1
|
Cara-Fuentes G, Verma R, Venkatareddy M, Bauer C, Piani F, Aksoy ST, Vazzalwar N, Garcia GE, Banks M, Ordoñez FA, de Lucas-Collantes C, Bjornstad P, González Rodríguez JD, Johnson RJ, Garg P. β1-Integrin blockade prevents podocyte injury in experimental models of minimal change disease. Nefrologia 2024; 44:90-99. [PMID: 37150673 DOI: 10.1016/j.nefroe.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/10/2022] [Indexed: 05/09/2023] Open
Abstract
INTRODUCTION Activation of the focal adhesion kinase (FAK) in podocytes is involved in the pathogenesis of minimal change disease (MCD), but the pathway leading to its activation in this disease is unknown. Here, we tested whether podocyte β1 integrin is the upstream modulator of FAK activation and podocyte injury in experimental models of MCD-like injury. METHODS We used lipopolysaccharide (LPS) and MCD sera to induce MCD-like changes in vivo and in cultured human podocytes, respectively. We performed functional studies using specific β1 integrin inhibitors in vivo and in vitro, and integrated histological analysis, western blotting, and immunofluorescence to assess for morphological and molecular changes in podocytes. By ELISA, we measured serum LPS levels in 35 children with MCD or presumed MCD (idiopathic nephrotic syndrome [INS]) and in 18 healthy controls. RESULTS LPS-injected mice showed morphological (foot process effacement, and normal appearing glomeruli on light microscopy) and molecular features (synaptopodin loss, nephrin mislocalization, FAK phosphorylation) characteristic of human MCD. Administration of a β1 integrin inhibitor to mice abrogated FAK phosphorylation, and ameliorated proteinuria and podocyte injury following LPS. Children with MCD/INS in relapse had higher serum LPS levels than controls. In cultured human podocytes, β1 integrin blockade prevented cytoskeletal rearrangements following exposure to MCD sera in relapse. CONCLUSIONS Podocyte β1 integrin activation is an upstream mediator of FAK phosphorylation and podocyte injury in models of MCD-like injury.
Collapse
|
2
|
Kraus A, Rose V, Krüger R, Sarau G, Kling L, Schiffer M, Christiansen S, Müller-Deile J. Characterizing Intraindividual Podocyte Morphology In Vitro with Different Innovative Microscopic and Spectroscopic Techniques. Cells 2023; 12:cells12091245. [PMID: 37174644 PMCID: PMC10177567 DOI: 10.3390/cells12091245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/14/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Podocytes are critical components of the glomerular filtration barrier, sitting on the outside of the glomerular basement membrane. Primary and secondary foot processes are characteristic for podocytes, but cell processes that develop in culture were not studied much in the past. Moreover, protocols for diverse visualization methods mostly can only be used for one technique, due to differences in fixation, drying and handling. However, we detected by single-cell RNA sequencing (scRNAseq) analysis that cells reveal high variability in genes involved in cell type-specific morphology, even within one cell culture dish, highlighting the need for a compatible protocol that allows measuring the same cell with different methods. Here, we developed a new serial and correlative approach by using a combination of a wide variety of microscopic and spectroscopic techniques in the same cell for a better understanding of podocyte morphology. In detail, the protocol allowed for the sequential analysis of identical cells with light microscopy (LM), Raman spectroscopy, scanning electron microscopy (SEM) and atomic force microscopy (AFM). Skipping the fixation and drying process, the protocol was also compatible with scanning ion-conductance microscopy (SICM), allowing the determination of podocyte surface topography of nanometer-range in living cells. With the help of nanoGPS Oxyo®, tracking concordant regions of interest of untreated podocytes and podocytes stressed with TGF-β were analyzed with LM, SEM, Raman spectroscopy, AFM and SICM, and revealed significant morphological alterations, including retraction of podocyte process, changes in cell surface morphology and loss of cell-cell contacts, as well as variations in lipid and protein content in TGF-β treated cells. The combination of these consecutive techniques on the same cells provides a comprehensive understanding of podocyte morphology. Additionally, the results can also be used to train automated intelligence networks to predict various outcomes related to podocyte injury in the future.
Collapse
Affiliation(s)
- Annalena Kraus
- Institute for Nanotechnology and Correlative Microscopy, INAM, 91301 Forchheim, Germany
| | - Victoria Rose
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - René Krüger
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - George Sarau
- Institute for Nanotechnology and Correlative Microscopy, INAM, 91301 Forchheim, Germany
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, 91301 Forchheim, Germany
- Leuchs Emeritus Group, Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
| | - Lasse Kling
- Institute for Nanotechnology and Correlative Microscopy, INAM, 91301 Forchheim, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Research Center on Rare Kidney Diseases (RECORD), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Silke Christiansen
- Institute for Nanotechnology and Correlative Microscopy, INAM, 91301 Forchheim, Germany
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS, 91301 Forchheim, Germany
- Physics Department, Freie Universität Berlin, 14195 Berlin, Germany
| | - Janina Müller-Deile
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Research Center on Rare Kidney Diseases (RECORD), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
3
|
Li D, Liu L, Murea M, Freedman BI, Ma L. Bioinformatics Analysis Reveals a Shared Pathway for Common Forms of Adult Nephrotic Syndrome. KIDNEY360 2023; 4:e515-e524. [PMID: 36763793 PMCID: PMC10278839 DOI: 10.34067/kid.0000000000000074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/20/2023] [Indexed: 02/12/2023]
Abstract
Key Points Dysregulation of the focal adhesion pathway is present in the three most common forms of glomerular disease, that is, Focal segmental glomerulosclerosis, membranous nephropathy, and minimal change disease. Zyxin is seen to be upregulated in the glomerular compartment of patients with the three most common forms of glomerular disease. Background Focal segmental glomerulosclerosis, membranous nephropathy, and minimal change disease are common causes of nephrotic syndrome. Although triggers for these diseases differ, disease progression may share common molecular mechanisms. The aim of this study was to investigate the presence of molecular pathways that are dysregulated across these glomerular diseases. Methods The gene expression dataset GSE200828 from the Nephrotic Syndrome Study Network study was obtained from the Gene Expression Omnibus database. R and Python packages, Cytoscape software, and online tools (DAVID and STRING) were used to identify core genes and topologically relevant nodes and molecular pathways. Single-cell RNA sequencing analysis was applied to identify the expression patterns of core genes across kidney cell types in glomerular compartments. Results A total of 1087 differentially expressed genes were identified, including 691 upregulated genes and 396 downregulated genes, which are common in all three forms of nephrotic syndrome compared with kidney donor controls (FDR P <0.01). A multiapproach bioinformatics analysis narrowed down to 28 similarly dysregulated genes across the three proteinuric glomerulopathies. The most topologically relevant nodes belonged to the adherens junction, focal adhesion, and cytoskeleton pathways, where zyxin covers all of those gene ontology terms. Conclusions We report that dysregulation of cell adhesion complexes was present in the three most common forms of glomerular disease. Zyxin could be a biomarker in all three common forms of nephrotic syndrome. If further functional studies confirm its role in their development, zyxin could be a potential therapeutic target.
Collapse
Affiliation(s)
- DengFeng Li
- Informatics and Analytics, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Liang Liu
- Bioinformatics Shared Resource, Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Mariana Murea
- Department of Internal Medicine—Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Barry I. Freedman
- Department of Internal Medicine—Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Lijun Ma
- Department of Internal Medicine—Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
4
|
VEGFA promotes the occurrence of PLA2R-associated idiopathic membranous nephropathy by angiogenesis via the PI3K/AKT signalling pathway. BMC Nephrol 2022; 23:313. [PMID: 36114523 PMCID: PMC9482157 DOI: 10.1186/s12882-022-02936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background The M-type phospholipase A2 receptor (PLA2R)-associated idiopathic membranous nephropathy (IMN) is a common immune-related disease in adults. Vascular endothelial growth factor A (VEGFA) is the key mediator of angiogenesis, which leads to numerous kidney diseases. However, the role of VEGFA in IMN is poorly understood. Methods In the present study, we downloaded the microarray data GSE115857 from Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) were identified with R software. The cytoHubba plug-in were used to identify hub genes from the protein–protein interaction network. Gene set enrichment analysis (GSEA) was used to identify signalling pathway in IMN. CCK8 was performed to assess the cell viability in human vascular endothelial cells (HVECs). Then, passive Heymann nephritis (PHN) was induced in rats by a single tail vein injection of anti-Fx1A antiserum. Animals treated with VEGFA inhibitor bevacizumab (BV), with saline as a positive control. Proteinuria was evaluated by biochemical measurements. Immunohistochemistry and immunofluorescence was used to evaluate relative proteins expression. Electron microscopy was performed to observe the thickness of the glomerular basement membrane (GBM). Results We revealed 3 hub genes, including one up-regulated gene VEGFA and two down-regulated genes JUN and FOS, which are closely related to the development of PLA2R-associated IMN. Pathway enrichment analysis found that the biological process induced by VEGFA is associated with PI3K/Akt signalling. GSEA showed that the signalling pathway of DEGs in GSE115857 was focused on angiogenesis, in which VEGFA acts as a core gene. We confirmed the high expression of VEGFA, PI3K, and AKT in IMN renal biopsy samples with immunohistochemistry. In HVECs, we found that BV suppresses cell viability in a time and dose dependent manner. In vivo, we found low dose of BV attenuates proteinuria via inhibiting VEGFA/PI3K/AKT signalling. Meanwhile, low dose of BV alleviates the thickening of the GBM. Conclusion VEGFA/PI3K/AKT signalling may play significant roles in the pathogenesis of IMN, which may provide new targets for the treatment of IMN. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02936-y.
Collapse
|
5
|
Cara-Fuentes G, Andres-Hernando A, Bauer C, Banks M, Garcia GE, Cicerchi C, Kuwabara M, Shimada M, Johnson RJ, Lanaspa MA. Pulmonary surfactants and the respiratory-renal connection in steroid-sensitive nephrotic syndrome of childhood. iScience 2022; 25:104694. [PMID: 35847557 PMCID: PMC9284382 DOI: 10.1016/j.isci.2022.104694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/23/2022] [Accepted: 06/24/2022] [Indexed: 01/21/2023] Open
Abstract
Steroid-sensitive nephrotic syndrome (SSNS) in childhood is usually due to minimal change disease (MCD). Unlike many glomerular conditions, SSNS/MCD is commonly precipitated by respiratory infections. Of interest, pulmonary inflammation releases surfactants in circulation which are soluble agonists of SIRPα, a podocyte receptor that regulates integrin signaling. Here, we characterized this pulmonary-renal connection in MCD and performed studies to determine its importance. Children with SSNS/MCD in relapse but not remission had elevated plasma surfactants and urinary SIRPα. Sera from relapsing subjects triggered podocyte SIRPα signaling via tyrosine phosphatase SHP-2 and nephrin dephosphorylation, a marker of podocyte activation. Further, addition of surfactants to MCD sera from patients in remission replicated these findings. Similarly, nasal instillation of toll-like receptor 3 and 4 agonists in mice resulted in elevated serum surfactants and their binding to glomeruli triggering proteinuria. Together, our data document a critical pulmonary-podocyte signaling pathway involving surfactants and SIRPα signaling in SSNS/MCD.
