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Zhi J He
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zi J Zhou
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiu Y Huang
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Günter Fingerle-Rowson
- Department I of Internal Medicine, University Hospital Cologne, and Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - An P Xu
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhi H Zheng
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Kuppe C, Leuchtle K, Wagner A, Kabgani N, Saritas T, Puelles VG, Smeets B, Hakroush S, van der Vlag J, Boor P, Schiffer M, Gröne HJ, Fogo A, Floege J, Moeller MJ. Novel parietal epithelial cell subpopulations contribute to focal segmental glomerulosclerosis and glomerular tip lesions. Kidney Int 2019; 96:80-93. [PMID: 31029503 PMCID: PMC7292612 DOI: 10.1016/j.kint.2019.01.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
Abstract
Beside the classical flat parietal epithelial cells (PECs), we investigated proximal tubular epithelial-like cells, a neglected subgroup of PECs. These cells, termed cuboidal PECs, make up the most proximal part of the proximal tubule and may also line parts of Bowman's capsule. Additionally, a third intermediate PEC subgroup was identified at the junction between the flat and cuboidal PEC subgroups at the tubular orifice. The transgenic mouse line PEC-rtTA labeled all three PEC subgroups. Here we show that the inducible Pax8-rtTA mouse line specifically labeled only cuboidal and intermediate PECs, but not flat PECs. In aging Pax8-rtTA mice, cell fate mapping showed no evidence for significant transdifferentiation from flat PECs to cuboidal or intermediate PECs or vice versa. In murine glomerular disease models of crescentic glomerulonephritis, and focal segmental glomerulosclerosis (FSGS), intermediate PECs became more numerous. These intermediate PECs preferentially expressed activation markers CD44 and Ki-67, suggesting that this subgroup of PECs was activated more easily than the classical flat PECs. In mice with FSGS, cuboidal and intermediate PECs formed sclerotic lesions. In patients with FSGS, cells forming the tip lesions expressed markers of intermediate PECs. These novel PEC subgroups form sclerotic lesions and were more prone to cellular activation compared to the classical flat PECs in disease. Thus, colonization of Bowman's capsule by cuboidal PECs may predispose to lesion formation and chronic kidney disease. We propose that tip lesions originate from this novel subgroup of PECs in patients with FSGS.
Collapse
Affiliation(s)
- Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| | - Katja Leuchtle
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Anton Wagner
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nazanin Kabgani
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Turgay Saritas
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Victor G Puelles
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Cardiovascular Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, School of Biomedical Sciences, and Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia; Department of Nephrology, Monash Health, Melbourne Australia
| | - Bart Smeets
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Samy Hakroush
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, University of Erlangen, Erlangen, Germany
| | - Hermann-Josef Gröne
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Agnes Fogo
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Marcus Johannes Moeller
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| |
Collapse
|
46
|
Abstract
BACKGROUND Macrophage migration-inhibitory factor (MIF) is a cytokine best known for its proinflammatory and disease-aggravating role in a number of conditions, including atherosclerosis, autoimmune diseases, sepsis, and glomerulonephritides. OBJECTIVES In our studies we aimed to define the role of MIF on local renal resident cells, in particular the renal epithelium. RESULTS We have shown that MIF exerts local effects on glomerular cells, in particular the parietal epithelial cells and mesangial cells, promoting their pathological proliferation and aggravating disease course of a murine model of immune-mediated glomerulonephritis. In contrast, in a large set of animal and in vitro experiments, we have shown that in the setting of chronic kidney disease, MIF had an unexpected and potent antifibrotic and anti-inflammatory effect. This was mediated by enhanced regeneration and reduced cell-cycle arrest of tubular epithelial cells. Finally, in a combined approach using clinical studies, animal models, and in vitro experiments, we have shown that MIF is also renoprotective in the setting of acute kidney injury. In this setting, MIF-modulated programmed cell death of tubular cells and thereby reduced necroinflammation and kidney injury. CONCLUSIONS Taken together, MIF has a dual role in kidney diseases, promoting (auto)immune glomerular diseases and limiting tubular cell injury in the setting of acute and chronic kidney diseases. These data suggest potential safety issues of systemic MIF targeted therapies, but also open new therapeutic options by targeting MIF or its analogues to tubular cells.