Collapse
Affiliation(s)
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA,Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, OR, USA
| | - Colin Bauer
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Mindy Banks
- Rocky Mountain Pediatric Kidney Center, Denver, CO, USA
| | - Gabriela E. Garcia
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Christina Cicerchi
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Masanari Kuwabara
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Michiko Shimada
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA,Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, OR, USA,Corresponding author
| |
Collapse
|
6
|
Ye Q, Lan B, Liu H, Persson PB, Lai EY, Mao J. A critical role of the podocyte cytoskeleton in the pathogenesis of glomerular proteinuria and autoimmune podocytopathies. Acta Physiol (Oxf) 2022; 235:e13850. [PMID: 35716094 DOI: 10.1111/apha.13850] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/23/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023]
Abstract
Selective glomerular filtration relies on the membrane separating the glomerular arterioles from the Bowman space. As a major component of the glomerular filtration barrier, podocytes form foot processes by the actin cytoskeleton, which dynamically adjusts in response to environmental changes to maintain filtration barrier integrity. The slit diaphragms bridge the filtration slits between neighboring foot processes and act as signaling hubs interacting with the actin cytoskeleton. Focal adhesions relay signals to regulate actin dynamics while allowing podocyte adherence to the basement membrane. Mutations in actin regulatory and signaling proteins may disrupt the actin cytoskeleton, resulting in foot process retraction, effacement, and proteinuria. Large-scale gene expression profiling platforms, transgenic animal models, and other in vivo gene delivery methods now enhance our understanding of the interactions among podocyte focal adhesions, slit diaphragms, and actin dynamics. In addition, our team found that at least 66% of idiopathic nephrotic syndrome (INS) children have podocyte autoantibodies, which was defined as a new disease subgroup-, autoimmune podocytopathies. This review outlines the pathophysiological mechanisms of podocyte cytoskeleton protein interactions in proteinuria and glomerular podocytopathy.
Collapse
Affiliation(s)
- Qing Ye
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Bing Lan
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Huihui Liu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Translational Physiology, Berlin, Germany
| | - En Yin Lai
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Translational Physiology, Berlin, Germany.,Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| |
Collapse
|
7
|
Sasaki H, Sasaki N. Tensin 2-deficient nephropathy - mechanosensitive nephropathy, genetic susceptibility. Exp Anim 2022; 71:252-263. [PMID: 35444113 PMCID: PMC9388341 DOI: 10.1538/expanim.22-0031] [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] [Indexed: 11/30/2022] Open
Abstract
Tensin 2 (TNS2), a focal adhesion protein, is considered to anchor focal adhesion proteins to β integrin as an integrin adaptor protein and/or serve as a scaffold to facilitate the
interactions of these proteins. In the kidney, TNS2 localizes to the basolateral surface of glomerular epithelial cells, i.e., podocytes. Loss of TNS2 leads to the development of glomerular
basement membrane lesions and abnormal accumulation of extracellular matrix in maturing glomeruli during the early postnatal stages. It subsequently results in podocyte foot process
effacement, eventually leading to glomerulosclerosis. Histopathological features of the affected glomeruli in the middle stage of the disease include expansion of the mesangial matrix
without mesangial cell proliferation. In this review, we provide an overview of TNS2-deficient nephropathy and discuss the potential mechanism underlying this mechanosensitive nephropathy,
which may be applicable to other glomerulonephropathies, such as CD151-deficient nephropathy and Alport syndrome. The onset of TNS2-deficient nephropathy strictly depends on the genetic
background, indicating the presence of critical modifier genes. A better understanding of molecular mechanisms of mechanosensitive nephropathy may open new avenues for the management of
patients with glomerulonephropathies.
Collapse
Affiliation(s)
- Hayato Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University
| |
Collapse
|
8
|
Maywald ML, Picciotto C, Lepa C, Bertgen L, Yousaf FS, Ricker A, Klingauf J, Krahn MP, Pavenstädt H, George B. Rap1 Activity Is Essential for Focal Adhesion and Slit Diaphragm Integrity. Front Cell Dev Biol 2022; 10:790365. [PMID: 35372328 PMCID: PMC8972170 DOI: 10.3389/fcell.2022.790365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
Glomerular podocytes build, with their intercellular junctions, part of the kidney filter. The podocyte cell adhesion protein, nephrin, is essential for developing and maintaining slit diaphragms as functional loss in humans results in heavy proteinuria. Nephrin expression and function are also altered in many adult-onset glomerulopathies. Nephrin signals from the slit diaphragm to the actin cytoskeleton and integrin β1 at focal adhesions by recruiting Crk family proteins, which can interact with the Rap guanine nucleotide exchange factor 1 C3G. As Rap1 activity affects focal adhesion formation, we hypothesize that nephrin signals via Rap1 to integrin β. To address this issue, we combined Drosophila in vivo and mammalian cell culture experiments. We find that Rap1 is necessary for correct targeting of integrin β to focal adhesions in Drosophila nephrocytes, which also form slit diaphragm-like structures. In the fly, the Rap1 activity is important for signaling of the nephrin ortholog to integrin β, as well as for nephrin-dependent slit diaphragm integrity. We show by genetic interaction experiments that Rap1 functions downstream of nephrin signaling to integrin β and downstream of nephrin signaling necessary for slit diaphragm integrity. Similarly, in human podocyte culture, nephrin activation results in increased activation of Rap1. Thus, Rap1 is necessary for downstream signal transduction of nephrin to integrin β.
Collapse
Affiliation(s)
- Mee-Ling Maywald
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Cara Picciotto
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Carolin Lepa
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Luisa Bertgen
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | | | - Andrea Ricker
- Institute of Medical Physics and Biophysics, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Michael P. Krahn
- Medizinische Klinik D, Medical Cell Biology, University Hospital Münster, Münster, Germany
| | | | - Britta George
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
- *Correspondence: Britta George,
| |
Collapse
|
9
|
Purohit S, Piani F, Ordoñez FA, de Lucas-Collantes C, Bauer C, Cara-Fuentes G. Molecular Mechanisms of Proteinuria in Minimal Change Disease. Front Med (Lausanne) 2022; 8:761600. [PMID: 35004732 PMCID: PMC8733331 DOI: 10.3389/fmed.2021.761600] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Minimal change disease (MCD) is the most common type of idiopathic nephrotic syndrome in childhood and represents about 15% cases in adults. It is characterized by massive proteinuria, edema, hypoalbuminemia, and podocyte foot process effacement on electron microscopy. Clinical and experimental studies have shown an association between MCD and immune dysregulation. Given the lack of inflammatory changes or immunocomplex deposits in the kidney tissue, MCD has been traditionally thought to be mediated by an unknown circulating factor(s), probably released by T cells that directly target podocytes leading to podocyte ultrastructural changes and proteinuria. Not surprisingly, research efforts have focused on the role of T cells and podocytes in the disease process. Nevertheless, the pathogenesis of the disease remains a mystery. More recently, B cells have been postulated as an important player in the disease either by activating T cells or by releasing circulating autoantibodies against podocyte targets. There are also few reports of endothelial injury in MCD, but whether glomerular endothelial cells play a role in the disease remains unexplored. Genome-wide association studies are providing insights into the genetic susceptibility to develop the disease and found a link between MCD and certain human haplotype antigen variants. Altogether, these findings emphasize the complex interplay between the immune system, glomerular cells, and the genome, raising the possibility of distinct underlying triggers and/or mechanisms of proteinuria among patients with MCD. The heterogeneity of the disease and the lack of good animal models of MCD remain major obstacles in the understanding of MCD. In this study, we will review the most relevant candidate mediators and mechanisms of proteinuria involved in MCD and the current models of MCD-like injury.
Collapse
Affiliation(s)
- Shrey Purohit
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Pediatrics, Section of Pediatric Nephrology, Children's Hospital Colorado, Aurora, CO, United States
| | - Federica Piani
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Medicine and Surgery Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Flor A Ordoñez
- Division of Pediatric Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Colin Bauer
- Department of Pediatrics, Section of Pediatric Nephrology, Children's Hospital Colorado, Aurora, CO, United States
| | - Gabriel Cara-Fuentes
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Pediatrics, Section of Pediatric Nephrology, Children's Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
10
|
ElShaer A, Almasry M, Alawar M, Masoud H, El Kinge AR. Dasatinib-Induced Nephrotic Syndrome: A Case Report. Cureus 2021; 13:e20330. [PMID: 34912656 PMCID: PMC8665416 DOI: 10.7759/cureus.20330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 12/22/2022] Open
Abstract
Second-generation tyrosine kinase inhibitors (TKI), such as nilotinib and dasatinib, are used in the first-line treatment of chronic myeloid leukemia (CML), usually after the failure or resistance to imatinib. Despite a good safety profile, medications in this category have an increased incidence of specific adverse events such as pulmonary hypertension, pleural effusion, and cardiovascular/peripheral arterial events. However, renal complications are rarely reported and observed. We herein report a case of a 46-year-old patient with CML who developed nephrotic syndrome upon switching from imatinib to dasatinib therapy, with the resolution of symptoms upon treatment discontinuation and switching to nilotinib. Limited cases were reported in the literature. It is thought that the inhibition of the vascular endothelial growth factor (VEGF) pathway is the main mechanism leading to proteinuria. Dasatinib-induced nephrotic syndrome should be looked for as it can be resolved by either reducing the dose or stopping it altogether and switching to another TKI.
Collapse
Affiliation(s)
- Ahmed ElShaer
- Internal Medicine, Alfaisal University College of Medicine, Riyadh, SAU
| | - Mazen Almasry
- Internal Medicine, Alfaisal University College of Medicine, Riyadh, SAU
| | - Maher Alawar
- Nephrology, Specialized Medical Center, Riyadh, SAU
| | | | | |
Collapse
|
11
|
Liu W, Huang G, Rui H, Geng J, Hu H, Huang Y, Huo G, Liu B, Xu A. Course monitoring of membranous nephropathy: Both autoantibodies and podocytes require multidimensional attention. Autoimmun Rev 2021; 21:102976. [PMID: 34757091 DOI: 10.1016/j.autrev.2021.102976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/18/2021] [Accepted: 10/24/2021] [Indexed: 01/15/2023]
Abstract
A variety of podocyte antigens have been identified in human membranous nephropathy (MN), which is divided into various antigen-dominated subtypes, confirming the concept that MN is the common pattern of glomerular injury in multiple autoimmune responses. The detection of autoantibodies has been widely used, which promoted the clinical practice of MN toward personalized precision medicine. However, given the potential risks of immunosuppressive therapy, more autoantibodies and biomarkers need to be identified to predict the prognosis and therapeutic response of MN more accurately. In this review, we attempted to summarize the autoantigens/autoantibodies and autoimmune mechanisms that can predict disease states based on the current understanding of MN pathogenesis, especially the podocyte injury manifestations. In conclusion, both the autoimmune response and podocyte injury require multidimensional attention in the disease course of MN.
Collapse
Affiliation(s)
- Wenbin Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hongliang Rui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jie Geng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Haikun Hu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yujiao Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guiyang Huo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
12
|
Jehn U, Bayraktar S, Pollmann S, Van Marck V, Weide T, Pavenstädt H, Brand E, Lenders M. α-Galactosidase a Deficiency in Fabry Disease Leads to Extensive Dysregulated Cellular Signaling Pathways in Human Podocytes. Int J Mol Sci 2021; 22:ijms222111339. [PMID: 34768768 PMCID: PMC8583658 DOI: 10.3390/ijms222111339] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
Fabry disease (FD) is caused by mutations in the α-galactosidase A (GLA) gene encoding the lysosomal AGAL enzyme. Loss of enzymatic AGAL activity and cellular accumulation of sphingolipids (mainly globotriaosylcermide) may lead to podocyturia and renal loss of function with increased cardiovascular morbidity and mortality in affected patients. To identify dysregulated cellular pathways in FD, we established a stable AGAL-deficient podocyte cell line to perform a comprehensive proteome analysis. Imbalanced protein expression and function were analyzed in additional FD cell lines including endothelial, epithelial kidney, patient-derived urinary cells and kidney biopsies. AGAL-deficient podocytes showed dysregulated proteins involved in thermogenesis, lysosomal trafficking and function, metabolic activity, cell-cell interactions and cell cycle. Proteins associated with neurological diseases were upregulated in AGAL-deficient podocytes. Rescues with inducible AGAL expression only partially normalized protein expression. A disturbed protein expression was confirmed in endothelial, epithelial and patient-specific cells, pointing toward fundamental pathway disturbances rather than to cell type-specific alterations in FD. We conclude that a loss of AGAL function results in profound changes of cellular pathways, which are ubiquitously in different cell types. Due to these profound alterations, current approved FD-specific therapies may not be sufficient to completely reverse all dysregulated pathways.
Collapse
Affiliation(s)
- Ulrich Jehn
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany; (U.J.); (S.B.); (T.W.); (H.P.)
| | - Samet Bayraktar
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany; (U.J.); (S.B.); (T.W.); (H.P.)
| | - Solvey Pollmann
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center (IFAZ), University Hospital Münster, 48149 Münster, Germany; (S.P.); (E.B.)
| | - Veerle Van Marck
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, 48149 Münster, Germany;
| | - Thomas Weide
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany; (U.J.); (S.B.); (T.W.); (H.P.)
| | - Hermann Pavenstädt
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology, and Rheumatology, University Hospital Münster, 48149 Münster, Germany; (U.J.); (S.B.); (T.W.); (H.P.)
| | - Eva Brand
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center (IFAZ), University Hospital Münster, 48149 Münster, Germany; (S.P.); (E.B.)
| | - Malte Lenders
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center (IFAZ), University Hospital Münster, 48149 Münster, Germany; (S.P.); (E.B.)
- Correspondence: ; Tel.: +49-251-8348-104
| |
Collapse
|
13
|
Li S, Luo Z, Meng S, Qiu X, Zheng F, Dai W, Zhang X, Sui W, Yan Q, Tang D, Dai Y. Label-free quantitative proteomic and phosphoproteomic analyses of renal biopsy tissues in membranous nephropathy. Proteomics Clin Appl 2021; 16:e2000069. [PMID: 34543527 DOI: 10.1002/prca.202000069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/01/2021] [Accepted: 09/17/2021] [Indexed: 11/11/2022]
Abstract
PURPOSE Membranous nephropathy (MN) is a common cause of nephrotic syndrome in adults. However, the underlying mechanisms of its occurrence and development are not completely clear. Thus, it is essential to explore the mechanisms. EXPERIMENTAL DESIGN Here, we employed label-free quantification and liquid chromatography-tandem mass spectrometry analysis techniques to investigate the proteomic and phosphoproteomic alterations in renal biopsy tissues of MN patients. Samples were collected from 16 MN patients and 10 controls. Immunohistochemistry (IHC) was performed to validate the hub phosphoprotein. RESULTS We focused on the changes in the phosphoproteome in MN group versus control group (CG). Totally, 1704 phosphoproteins containing 3241 phosphosites were identified and quantified. The phosphorylation levels of 216 phosphoproteins containing 297 phosphosites were differentially regulated in stage II MN group versus CG, and 333 phosphoproteins containing 461 phosphosites were differentially phosphorylated in stage III MN group versus CG. In each comparison, several differential phosphoproteins were factors, kinases and receptors involved in cellular processes, biological regulation and other biological processes. The subcellular location of most of the differential phosphoproteins was the nucleus. Protein-protein interaction analysis showed that the connections among the differential phosphoproteins were extremely complex, and several signalling pathways probably associated with MN were identified. The hub phosphoprotein was validated by IHC. CONCLUSIONS AND CLINICAL RELEVANCE This investigation can provide direct insight into the global phosphorylation events in MN group versus CG and may help to shed light on the potential pathogenic mechanisms of MN.
Collapse
Affiliation(s)
- Shanshan Li
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Zhifeng Luo
- Guangxi Key Laboratory of Metabolic Disease Research, Department of Nephrology, The No. 924 Hospital of the Joint Logistic Support Force of the Chinese People's Liberation Army, Guilin, China
| | - Shuhui Meng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaofen Qiu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Department of Nephrology, The No. 924 Hospital of the Joint Logistic Support Force of the Chinese People's Liberation Army, Guilin, China
| | - Fengping Zheng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, Texas, USA
| | - Xinzhou Zhang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Weiguo Sui
- Guangxi Key Laboratory of Metabolic Disease Research, Department of Nephrology, The No. 924 Hospital of the Joint Logistic Support Force of the Chinese People's Liberation Army, Guilin, China
| | - Qiang Yan
- Guangxi Key Laboratory of Metabolic Disease Research, Department of Nephrology, The No. 924 Hospital of the Joint Logistic Support Force of the Chinese People's Liberation Army, Guilin, China
| | - Donge Tang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
14
|
Martin CE, Jones N. ShcA expression in podocytes is dispensable for glomerular development but its upregulation is associated with kidney disease. Am J Transl Res 2021; 13:9874-9882. [PMID: 34540124 PMCID: PMC8430102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND ShcA (SHC1) is a phosphotyrosine adaptor protein which plays broad signaling roles within the cell. Systemic loss of ShcA during embryogenesis is lethal, while its aberrant expression contributes to disease. We recently demonstrated that ShcA is highly expressed during glomerular development and that it is upregulated within podocytes in experimental kidney injury and chronic kidney disease. The objective of this study was to analyze the in vivo role of ShcA in podocytes. METHODS We selectively deleted all three isoforms of ShcA from mouse kidney podocytes using the Cre/lox system driven by the podocyte-specific podocin promoter (Nphs2). Immunostaining of kidney sections was used to confirm ShcA deletion in podocytes. Coomassie blue staining of protein gels was used to detect urinary albumin. Light and electron microscopy were used to assess glomerular morphology. Transcript levels of SHC1 in human renal disease were assessed using the Nephroseq database. RESULTS Mice lacking podocyte ShcA were born at the expected Mendelian frequency and did not display overt renal impairment or changes in podocyte architecture beyond one year of age. In parallel, we correlated increased ShcA mRNA expression in the human kidney with proteinuria and reduced glomerular filtration rate. CONCLUSION Our studies reveal that ShcA is dispensable for normal kidney function, but its upregulation is associated with disease.
Collapse
Affiliation(s)
- Claire E Martin
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph Guelph, ON, Canada
| |
Collapse
|
15
|
Role of Rho GTPase Interacting Proteins in Subcellular Compartments of Podocytes. Int J Mol Sci 2021; 22:ijms22073656. [PMID: 33915776 PMCID: PMC8037304 DOI: 10.3390/ijms22073656] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
The first step of urine formation is the selective filtration of the plasma into the urinary space at the kidney structure called the glomerulus. The filtration barrier of the glomerulus allows blood cells and large proteins such as albumin to be retained while eliminating the waste products of the body. The filtration barrier consists of three layers: fenestrated endothelial cells, glomerular basement membrane, and podocytes. Podocytes are specialized epithelial cells featured by numerous, actin-based projections called foot processes. Proteins on the foot process membrane are connected to the well-organized intracellular actin network. The Rho family of small GTPases (Rho GTPases) act as intracellular molecular switches. They tightly regulate actin dynamics and subsequent diverse cellular functions such as adhesion, migration, and spreading. Previous studies using podocyte-specific transgenic or knockout animal models have established that Rho GTPases are crucial for the podocyte health and barrier function. However, little attention has been paid regarding subcellular locations where distinct Rho GTPases contribute to specific functions. In the current review, we discuss cellular events involving the prototypical Rho GTPases (RhoA, Rac1, and Cdc42) in podocytes, with particular focus on the subcellular compartments where the signaling events occur. We also provide our synthesized views of the current understanding and propose future research directions.
Collapse
|
16
|
Park T. Crk and CrkL as Therapeutic Targets for Cancer Treatment. Cells 2021; 10:cells10040739. [PMID: 33801580 PMCID: PMC8065463 DOI: 10.3390/cells10040739] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 02/03/2023] Open
Abstract
Crk and CrkL are cellular counterparts of the viral oncoprotein v-Crk. Crk and CrkL are overexpressed in many types of human cancer, correlating with poor prognosis. Furthermore, gene knockdown and knockout of Crk and CrkL in tumor cell lines suppress tumor cell functions, including cell proliferation, transformation, migration, invasion, epithelial-mesenchymal transition, resistance to chemotherapy drugs, and in vivo tumor growth and metastasis. Conversely, overexpression of tumor cells with Crk or CrkL enhances tumor cell functions. Therefore, Crk and CrkL have been proposed as therapeutic targets for cancer treatment. However, it is unclear whether Crk and CrkL make distinct or overlapping contributions to tumor cell functions in various cancer types because Crk or CrkL have been examined independently in most studies. Two recent studies using colorectal cancer and glioblastoma cells clearly demonstrated that Crk and CrkL need to be ablated individually and combined to understand distinct and overlapping roles of the two proteins in cancer. A comprehensive understanding of individual and overlapping roles of Crk and CrkL in tumor cell functions is necessary to develop effective therapeutic strategies. This review systematically discusses crucial functions of Crk and CrkL in tumor cell functions and provides new perspectives on targeting Crk and CrkL in cancer therapy.
Collapse
Affiliation(s)
- Taeju Park
- Children's Mercy Research Institute, Children's Mercy Kansas City, Department of Pediatrics, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
17
|
Feng D. Phosphorylation of key podocyte proteins and the association with proteinuric kidney disease. Am J Physiol Renal Physiol 2020; 319:F284-F291. [PMID: 32686524 DOI: 10.1152/ajprenal.00002.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Podocyte dysfunction contributes to proteinuric chronic kidney disease. A number of key proteins are essential for podocyte function, including nephrin, podocin, CD2-associated protein (CD2AP), synaptopodin, and α-actinin-4 (ACTN4). Although most of these proteins were first identified through genetic studies associated with human kidney disease, subsequent studies have identified phosphorylation of these proteins as an important posttranslational event that regulates their function. In this review, a brief overview of the function of these key podocyte proteins is provided. Second, the role of phosphorylation in regulating the function of these proteins is described. Third, the association between these phosphorylation pathways and kidney disease is reviewed. Finally, challenges and future directions in studying phosphorylation are discussed. Better characterization of these phosphorylation pathways and others yet to be discovered holds promise for translating this knowledge into new therapies for patients with proteinuric chronic kidney disease.