Collapse
Affiliation(s)
- P Boor
- Institute of Pathology, University Clinic, RWTH University of Aachen, Pauwelsstr. 30, 52074, Aachen, Germany. .,Division of Nephrology, RWTH University of Aachen, Aachen, Germany.
| |
Collapse
|
47
|
|
48
|
[MIF in kidney diseases : A story of Dr. Jekyll and Mr. Hyde (German version)]. DER PATHOLOGE 2018; 39:177-184. [PMID: 30406830 DOI: 10.1007/s00292-018-0535-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Macrophage migration-inhibitory factor (MIF) is a cytokine best known for its proinflammatory and disease-aggravating role in a number of conditions, including atherosclerosis, autoimmune diseases, sepsis, and glomerulonephritides. OBJECTIVES In our studies we aimed to define the role of MIF on local renal resident cells, in particular the renal epithelium. RESULTS We have shown that MIF exerts local effects on glomerular cells, in particular the parietal epithelial cells and mesangial cells, promoting their pathological proliferation and aggravating disease course of a murine model of immune-mediated glomerulonephritis. In contrast, in a large set of animal and in vitro experiments, we have shown that in the setting of chronic kidney disease, MIF had an unexpected and potent antifibrotic and anti-inflammatory effect. This was mediated by enhanced regeneration and reduced cell-cycle arrest of tubular epithelial cells. Finally, in a combined approach using clinical studies, animal models, and in vitro experiments, we have shown that MIF is also renoprotective in the setting of acute kidney injury. In this setting, MIF-modulated programmed cell death of tubular cells and thereby reduced necroinflammation and kidney injury. CONCLUSIONS Taken together, MIF has a dual role in kidney diseases, promoting (auto)immune glomerular diseases and limiting tubular cell injury in the setting of acute and chronic kidney diseases. These data suggest potential safety issues of systemic MIF targeted therapies, but also open new therapeutic options by targeting MIF or its analogues to tubular cells.
Collapse
|
49
|
Moellmann J, Klinkhammer BM, Onstein J, Stöhr R, Jankowski V, Jankowski J, Lebherz C, Tacke F, Marx N, Boor P, Lehrke M. Glucagon-Like Peptide 1 and Its Cleavage Products Are Renoprotective in Murine Diabetic Nephropathy. Diabetes 2018; 67:2410-2419. [PMID: 30104246 DOI: 10.2337/db17-1212] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/27/2018] [Indexed: 11/13/2022]
Abstract
Incretin-based therapies, including glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors, are potent glucose-lowering drugs. Still, only GLP-1 receptor agonists with close peptide homology to GLP-1 (liraglutide and semaglutide) but neither exenatide-based GLP-1 receptor agonists nor DPP-4 inhibitors were found to reduce cardiovascular events. This different response might relate to GLP-1 receptor-independent actions of GLP-1 caused by cleavage products only liberated by GLP-1 receptor agonists with close peptide structure to GLP-1. To test this hypothesis, we directly compared metabolic, renal, and cardiac effects of GLP-1 and its cleavage products in diabetic db/db mice. Using an adeno-associated viral vector system, we overexpressed DPP-4-resistant GLP-1 (7-37 Mut8) and the two GLP-1 cleavage products, GLP-1 (9-37) and GLP-1 (28-37), in diabetic db/db mice. Only GLP-1 (7-37 Mut8), but none of the cleavage products, significantly improved glucose metabolism. Still, all GLP-1 constructs significantly reduced tubulointerstitial renal damage, lowered expression of the tubular injury markers, and attenuated renal accumulation of macrophages and T cells. This was associated with a systemic immunomodulatory effect, which was similarly found in an acute renal ischemia/reperfusion injury model. In conclusion, GLP-1 cleavage products proved sufficient to mediate organ-protective effects, which might help to explain differences between GLP-1 receptor agonists.