Collapse
Affiliation(s)
- Di Feng
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Blaine J, Dylewski J. Regulation of the Actin Cytoskeleton in Podocytes. Cells 2020; 9:cells9071700. [PMID: 32708597 PMCID: PMC7408282 DOI: 10.3390/cells9071700] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Podocytes are an integral part of the glomerular filtration barrier, a structure that prevents filtration of large proteins and macromolecules into the urine. Podocyte function is dependent on actin cytoskeleton regulation within the foot processes, structures that link podocytes to the glomerular basement membrane. Actin cytoskeleton dynamics in podocyte foot processes are complex and regulated by multiple proteins and other factors. There are two key signal integration and structural hubs within foot processes that regulate the actin cytoskeleton: the slit diaphragm and focal adhesions. Both modulate actin filament extension as well as foot process mobility. No matter what the initial cause, the final common pathway of podocyte damage is dysregulation of the actin cytoskeleton leading to foot process retraction and proteinuria. Disruption of the actin cytoskeleton can be due to acquired causes or to genetic mutations in key actin regulatory and signaling proteins. Here, we describe the major structural and signaling components that regulate the actin cytoskeleton in podocytes as well as acquired and genetic causes of actin dysregulation.
Collapse
Affiliation(s)
- Judith Blaine
- Renal Division, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - James Dylewski
- Renal Division, University of Colorado Anschutz Medical Campus and Denver Health Medical Center, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +303-724-4841
| |
Collapse
|
19
|
Crk1/2 and CrkL play critical roles in maintaining podocyte morphology and function. Exp Cell Res 2020; 394:112135. [PMID: 32535035 DOI: 10.1016/j.yexcr.2020.112135] [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: 01/06/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022]
Abstract
Podocytes are actin-rich epithelial cells whose effacement and detachment are the main cause of glomerular disease. Crk family proteins: Crk1/2 and CrkL are reported to be important intracellular signaling proteins that are involved in many biological processes. However, the roles of them in maintaining podocyte morphology and function remain poorly understood. In this study, specific knocking down of Crk1/2 and CrkL in podocytes caused abnormal cell morphology, actin cytoskeleton rearrangement and dysfunction in cell adhesion, spreading, migration, and viability. The p130Cas, focal adhesion kinase, phosphatidylinositol 3-kinase/Akt, p38 and JNK signaling pathways involved in these alterations. Furthermore, knocking down CrkL alone conferred a more modest phenotype than did the Crk1/2 knockdown and the double knockdown. Kidney biopsy specimens from patients with focal segmental glomerulosclerosis and minimal change nephropathy showed downregulation of Crk1/2 and CrkL in glomeruli. In zebrafish embryos, Crk1/2 and CrkL knockdown compromised the morphology and caused abnormal glomerular development. Thus, our results suggest that Crk1/2 and CrkL expression are important in podocytes; loss of either will cause podocyte dysfunction, leading to foot process effacement and podocyte detachment.
Collapse
|
20
|
Pisarek-Horowitz A, Fan X, Kumar S, Rasouly HM, Sharma R, Chen H, Coser K, Bluette CT, Hirenallur-Shanthappa D, Anderson SR, Yang H, Beck LH, Bonegio RG, Henderson JM, Berasi SP, Salant DJ, Lu W. Loss of Roundabout Guidance Receptor 2 (Robo2) in Podocytes Protects Adult Mice from Glomerular Injury by Maintaining Podocyte Foot Process Structure. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:799-816. [PMID: 32220420 PMCID: PMC7217334 DOI: 10.1016/j.ajpath.2019.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/24/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Roundabout guidance receptor 2 (ROBO2) plays an important role during early kidney development. ROBO2 is expressed in podocytes, inhibits nephrin-induced actin polymerization, down-regulates nonmuscle myosin IIA activity, and destabilizes kidney podocyte adhesion. However, the role of ROBO2 during kidney injury, particularly in mature podocytes, is not known. Herein, we report that loss of ROBO2 in podocytes [Robo2 conditional knockout (cKO) mouse] is protective from glomerular injuries. Ultrastructural analysis reveals that Robo2 cKO mice display less foot process effacement and better-preserved slit-diaphragm density compared with wild-type littermates injured by either protamine sulfate or nephrotoxic serum (NTS). The Robo2 cKO mice also develop less proteinuria after NTS injury. Further studies reveal that ROBO2 expression in podocytes is up-regulated after glomerular injury because its expression levels are higher in the glomeruli of NTS injured mice and passive Heymann membranous nephropathy rats. Moreover, the amount of ROBO2 in the glomeruli is also elevated in patients with membranous nephropathy. Finally, overexpression of ROBO2 in cultured mouse podocytes compromises cell adhesion. Taken together, these findings suggest that kidney injury increases glomerular ROBO2 expression that might compromise podocyte adhesion and, thus, loss of Robo2 in podocytes could protect from glomerular injury by enhancing podocyte adhesion that helps maintain foot process structure. Our findings also suggest that ROBO2 is a therapeutic target for podocyte injury and podocytopathy.
Collapse
Affiliation(s)
- Anna Pisarek-Horowitz
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Xueping Fan
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Sudhir Kumar
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Hila M Rasouly
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Richa Sharma
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Kathryn Coser
- Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts
| | | | | | - Sarah R Anderson
- Global Pathology, Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut
| | - Hongying Yang
- Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts
| | - Laurence H Beck
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Ramon G Bonegio
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Stephen P Berasi
- Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts
| | - David J Salant
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts.
| |
Collapse
|
21
|
Angiotensin II promotes podocyte injury by activating Arf6-Erk1/2-Nox4 signaling pathway. PLoS One 2020; 15:e0229747. [PMID: 32119711 PMCID: PMC7051060 DOI: 10.1371/journal.pone.0229747] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/13/2020] [Indexed: 11/20/2022] Open
Abstract
Angiotensin II (Ang II) is a key contributor to glomerular disease by predominantly resulting in podocyte injury, whereas the underlying molecular mechanisms has not been fully understood. This study aimed to investigate if and how ADP-ribosylation factor 6 (Arf6), a small GTP-binding protein, involves Ang II-induced cellular injury in cultured human podocytes. Cellular injury was evaluated with caspase 3 activity, reactive oxygen species (ROS) level and TUNEL assay. Arf6 activity was measured using an Arf6-GTP Pull-Down Assay. Ang II significantly enhanced Arf6 expressions accompanied by increase of Arf6-GTP. The TUNEL-positive cells as well as activated caspase 3, NADPH oxidase 4 protein (Nox4) and ROS levels were dramatically increased in Ang II-treated podocytes, which was prevented by secinH3, an Arf6 activity inhibitor. Induction of ROS by Ang II was inhibited in podocytes with Nox4 knockdown. Ang II-induced elevation of Nox4 and ROS was prevented by Arf6 knockdown. Phpspho-Erk1/2Thr202/Tyr204 levels were upregulated remarkably following Ang II treatment, and Erk inhibitor LY3214996 significantly downregulated Nox4 expression. In addition, Ang II decreased CD2AP expression. Overexpression of CD2AP prevented Ang II-induced upregulation of Arf6-GTP. Our data demonstrated that Ang II promotes ROS production and podocytes injury through activation of Arf6-Erk1/2-Nox4 signaling. We also provided evidence that Ang II activates Arf6 by degradation of CD2AP.
Collapse
|
22
|
Hall G, Wang L, Spurney RF. TRPC Channels in Proteinuric Kidney Diseases. Cells 2019; 9:cells9010044. [PMID: 31877991 PMCID: PMC7016871 DOI: 10.3390/cells9010044] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
Over a decade ago, mutations in the gene encoding TRPC6 (transient receptor potential cation channel, subfamily C, member 6) were linked to development of familial forms of nephrosis. Since this discovery, TRPC6 has been implicated in the pathophysiology of non-genetic forms of kidney disease including focal segmental glomerulosclerosis (FSGS), diabetic nephropathy, immune-mediated kidney diseases, and renal fibrosis. On the basis of these findings, TRPC6 has become an important target for the development of therapeutic agents to treat diverse kidney diseases. Although TRPC6 has been a major focus for drug discovery, more recent studies suggest that other TRPC family members play a role in the pathogenesis of glomerular disease processes and chronic kidney disease (CKD). This review highlights the data implicating TRPC6 and other TRPC family members in both genetic and non-genetic forms of kidney disease, focusing on TRPC3, TRPC5, and TRPC6 in a cell type (glomerular podocytes) that plays a key role in proteinuric kidney diseases.
Collapse
|
23
|
Chen Y, Liu Q, Shan Z, Mi W, Zhao Y, Li M, Wang B, Zheng X, Feng W. Catalpol Ameliorates Podocyte Injury by Stabilizing Cytoskeleton and Enhancing Autophagy in Diabetic Nephropathy. Front Pharmacol 2019; 10:1477. [PMID: 31920663 PMCID: PMC6914850 DOI: 10.3389/fphar.2019.01477] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/13/2019] [Indexed: 12/30/2022] Open
Abstract
Catalpol, an iridoid glycoside extracted from Rehmannia glutinosa, has been found to ameliorate diabetic nephropathy (DN), but the mechanism has not been clarified. Podocyte injury play a key role in the pathogenesis of DN. This study mainly investigated the protective effect and potential mechanism of catalpol on podocyte injury of DN in vivo and in vitro. The results indicated that the pathological features of DN in mice were markedly ameliorated after treatment with catalpol. Moreover, podocyte foot process effacement, and down-regulation of nephrin and synaptopodin expression in DN mice were also significantly improved after treatment with catalpol. In vitro, catalpol rescued disrupted cytoskeleton and increased migration ratio in podocytes induced by high glucose, the effect might be attributable to the inhibition of RhoA and Cdc42 activities but not Rac1. Furthermore, the impaired podocyte autophagy in DN mice was significantly enhanced after catalpol treatment. And catalpol also enhanced autophagy and lysosome biogenesis in cultured podocytes under high glucose condition. In addition, we found that catalpol could inhibit mTOR activity and promote TFEB nuclear translocation in vivo and in vitro experiments. Our study demonstrated that catalpol could ameliorate podocyte injury in DN, and the protective effect of catalpol might be attributed to the stabilization of podocyte cytoskeleton and the improvement of impaired podocyte autophagy.
Collapse
Affiliation(s)
- Yan Chen
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qingpu Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zengfu Shan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wangyang Mi
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yingying Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Meng Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Baiyan Wang
- College of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China.,Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China.,Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
24
|
Inoue K, Tian X, Velazquez H, Soda K, Wang Z, Pedigo CE, Wang Y, Cross E, Groener M, Shin JW, Li W, Hassan H, Yamamoto K, Mundel P, Ishibe S. Inhibition of Endocytosis of Clathrin-Mediated Angiotensin II Receptor Type 1 in Podocytes Augments Glomerular Injury. J Am Soc Nephrol 2019; 30:2307-2320. [PMID: 31511362 PMCID: PMC6900791 DOI: 10.1681/asn.2019010053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inhibition of the renin-angiotensin system remains a cornerstone in reducing proteinuria and progression of kidney failure, effects believed to be the result of reduction in BP and glomerular hyperfiltration. However, studies have yielded conflicting results on whether podocyte-specific angiotensin II (AngII) signaling directly induces podocyte injury. Previous research has found that after AngII stimulation, β-arrestin-bound angiotensin II receptor type 1 (AT1R) is internalized in a clathrin- and dynamin-dependent manner, and that Dynamin1 and Dynamin2 double-knockout mice exhibit impaired clathrin-mediated endocytosis. METHODS We used podocyte-specific Dyn double-knockout mice to examine AngII-stimulated AT1R internalization and signaling in primary podocytes and controls. We also examined the in vivo effect of AngII in these double-knockout mice through renin-angiotensin system blockers and through deletion of Agtr1a (which encodes the predominant AT1R isoform expressed in kidney, AT1aR). We tested calcium influx, Rac1 activation, and lamellipodial extension in control and primary podocytes of Dnm double-knockout mice treated with AngII. RESULTS We confirmed augmented AngII-stimulated AT1R signaling in primary Dnm double-knockout podocytes resulting from arrest of clathrin-coated pit turnover. Genetic ablation of podocyte Agtr1a in Dnm double-knockout mice demonstrated improved albuminuria and kidney function compared with the double-knockout mice. Isolation of podocytes from Dnm double-knockout mice revealed abnormal membrane dynamics, with increased Rac1 activation and lamellipodial extension, which was attenuated in Dnm double-knockout podocytes lacking AT1aR. CONCLUSIONS Our results indicate that inhibiting aberrant podocyte-associated AT1aR signaling pathways has a protective effect in maintaining the integrity of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Kazunori Inoue
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Heino Velazquez
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Keita Soda
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Zhen Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Christopher E Pedigo
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Ying Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Elizabeth Cross
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Marwin Groener
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jee-Won Shin
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Wei Li
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Hossam Hassan
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Koichi Yamamoto
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; and
| | - Peter Mundel
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut;
| |
Collapse
|
25
|
Sasaki H, Takahashi Y, Ogawa T, Hiura K, Nakano K, Sugiyama M, Okamura T, Sasaki N. Deletion of the Tensin2 SH2-PTB domain, but not the loss of its PTPase activity, induces podocyte injury in FVB/N mouse strain. Exp Anim 2019; 69:135-143. [PMID: 31723089 PMCID: PMC7220710 DOI: 10.1538/expanim.19-0101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Tensin2 (TNS2) is a focal adhesion-localized protein possessing N-terminal tandem protein
tyrosine phosphatase (PTPase) and C2 domains, and C-terminal tandem Src homology 2 (SH2)
and phosphotyrosine binding (PTB) domains. Genetic deletion of Tns2 in a
susceptible murine strain leads to podocyte alterations after birth. To clarify the domain
contributions to podocyte maintenance, we generated two Tns2-mutant mice
with the genetic background of the susceptible FVB/NJ strain,
Tns2∆C and Tns2CS mice, carrying
a SH2-PTB domain deletion and a PTPase domain inactivation, respectively. The
Tns2∆C mice developed massive albuminuria, severe
glomerular injury and podocyte alterations similarly to those in
Tns2-deficient mice. In contrast, the Tns2CS
mice showed no obvious phenotypic abnormalities. These results indicate that the TNS2
SH2-PTB domain, but not its PTPase activity, plays a role in podocyte maintenance.
Furthermore, in a podocyte cell line, the truncated TNS2 mutant lacking the SH2-PTB domain
lost the ability to localize to focal adhesion. Taken together, these data suggest that
TNS2 recruitment to focal adhesion is required to maintain postnatal podocytes on a
susceptible genetic background.
Collapse
Affiliation(s)
- Hayato Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| | - Yuki Takahashi
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| | - Tsubasa Ogawa
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| | - Koki Hiura
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| | - Kenta Nakano
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan.,Department of Laboratory Animal Medicine, Section of Animal Models, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Section of Animal Models, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, 35-1 Higashi-23, Towada, Aomori 034-8628, Japan
| |
Collapse
|
26
|
Dlugos CP, Picciotto C, Lepa C, Krakow M, Stöber A, Eddy ML, Weide T, Jeibmann A, P Krahn M, Van Marck V, Klingauf J, Ricker A, Wedlich-Söldner R, Pavenstädt H, Klämbt C, George B. Nephrin Signaling Results in Integrin β1 Activation. J Am Soc Nephrol 2019; 30:1006-1019. [PMID: 31097607 DOI: 10.1681/asn.2018040362] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with certain mutations in the gene encoding the slit diaphragm protein Nephrin fail to develop functional slit diaphragms and display severe proteinuria. Many adult-onset glomerulopathies also feature alterations in Nephrin expression and function. Nephrin signals from the podocyte slit diaphragm to the Actin cytoskeleton by recruiting proteins that can interact with C3G, a guanine nucleotide exchange factor of the small GTPase Rap1. Because Rap activity affects formation of focal adhesions, we hypothesized that Nephrin transmits signals to the Integrin receptor complex, which mediates podocyte adhesion to the extracellular matrix. METHODS To investigate Nephrin's role in transmitting signals to the Integrin receptor complex, we conducted genetic studies in Drosophila nephrocytes and validated findings from Drosophila in a cultured human podocyte model. RESULTS Drosophila nephrocytes form a slit diaphragm-like filtration barrier and express the Nephrin ortholog Sticks and stones (Sns). A genetic screen identified c3g as necessary for nephrocyte function. In vivo, nephrocyte-specific gene silencing of sns or c3g compromised nephrocyte filtration and caused nephrocyte diaphragm defects. Nephrocytes with impaired Sns or C3G expression displayed an altered localization of Integrin and the Integrin-associated protein Talin. Furthermore, gene silencing of c3g partly rescued nephrocyte diaphragm defects of an sns overexpression phenotype, pointing to genetic interaction of sns and c3g in nephrocytes. We also found that activated Nephrin recruited phosphorylated C3G and resulted in activation of Integrin β1 in cultured podocytes. CONCLUSIONS Our findings suggest that Nephrin can mediate a signaling pathway that results in activation of Integrin β1 at focal adhesions, which may affect podocyte attachment to the extracellular matrix.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christian Klämbt
- Neurobiology, Westfälische-Wilhelms University Münster, Münster, Germany
| | | |
Collapse
|
27
|
A therapeutic approach towards microRNA29 family in vascular diabetic complications: A boon or curse? J Diabetes Metab Disord 2019; 18:243-254. [PMID: 31275895 DOI: 10.1007/s40200-019-00409-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/02/2019] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus (DM) is one of the major metabolic disorders and its severity leads to death. Enhancement in hyperglycaemic conditions of DM gives rise to endothelial impairment in small and large blood vessels contributing towards microvascular and macrovascular complications respectively. The pathogenesis of diabetic complications is associated with interruption of various signal transduction pathways due to epigenetic modifications such as aberrant histone modifications, DNA methylation and expression of miRNAs along with the long non-coding RNAs (lncRNAs). Amongst these epigenetic alterations, modulated expressions of miRNAs confer to apoptosis and endothelial dysfunction of organs that gives rise to vascular complications. In this review, we principally focussed on physiological role of miR29 family in DM and have discussed crosstalk between miR29 family and numerous genes involved in signal transduction pathways of Diabetic vascular complications. Incidences of diabetic retinopathy exploiting the role of miR29 in regulation of EMT process, differential expression patterns of miR29 and promising therapeutic role of miR29 have been discussed. We have summarised the therapeutic role of miR29 in podocyte impairment and how miR29 regulates the expressions of profibrotic, inflammatory and ECM encoding genes in renal fibrosis under diabetic conditions. We have also highlighted impact of miR29 expression patterns in cardiac angiopathy, cardiomyocyte's apoptosis and cardiac fibrosis. Additionally, we have also presented the contradictory actions of miR29 family in amelioration as well as in enhancement of diabetic complications.
Collapse
|
28
|
Hall G, Spurney RF. Losing their footing: Rac1 signaling causes podocyte detachment and FSGS. Kidney Int 2019; 92:283-285. [PMID: 28709595 DOI: 10.1016/j.kint.2017.03.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 12/26/2022]
Abstract
Selective modulation of Rho GTPase activity in podocytes recapitulates characteristic features of human nephrosis. Using a mouse model, Robins et al. found that high levels of Rac1 activation in podocytes caused podocyte detachment and glomerulosclerosis. Podocyte Rac1 activity was enhanced in biopsy specimens from patients with nephrosis, and serum from this patient population activated Rac1 in cultured podocytes. These data provide a causal link between podocyte Rac1 activation and human nephrotic diseases.
Collapse
Affiliation(s)
- Gentzon Hall
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | - Robert F Spurney
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA.
| |
Collapse
|
29
|
Arif E, Solanki AK, Srivastava P, Rahman B, Tash BR, Holzman LB, Janech MG, Martin R, Knölker HJ, Fitzgibbon WR, Deng P, Budisavljevic MN, Syn WK, Wang C, Lipschutz JH, Kwon SH, Nihalani D. The motor protein Myo1c regulates transforming growth factor-β-signaling and fibrosis in podocytes. Kidney Int 2019; 96:139-158. [PMID: 31097328 DOI: 10.1016/j.kint.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 01/19/2023]
Abstract
Transforming growth factor-β (TGF-β) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-β signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-β-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-β-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-β signaling through downregulation of canonical and non-canonical TGF-β pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-β signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-β responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-β-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.