Collapse
Affiliation(s)
- Julia Moellmann
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Julia Onstein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Robert Stöhr
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Corinna Lebherz
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Internal Medicine II, Nephrology and Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Electron Microscopy Facility, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
50
|
Sreih AG, Ezzedine R, Leng L, Fan J, Yao J, Reid D, Piecychna M, Carette S, Cuthbertson D, Dellaripa P, Hoffman GS, Khalidi NA, Koening CL, Langford CA, Mahr A, McAlear CA, Maksimowicz-Mckinnon K, Monach PA, Seo P, Specks U, St Clair EW, Stone JH, Ytterberg SR, Edberg J, Merkel PA, Bucala R. Role of Macrophage Migration Inhibitory Factor in Granulomatosis With Polyangiitis. Arthritis Rheumatol 2018; 70:2077-2086. [PMID: 29953750 DOI: 10.1002/art.40655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/26/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To examine the association between macrophage migration inhibitory factor (MIF) promoter polymorphisms and granulomatosis with polyangiitis (GPA) in human subjects, and to assess the role of MIF in a murine model of granulomatous vasculitis. METHODS The human study involved 1,077 patients with GPA and healthy controls whose serum was genotyped by capillary electrophoresis for the MIF -794 CATT5-8 promoter microsatellite (rs5844572). MIF promoter, CATT-length-dependent gene expression in response to β-glucan was assessed by gene reporter assays. In mouse studies, granulomatous disease was induced by injection of Candida albicans β-glucan into wild-type (WT) or Mif-knockout (Mif-KO) C57BL/6 mice and C57BL/6 mice transgenically overexpressing Mif in lung epithelium (Mif lung-Tg2.1). Mice were treated with a neutralizing anti-MIF antibody and analyzed for the density of pulmonary granulomas, expression of inflammatory chemokines, and frequency of mortality. RESULTS The percentage of human subjects carrying >5 CATT repeats in each MIF allele (high genotypic MIF expressers) was 60.2% among patients with GPA and 53.9% among healthy controls (adjusted P = 0.049). In response to granulomatous stimulation, human MIF gene expression increased proportionally with CATT length. Mif lung-Tg2.1 mice exhibited more pulmonary granulomas than WT mice, which in turn showed more granulomas than Mif-KO mice. A significantly higher percentage of Mif lung-Tg2.1 mice, compared to Mif-KO or WT mice, died when injected with Candida albicans β-glucan, and treatment of these mice with an anti-MIF monoclonal antibody protected against a lethal outcome. Levels of MIF-dependent neutrophil/macrophage chemokines were elevated in the bronchoalveolar lavage fluid or plasma of Mif lung-Tg2.1 mice. CONCLUSION Patients with GPA have an increased frequency of high MIF expression CATT alleles. Higher Mif expression increases the incidence of mortality and pulmonary granulomas in Mif lung-Tg2.1 mice, while anti-MIF treatment protects these mice against death. Blockade of MIF in high genotypic MIF expressers may therefore offer a selective pharmacologic therapy for GPA.
Collapse
Affiliation(s)
| | | | - Lin Leng
- Yale School of Medicine, New Haven, Connecticut
| | - Juan Fan
- Yale School of Medicine, New Haven, Connecticut
| | - Jie Yao
- Yale School of Medicine, New Haven, Connecticut
| | - Duncan Reid
- Yale School of Medicine, New Haven, Connecticut
| | | | - Simon Carette
- Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Paul Dellaripa
- Brigham and Women's Hospital, and Harvard University, Boston, Massachusetts
| | | | - Nader A Khalidi
- St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | - Philip Seo
- Johns Hopkins University, Baltimore, Maryland
| | - Ulrich Specks
- Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | - John H Stone
- Massachusetts General Hospital and Harvard University, Boston, Massachusetts
| | | | | | | | | |
Collapse
|