Collapse
Affiliation(s)
- Ehtesham Arif
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ashish K Solanki
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Pankaj Srivastava
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bushra Rahman
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian R Tash
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lawrence B Holzman
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael G Janech
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA; College of Charleston, Charleston, South Carolina, USA
| | - René Martin
- Department of Chemistry, TU Dresden, Dresden, Germany
| | | | - Wayne R Fitzgibbon
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peifeng Deng
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Milos N Budisavljevic
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wing-Kin Syn
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA; Section of Gastroenterology, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, (UPV/EHU), Vizcaya, Spain
| | - Cindy Wang
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joshua H Lipschutz
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Deepak Nihalani
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
30
|
Yu SMW, Nissaisorakarn P, Husain I, Jim B. Proteinuric Kidney Diseases: A Podocyte's Slit Diaphragm and Cytoskeleton Approach. Front Med (Lausanne) 2018; 5:221. [PMID: 30255020 PMCID: PMC6141722 DOI: 10.3389/fmed.2018.00221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/18/2018] [Indexed: 01/19/2023] Open
Abstract
Proteinuric kidney diseases are a group of disorders with diverse pathological mechanisms associated with significant losses of protein in the urine. The glomerular filtration barrier (GFB), comprised of the three important layers, the fenestrated glomerular endothelium, the glomerular basement membrane (GBM), and the podocyte, dictates that disruption of any one of these structures should lead to proteinuric disease. Podocytes, in particular, have long been considered as the final gatekeeper of the GFB. This specialized visceral epithelial cell contains a complex framework of cytoskeletons forming foot processes and mediate important cell signaling to maintain podocyte health. In this review, we will focus on slit diaphragm proteins such as nephrin, podocin, TRPC6/5, as well as cytoskeletal proteins Rho/small GTPases and synaptopodin and their respective roles in participating in the pathogenesis of proteinuric kidney diseases. Furthermore, we will summarize the potential therapeutic options targeting the podocyte to treat this group of kidney diseases.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States
| | | | - Irma Husain
- Department of Medicine, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Belinda Jim
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States.,Renal Division, Jacobi Medical Center, Bronx, NY, United States
| |
Collapse
|
31
|
Verma R, Venkatareddy M, Kalinowski A, Li T, Kukla J, Mollin A, Cara-Fuentes G, Patel SR, Garg P. Nephrin is necessary for podocyte recovery following injury in an adult mature glomerulus. PLoS One 2018; 13:e0198013. [PMID: 29924795 PMCID: PMC6010211 DOI: 10.1371/journal.pone.0198013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/12/2018] [Indexed: 11/28/2022] Open
Abstract
Nephrin (Nphs1) is an adhesion protein that is expressed at the podocyte intercellular junction in the glomerulus. Nphs1 mutations in humans or deletion in animal genetic models results in a developmental failure of foot process formation. A number of studies have shown decrease in expression of nephrin in various proteinuric kidney diseases as well as in animal models of glomerular disease. Decrease in nephrin expression has been suggested to precede podocyte loss and linked to the progression of kidney disease. Whether the decrease in expression of nephrin is related to loss of podocytes or lead to podocyte detachment is unclear. To answer this central question we generated an inducible model of nephrin deletion (Nphs1Tam-Cre) in order to lower nephrin expression in healthy adult mice. Following tamoxifen-induction there was a 75% decrease in nephrin expression by 14 days. The Nphs1Tam-Cre mice had normal foot process ultrastructure and intact filtration barriers up to 4-6 weeks post-induction. Despite the loss of nephrin expression, the podocyte number and density remained unchanged during the initial period. Unexpectedly, nephrin expression, albeit at low levels persisted at the slit diaphragm up to 16-20 weeks post-tamoxifen induction. The mice became progressively proteinuric with glomerular hypertrophy and scarring reminiscent of focal and segmental glomerulosclerosis at 20 weeks. Four week-old Nphs1 knockout mice subjected to protamine sulfate model of podocyte injury demonstrated failure to recover from foot process effacement following heparin sulfate. Similarly, Nphs1 knockout mice failed to recover following nephrotoxic serum (NTS) with persistence of proteinuria and foot process effacement. Our results suggest that as in development, nephrin is necessary for maintenance of a healthy glomerular filter. In contrast to the developmental phenotype, lowering nephrin expression in a mature glomerulus resulted in a slowly progressive disease that histologically resembles FSGS a disease linked closely with podocyte depletion. Podocytes with low levels of nephrin expression are both susceptible and unable to recover following perturbation. Our results suggest that decreased nephrin expression independent of podocyte loss occurring as an early event in proteinuric kidney diseases might play a role in disease progression.
Collapse
Affiliation(s)
- Rakesh Verma
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Madhusudan Venkatareddy
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Anne Kalinowski
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Theodore Li
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Joanna Kukla
- Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | | | - Gabriel Cara-Fuentes
- Division of Pediatric Nephrology, Motts Children Hospital, Ann Arbor, Michigan, United States of America
| | - Sanjeevkumar R. Patel
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- Veterans Administration, VAMC, Ann Arbor, Michigan, United States of America
| | - Puneet Garg
- Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- Veterans Administration, VAMC, Ann Arbor, Michigan, United States of America
| |
Collapse
|
32
|
Pan Y, Jiang S, Hou Q, Qiu D, Shi J, Wang L, Chen Z, Zhang M, Duan A, Qin W, Zen K, Liu Z. Dissection of Glomerular Transcriptional Profile in Patients With Diabetic Nephropathy: SRGAP2a Protects Podocyte Structure and Function. Diabetes 2018; 67:717-730. [PMID: 29242313 DOI: 10.2337/db17-0755] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/26/2017] [Indexed: 01/19/2023]
Abstract
Podocytes play a pivotal role in maintaining glomerular filtration function through their interdigitated foot processes. However, the mechanisms that govern the podocyte cytoskeletal rearrangement remain unclear. Through analyzing the transcriptional profile of renal biopsy specimens from patients with diabetic nephropathy (DN) and control donors, we identify SLIT-ROBO ρGTPase-activating protein 2a (SRGAP2a) as one of the main hub genes strongly associated with proteinuria and glomerular filtration in type 2 DN. Immunofluorescence staining and Western blot analysis revealed that human and mouse SRGAP2a is primarily localized at podocytes and largely colocalized with synaptopodin. Moreover, podocyte SRGAP2a is downregulated in patients with DN and db/db mice at both the mRNA and the protein level. SRGAP2a reduction is observed in cultured podocytes treated with tumor growth factor-β or high concentrations of glucose. Functional and mechanistic studies show that SRGAP2a suppresses podocyte motility through inactivating RhoA/Cdc42 but not Rac1. The protective role of SRGAP2a in podocyte function also is confirmed in zebrafish, in which knockdown of SRGAP2a, a SRGAP2 ortholog in zebrafish, recapitulates podocyte foot process effacement. Finally, increasing podocyte SRGAP2a levels in db/db mice through administration of adenovirus-expressing SRGAP2a significantly mitigates podocyte injury and proteinuria. The results demonstrate that SRGAP2a protects podocytes by suppressing podocyte migration.
Collapse
Affiliation(s)
- Yu Pan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Song Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Qing Hou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Dandan Qiu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Ling Wang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Zhaohong Chen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Aiping Duan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Fukusumi Y, Zhang Y, Yamagishi R, Oda K, Watanabe T, Matsui K, Kawachi H. Nephrin-Binding Ephrin-B1 at the Slit Diaphragm Controls Podocyte Function through the JNK Pathway. J Am Soc Nephrol 2018; 29:1462-1474. [PMID: 29602834 DOI: 10.1681/asn.2017090993] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 11/03/2022] Open
Abstract
Background B-type ephrins are membrane-bound proteins that maintain tissue function in several organs. We previously reported that ephrin-B1 is localized at the slit diaphragm of glomerular podocytes. However, the function of ephrin-B1 at this location is unclear.Methods We analyzed the phenotype of podocyte-specific ephrin-B1 knockout mice and assessed the molecular association of ephrin-B1 and nephrin, a key molecule of the slit diaphragm, in HEK293 cells and rats with anti-nephrin antibody-induced nephropathy.Results Compared with controls, ephrin-B1 conditional knockout mice displayed altered podocyte morphology, disarrangement of the slit diaphragm molecules, and proteinuria. Ephrin-B1 expressed in HEK293 cells immunoprecipitated with nephrin, which required the basal regions of the extracellular domains of both proteins. Treatment of cells with an anti-nephrin antibody promoted the phosphorylation of nephrin and ephrin-B1. However, phosphorylation of ephrin-B1 did not occur in cells expressing a mutant nephrin lacking the ephrin-B1 binding site or in cells treated with an Src kinase inhibitor. The phosphorylation of ephrin-B1 enhanced the phosphorylation of nephrin and promoted the phosphorylation of c-Jun N-terminal kinase (JNK), which was required for ephrin-B1-promoted cell motility in wound-healing assays. Notably, phosphorylated JNK was detected in the glomeruli of control mice but not ephrin-B1 conditional knockout mice. In rats, the phosphorylation of ephrin-B1, JNK, and nephrin occurred in the early phase (24 hours) of anti-nephrin antibody-induced nephropathy.Conclusions Through interactions with nephrin, ephrin-B1 maintains the structure and barrier function of the slit diaphragm. Moreover, phosphorylation of ephrin-B1 and, consequently, JNK are involved in the development of podocyte injury.
Collapse
Affiliation(s)
- Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ying Zhang
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryohei Yamagishi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kanako Oda
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata, Japan
| | - Toru Watanabe
- Department of Pediatrics, Niigata City General Hospital, Niigata, Japan; and
| | - Katsuyuki Matsui
- Department of Internal Medicine IV, Teikyo University School of Medicine, Kawasaki, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan;
| |
Collapse
|
34
|
Gromnitza S, Lepa C, Weide T, Schwab A, Pavenstädt H, George B. Tropomyosin-related kinase C (TrkC) enhances podocyte migration by ERK-mediated WAVE2 activation. FASEB J 2018; 32:1665-1676. [PMID: 29162704 DOI: 10.1096/fj.201700703r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Podocyte malfunction is central to glomerular diseases and is marked by defective podocyte intercellular junctions and actin cytoskeletal dynamics. Podocytes share many morphologic features with neurons, so that similar sets of proteins appear to regulate cell process formation. One such protein is the tropomyosin-related kinase C (TrkC). TrkC deficiency in mice leads to proteinuria as a surrogate of defective kidney filter function. Activation of endogenous TrkC by its ligand neurotrophin-3 resulted in increased podocyte migration-a surrogate of podocyte actin dynamics in vivo. Employing a mutagenesis approach, we found that the Src homologous and collagen-like (Shc) binding site Tyr516 within the TrkC cytoplasmic domain was necessary for TrkC-induced migration of podocytes. TrkC activation led to a mobility shift of Wiskott-Aldrich syndrome family verprolin-homologous protein (WAVE)-2 which is known to orchestrate Arp2/3 activation and actin polymerization. Chemical inactivation of Erk or mutagenesis of 2 of 4 known Erk target sites within WAVE2, Thr346 and Ser351, abolished the TrkC-induced WAVE2 mobility shift. Knockdown of WAVE2 by shRNA abolished TrkC-induced podocyte migration. In summary, TrkC signals to the podocyte actin cytoskeleton to induce migration by phosphorylating WAVE2 Erk dependently. This signaling mechanism may be important for TrkC-mediated cytoskeletal dynamics in podocyte disease.-Gromnitza, S., Lepa, C., Weide, T., Schwab, A., Pavenstädt, H., George, B. Tropomyosin-related kinase C (TrkC) enhances podocyte migration by ERK-mediated WAVE2 activation.
Collapse
Affiliation(s)
- Sascha Gromnitza
- Medizinische Klinik D, Universitätsklinikum Münster, Muenster, Germany
| | - Carolin Lepa
- Medizinische Klinik D, Universitätsklinikum Münster, Muenster, Germany
| | - Thomas Weide
- Medizinische Klinik D, Universitätsklinikum Münster, Muenster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Westfälische-Wilhelms-Universität Münster, Muenster, Germany
| | | | - Britta George
- Medizinische Klinik D, Universitätsklinikum Münster, Muenster, Germany
| |
Collapse
|
35
|
Martin CE, Jones N. Nephrin Signaling in the Podocyte: An Updated View of Signal Regulation at the Slit Diaphragm and Beyond. Front Endocrinol (Lausanne) 2018; 9:302. [PMID: 29922234 PMCID: PMC5996060 DOI: 10.3389/fendo.2018.00302] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Podocytes are a major component of the glomerular blood filtration barrier, and alterations to the morphology of their unique actin-based foot processes (FP) are a common feature of kidney disease. Adjacent FP are connected by a specialized intercellular junction known as the slit diaphragm (SD), which serves as the ultimate barrier to regulate passage of macromolecules from the blood. While the link between SD dysfunction and reduced filtration selectivity has been recognized for nearly 50 years, our understanding of the underlying molecular circuitry began only 20 years ago, sparked by the identification of NPHS1, encoding the transmembrane protein nephrin. Nephrin not only functions as the core component of the extracellular SD filtration network but also as a signaling scaffold via interactions at its short intracellular region. Phospho-regulation of several conserved tyrosine residues in this region influences signal transduction pathways which control podocyte cell adhesion, shape, and survival, and emerging studies highlight roles for nephrin phospho-dynamics in mechanotransduction and endocytosis. The following review aims to summarize the last 5 years of advancement in our knowledge of how signaling centered at nephrin directs SD barrier formation and function. We further provide insight on promising frontiers in podocyte biology, which have implications for SD signaling in the healthy and diseased kidney.
Collapse
|
36
|
Gupta KH, Goldufsky JW, Wood SJ, Tardi NJ, Moorthy GS, Gilbert DZ, Zayas JP, Hahm E, Altintas MM, Reiser J, Shafikhani SH. Apoptosis and Compensatory Proliferation Signaling Are Coupled by CrkI-Containing Microvesicles. Dev Cell 2017. [PMID: 28633020 DOI: 10.1016/j.devcel.2017.05.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Apoptosis has been implicated in compensatory proliferation signaling (CPS), whereby dying cells induce proliferation in neighboring cells as a means to restore homeostasis. The nature of signaling between apoptotic cells and their neighboring cells remains largely unknown. Here we show that a fraction of apoptotic cells produce and release CrkI-containing microvesicles (distinct from exosomes and apoptotic bodies), which induce proliferation in neighboring cells upon contact. We provide visual evidence of CPS by videomicroscopy. We show that purified vesicles in vitro and in vivo are sufficient to stimulate proliferation in other cells. Our data demonstrate that CrkI inactivation by ExoT bacterial toxin or by mutagenesis blocks vesicle formation in apoptotic cells and inhibits CPS, thus uncoupling apoptosis from CPS. We further show that c-Jun amino-terminal kinase (JNK) plays a pivotal role in mediating vesicle-induced CPS in recipient cells. CPS could have important ramifications in diseases that involve apoptotic cell death.
Collapse
Affiliation(s)
- Kajal H Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Josef W Goldufsky
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Stephen J Wood
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Nicholas J Tardi
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gayathri S Moorthy
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Douglas Z Gilbert
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Janet P Zayas
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Eunsil Hahm
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H Shafikhani
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA; Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA; Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
37
|
Targeting Neph1 and ZO-1 protein-protein interaction in podocytes prevents podocyte injury and preserves glomerular filtration function. Sci Rep 2017; 7:12047. [PMID: 28935902 PMCID: PMC5608913 DOI: 10.1038/s41598-017-12134-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/05/2017] [Indexed: 11/08/2022] Open
Abstract
Targeting protein-protein interaction (PPI) is rapidly becoming an attractive alternative for drug development. While drug development commonly involves inhibiting a PPI, in this study, we show that stabilizing PPI may also be therapeutically beneficial. Junctional proteins Neph1 and ZO-1 and their interaction is an important determinant of the structural integrity of slit diaphragm, which is a critical component of kidney's filtration system. Since injury induces loss of this interaction, we hypothesized that strengthening this interaction may protect kidney's filtration barrier and preserve kidney function. In this study, Neph1-ZO-1 structural complex was screened for the presence of small druggable pockets formed from contributions from both proteins. One such pocket was identified and screened using a small molecule library. Isodesmosine (ISD) a rare naturally occurring amino acid and a biomarker for pulmonary arterial hypertension was selected as the best candidate and to establish the proof of concept, its ability to enhance Neph1-CD and ZO-1 binding was tested. Results from biochemical binding analysis showed that ISD enhanced Neph1 and ZO-1 interaction under in vitro and in vivo conditions. Importantly, ISD treated podocytes were resistant to injury-induced loss of transepithelial permeability. Finally, mouse and zebrafish studies show that ISD protects from injury-induced renal damage.
Collapse
|
38
|
ARF6 mediates nephrin tyrosine phosphorylation-induced podocyte cellular dynamics. PLoS One 2017; 12:e0184575. [PMID: 28880939 PMCID: PMC5589247 DOI: 10.1371/journal.pone.0184575] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/26/2017] [Indexed: 01/10/2023] Open
Abstract
ADP-ribosylation factor 6 (ARF6) is a small GTPase necessary for regulating cellular structure, motility, and vesicle trafficking. In several cellular systems, ARF6 was shown to regulate actin dynamics in coordination with Rac1, a Rho small GTPase. We examined the function of ARF6 in the kidney podocyte because Rac1 was implicated in kidney diseases involving this cell. We found that ARF6 expression was enriched in human podocytes and that it modulated podocyte cytoskeletal dynamics through a functional interaction with nephrin, an intercellular junction protein necessary for podocyte injury-induced signaling requiring activation by tyrosine phosphorylation of its cytoplasmic domain. ARF6 was necessary for nephrin activation-induced ruffling and focal adhesion turnover, possibly by altering Rac1 activity. In podocyte-specific Arf6 (ARF6_PodKO) knockout mice, ARF6 deficiency did not result in a spontaneous kidney developmental phenotype or proteinuria after aging. However, ARF6_PodKO mice exhibited distinct phenotypes in two in vivo glomerular injury models. In the protamine sulfate perfusion model, which induced acute podocyte effacement, ARF6_PodKO mice were protected from podocyte effacement. In the nephrotoxic serum nephritis model, which induced immune-complex mediated injury, ARF6_PodKO mice exhibited aggravated proteinuria. Together, these observations suggest that while ARF6 is necessary for nephrin tyrosine phosphorylation-induced cytoskeletal dynamics in cultured podocytes, ARF6 has pleotropic podocyte roles in vivo, where glomerular injury-specific mechanisms might activate distinct signaling pathways that dictate whether ARF6 activity is beneficial or deleterious for maintaining the integrity of the glomerular filtration barrier.
Collapse
|
39
|
Wan J, Hou X, Zhou Z, Geng J, Tian J, Bai X, Nie J. WT1 ameliorates podocyte injury via repression of EZH2/β-catenin pathway in diabetic nephropathy. Free Radic Biol Med 2017; 108:280-299. [PMID: 28315733 DOI: 10.1016/j.freeradbiomed.2017.03.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/26/2017] [Accepted: 03/13/2017] [Indexed: 10/20/2022]
Abstract
Epigenetic modulation of podocyte injury plays a pivotal role in diabetic nephropathy (DN). Wilm's tumor 1 (WT1) has been found to have opposing roles with β-catenin in podocyte biology. Herein, we asked whether the histone methyltransferase enzyme enhancer of zeste homolog 2 (EZH2) promotes WT1-induced podocyte injury via β-catenin activation and the underlying mechanisms. We found that WT1 antagonized EZH2 and ameliorated β-catenin-mediated podocyte injury as demonstrated by attenuated podocyte mesenchymal transition, maintenance of podocyte architectural integrity, decreased podocyte apoptosis and oxidative stress. Further, we provided mechanistical evidence that EZH2 was required in WT1-mediated β-catenin inactivation via repression of secreted frizzled-related protein 1 (SFRP-1), a Wnt antagonist. Moreover, EZH2-mediated silencing of SFRP-1 was due to increased histone 3 lysine 27 trimethylation (H3K27me3) on its promoter region. WT1 favored renal function and decreased podocyte injury in diabetic rats and DN patients. Notably, WT1 exhibited clinical and biological relevance as it was linked to dropped serum creatinine, decreased proteinuria and elevated estimated glomerular filtration rate (eGFR). We propose an epigenetic process via the WT1/EZH2/β-catenin axis in attenuating podocyte injury in DN. Targeting WT1 and EZH2 could be potential therapeutic approaches for DN.
Collapse
Affiliation(s)
- Jiao Wan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China
| | - Xiaoyan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China
| | - Zhanmei Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China
| | - Jian Geng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jianwei Tian
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China
| | - Xiaoyan Bai
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China.
| | - Jing Nie
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, Guangdong, PR China.
| |
Collapse
|
40
|
Abstract
In this article, I shall outline some of the most important aspects of the evidentiary basis of the so-called Kriz model for the development of glomerular sclerosis, a model that we continue to modify to this day. In my mind, the most important findings include the fact that podocytes are generally post-mitotic cells, so that loss of a significant number for any cause leads to podocyte insufficiency. Another pivotal finding is that in many experimental models and in human disease, podocytes detach from the GBM as living cells. These facts, together with biomechanical deduction, have led to the ongoing evolution of the original Heidelberg model.
Collapse
|
41
|
Adrenomedullin ameliorates podocyte injury induced by puromycin aminonucleoside in vitro and in vivo through modulation of Rho GTPases. Int Urol Nephrol 2017; 49:1489-1506. [DOI: 10.1007/s11255-017-1622-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 05/15/2017] [Indexed: 01/02/2023]
|
42
|
Swiatecka-Urban A. Endocytic Trafficking at the Mature Podocyte Slit Diaphragm. Front Pediatr 2017; 5:32. [PMID: 28286744 PMCID: PMC5324021 DOI: 10.3389/fped.2017.00032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/03/2017] [Indexed: 12/16/2022] Open
Abstract
Endocytic trafficking couples cell signaling with the cytoskeletal dynamics by organizing a crosstalk between protein networks in different subcellular compartments. Proteins residing in the plasma membrane are internalized and transported as cargo in endocytic vesicles (i.e., endocytosis). Subsequently, cargo proteins can be delivered to lysosomes for degradation or recycled back to the plasma membrane. The slit diaphragm is a modified tight junction connecting foot processes of the glomerular epithelial cells, podocytes. Signaling at the slit diaphragm plays a critical role in the kidney while its dysfunction leads to glomerular protein loss (proteinuria), manifesting as nephrotic syndrome, a rare condition with an estimated incidence of 2-4 new cases per 100,000 each year. Relatively little is known about the role of endocytic trafficking in podocyte signaling and maintenance of the slit diaphragm integrity. This review will focus on the role of endocytic trafficking at the mature podocyte slit diaphragm.
Collapse
Affiliation(s)
- Agnieszka Swiatecka-Urban
- Department of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Haas ME, Levenson AE, Sun X, Liao WH, Rutkowski JM, de Ferranti SD, Schumacher VA, Scherer PE, Salant DJ, Biddinger SB. The Role of Proprotein Convertase Subtilisin/Kexin Type 9 in Nephrotic Syndrome-Associated Hypercholesterolemia. Circulation 2016; 134:61-72. [PMID: 27358438 DOI: 10.1161/circulationaha.115.020912] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/28/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND In nephrotic syndrome, damage to the podocytes of the kidney produces severe hypercholesterolemia for which novel treatments are urgently needed. PCSK9 (proprotein convertase subtilisin/kexin type 9) has emerged as an important regulator of plasma cholesterol levels and therapeutic target. Here, we tested the role of PCSK9 in mediating the hypercholesterolemia of nephrotic syndrome. METHODS PCSK9 and plasma lipids were studied in nephrotic syndrome patients before and after remission of disease, mice with genetic ablation of the podocyte (Podocyte Apoptosis Through Targeted Activation of Caspase-8, Pod-ATTAC mice) and mice treated with nephrotoxic serum (NTS), which triggers immune-mediated podocyte damage. In addition, mice with hepatic deletion of Pcsk9 were treated with NTS to determine the contribution of PCSK9 to the dyslipidemia of nephrotic syndrome. RESULTS Patients with nephrotic syndrome showed a decrease in plasma cholesterol and plasma PCSK9 on remission of their disease (P<0.05, n=47-50). Conversely, Pod-ATTAC mice and NTS-treated mice showed hypercholesterolemia and a 7- to 24-fold induction in plasma PCSK9. The induction of plasma PCSK9 appeared to be attributable to increased secretion of PCSK9 from the hepatocyte coupled with decreased clearance. Interestingly, knockout of Pcsk9ameliorated the effects of NTS on plasma lipids. Thus, in the presence of NTS, mice lacking hepatic Pcsk9 showed a 40% to 50% decrease in plasma cholesterol and triglycerides. Moreover, the ability of NTS treatment to increase the percentage of low-density lipoprotein-associated cholesterol (from 9% in vehicle-treated Flox mice to 47% after NTS treatment), was lost in mice with hepatic deletion of Pcsk9 (5% in both the presence and absence of NTS). CONCLUSIONS Podocyte damage triggers marked inductions in plasma PCSK9, and knockout of Pcsk9 ameliorates dyslipidemia in a mouse model of nephrotic syndrome. These data suggest that PCSK9 inhibitors may be beneficial in patients with nephrotic syndrome-associated hypercholesterolemia.
Collapse
Affiliation(s)
- Mary E Haas
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Amy E Levenson
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Xiaowei Sun
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Wan-Hui Liao
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Joseph M Rutkowski
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Sarah D de Ferranti
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Valerie A Schumacher
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Philipp E Scherer
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - David J Salant
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.)
| | - Sudha B Biddinger
- From Division of Endocrinology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (M.E.H., A.E.L., X.S., W.-H.L., S.B.B.); Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (J.M.R., P.E.S.); Division of Cardiology, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, MA (S.D.d.F.); Division of Nephrology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA (V.A.S.); and Department of Medicine, Section of Nephrology, Boston University Medical Center, MA (D.J.S.).
| |
Collapse
|
44
|
Pathogenesis of proteinuria in idiopathic minimal change disease: molecular mechanisms. Pediatr Nephrol 2016; 31:2179-2189. [PMID: 27384691 DOI: 10.1007/s00467-016-3379-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/07/2016] [Accepted: 03/14/2016] [Indexed: 12/13/2022]
Abstract
Minimal change disease (MCD) is the most common type of nephrotic syndrome in children and adolescents. The pathogenesis of proteinuria in this condition is currently being reassessed. Following the Shalhoub hypothesis, most efforts have been placed on identifying the putative circulating factor, but recent advancement in podocyte biology has focused attention on the molecular changes at the glomerular capillary wall, which could explain the mechanism of proteinuria in MCD. This report critically reviews current knowledge on the different postulated mechanisms at the glomerular capillary wall level for increased permeability to plasma proteins in MCD. The report helps describe the rationale behind novel therapies and suggests future targeted therapies for MCD.
Collapse
|
45
|
Abstract
Genetic studies of hereditary forms of nephrotic syndrome have identified several proteins that are involved in regulating the permselective properties of the glomerular filtration system. Further extensive research has elucidated the complex molecular basis of the glomerular filtration barrier and clearly established the pivotal role of podocytes in the pathophysiology of glomerular diseases. Podocyte architecture is centred on focal adhesions and slit diaphragms - multiprotein signalling hubs that regulate cell morphology and function. A highly interconnected actin cytoskeleton enables podocytes to adapt in order to accommodate environmental changes and maintain an intact glomerular filtration barrier. Actin-based endocytosis has now emerged as a regulator of podocyte integrity, providing an impetus for understanding the precise mechanisms that underlie the steady-state control of focal adhesion and slit diaphragm components. This Review outlines the role of actin dynamics and endocytosis in podocyte biology, and discusses how molecular heterogeneity in glomerular disorders could be exploited to deliver more rational therapeutic interventions, paving the way for targeted medicine in nephrology.
Collapse
|
46
|
Subramanian B, Sun H, Yan P, Charoonratana VT, Higgs HN, Wang F, Lai KMV, Valenzuela DM, Brown EJ, Schlöndorff JS, Pollak MR. Mice with mutant Inf2 show impaired podocyte and slit diaphragm integrity in response to protamine-induced kidney injury. Kidney Int 2016; 90:363-372. [PMID: 27350175 DOI: 10.1016/j.kint.2016.04.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/18/2016] [Accepted: 04/28/2016] [Indexed: 01/08/2023]
Abstract
Mutations in the INF2 (inverted formin 2) gene, encoding a diaphanous formin family protein that regulates actin cytoskeleton dynamics, cause human focal segmental glomerulosclerosis (FSGS). INF2 interacts directly with certain other mammalian diaphanous formin proteins (mDia) that function as RhoA effector molecules. FSGS-causing INF2 mutations impair these interactions and disrupt the ability of INF2 to regulate Rho/Dia-mediated actin dynamics in vitro. However, the precise mechanisms by which INF2 regulates and INF2 mutations impair glomerular structure and function remain unknown. Here, we characterize an Inf2 R218Q point-mutant (knockin) mouse to help answer these questions. Knockin mice have no significant renal pathology or proteinuria at baseline despite diminished INF2 protein levels. INF2 mutant podocytes do show impaired reversal of protamine sulfate-induced foot process effacement by heparin sulfate perfusion. This is associated with persistent podocyte cytoplasmic aggregation, nephrin phosphorylation, and nephrin and podocin mislocalization, as well as impaired recovery of mDia membrane localization. These changes were partially mimicked in podocyte outgrowth cultures, in which podocytes from knockin mice show altered cellular protrusions compared to those from wild-type mice. Thus, in mice, normal INF2 function is not required for glomerular development but normal INF2 is required for regulation of the actin-based behaviors necessary for response to and/or recovery from injury.
Collapse
Affiliation(s)
- Balajikarthick Subramanian
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Hua Sun
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; University of Children's Hospital, Iowa City, Iowa, USA
| | - Paul Yan
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Victoria T Charoonratana
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Henry N Higgs
- Department of Biochemistry, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Fang Wang
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ka-Man V Lai
- Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | | | - Elizabeth J Brown
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Department of Pediatrics, Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Johannes S Schlöndorff
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
| |
Collapse
|
47
|
Sun H, Tian J, Xian W, Xie T, Yang X. Pentraxin-3 Attenuates Renal Damage in Diabetic Nephropathy by Promoting M2 Macrophage Differentiation. Inflammation 2016; 38:1739-47. [PMID: 25761429 DOI: 10.1007/s10753-015-0151-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As one of the most important long-term complications of diabetes, diabetic nephropathy (DN) is the major cause of end-stage renal disease and high mortality in diabetic patients. The long pentraxin 3 (Ptx3) is a member of a superfamily of conserved proteins characterized by a cyclic multimeric structure and a conserved C-terminal domain. Several clinical investigations have demonstrated that elevated plasma Ptx3 levels are associated with cardiovascular and chronic kidney diseases (CKD). However, the therapeutic effect of Ptx3 on DN has never been investigated. In our current study, we showed a crucial role for Ptx3 in attenuating renal damage in DN. In our mouse hyperglycemia-induced nephropathy model, Ptx3 treatment showed significantly increased expression of nephrin, acetylated nephrin, and Wilm's tumor-1 protein (WT-1) when compared with control. The number of CD4(+) T cells, CD8(+) T cells, Ly6G(+) neutrophils, and CD11b(+) macrophages were all significantly lower in the Ptx3-treated group than that in the control group in DN. The IL-4 and IL-13 levels in the Ptx3-treated group were markedly higher than that in the control group in DN. Correspondingly, the Ptx3-treated group showed increased numbers of Arg1- or CD206-expressing macrophages compared with the control group. Furthermore, inhibition of Ptx3-treated macrophages abrogated the alleviated renal damage induced by Ptx3 treatment. In conclusion, Ptx3 attenuates renal damage in DN by promoting M2 macrophage differentiation.
Collapse
Affiliation(s)
- Huaibin Sun
- Department of Hemodialysis, Qilu Hospital, Shandong University, Jinan, China
| | | | | | | | | |
Collapse
|
48
|
Pullen N, Fornoni A. Drug discovery in focal and segmental glomerulosclerosis. Kidney Int 2016; 89:1211-20. [PMID: 27165834 PMCID: PMC4875964 DOI: 10.1016/j.kint.2015.12.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 11/26/2022]
Abstract
Despite the high medical burden experienced by patients with focal segmental glomerulosclerosis, the etiology of the condition remains largely unknown. Focal segmental glomerulosclerosis is highly heterogeneous in clinical and morphologic manifestations. While this presents challenges for the development of new treatments, research investments over the last 2 decades have yielded a surfeit of potential avenues for therapeutic intervention. The development of many of those ideas and concepts into new therapies, however, has been very disappointing. Here, we describe some of the factors that have potentially contributed to the poor translational performance from this research investment, including the confidence we ascribe to a target, the conduct of experimental studies, and the availability of selective reagents to test hypotheses. We will discuss the significance of genetic and systems traits as well as other methods for reducing bias. We will analyze the limitations of a successful drug development. We will use specific examples hoping that these will guide a consensus for investment and drive greater translational quality. We hope that this substrate will serve to exemplify the tremendous opportunity for intervention as well as facilitate greater collaborative effort between industry, academia, and private foundations in promoting appropriate validation of these targets. Only then will we have achieved our goal for curative therapies for this devastating disease.
Collapse
Affiliation(s)
- Nick Pullen
- Pfizer Global Research & Development, Cambridge, Massachusetts, USA.
| | - Alessia Fornoni
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
49
|
Abstract
Chronic kidney disease (CKD) represents a leading cause of death in the United States. There is no cure for this disease, with current treatment strategies relying on blood pressure control through blockade of the renin-angiotensin system. Such approaches only delay the development of end-stage kidney disease and can be associated with serious side effects. Recent identification of several novel mechanisms contributing to CKD development - including vascular changes, loss of podocytes and renal epithelial cells, matrix deposition, inflammation and metabolic dysregulation - has revealed new potential therapeutic approaches for CKD. This Review assesses emerging strategies and agents for CKD treatment, highlighting the associated challenges in their clinical development.
Collapse
|
50
|
Tian X, Ishibe S. Targeting the podocyte cytoskeleton: from pathogenesis to therapy in proteinuric kidney disease. Nephrol Dial Transplant 2016; 31:1577-83. [PMID: 26968197 DOI: 10.1093/ndt/gfw021] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023] Open
Abstract
Glomerular injury often incites a progression to chronic kidney disease, which affects millions of patients worldwide. Despite our current understanding of this disease's pathogenesis, there is still a lack of therapy available to curtail its progression. However, exciting new data strongly suggest the podocyte-an actin-rich, terminally differentiated epithelial cell that lines the outside of the glomerular filtration barrier-as a therapeutic target. The importance of podocytes in the pathogenesis of human nephrotic syndrome is best characterized by identification of genetic mutations, many of which regulate the actin cytoskeleton. The intricate regulation of the podocyte actin cytoskeleton is fundamental in preserving an intact glomerular filtration barrier, and this knowledge has inspired new research targeting actin-regulating proteins in these cells. This review will shed light on recent findings, which have furthered our understanding of the molecular mechanisms regulating podocyte actin dynamics, as well as discoveries that have therapeutic implications in the treatment of proteinuric kidney disease.
Collapse
Affiliation(s)
- Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|