51
|
Hamatani H, Sakairi T, Ikeuchi H, Kaneko Y, Maeshima A, Nojima Y, Hiromura K. TGF‐β1 alters DNA methylation levels in promoter and enhancer regions of the
WT1
gene in human podocytes. Nephrology (Carlton) 2019; 24:575-584. [DOI: 10.1111/nep.13411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Hiroko Hamatani
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Toru Sakairi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoriaki Kaneko
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Akito Maeshima
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoshihisa Nojima
- Department of Rheumatology and NephrologyJapan Red Cross Maebashi Hospital Maebashi Japan
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| |
Collapse
|
52
|
Dorval G, Kuzmuk V, Gribouval O, Welsh GI, Bierzynska A, Schmitt A, Miserey-Lenkei S, Koziell A, Haq S, Benmerah A, Mollet G, Boyer O, Saleem MA, Antignac C. TBC1D8B Loss-of-Function Mutations Lead to X-Linked Nephrotic Syndrome via Defective Trafficking Pathways. Am J Hum Genet 2019; 104:348-355. [PMID: 30661770 DOI: 10.1016/j.ajhg.2018.12.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is characterized by high-range proteinuria and most often focal and segmental glomerulosclerosis (FSGS). Identification of mutations in genes causing SRNS has improved our understanding of disease mechanisms and highlighted defects in the podocyte, a highly specialized glomerular epithelial cell, as major factors in disease pathogenesis. By exome sequencing, we identified missense mutations in TBC1D8B in two families with an X-linked early-onset SRNS with FSGS. TBC1D8B is an uncharacterized Rab-GTPase-activating protein likely involved in endocytic and recycling pathways. Immunofluorescence studies revealed TBC1D8B presence in human glomeruli, and affected individual podocytes displayed architectural changes associated with migration defects commonly found in FSGS. In zebrafish we demonstrated that both knockdown and knockout of the unique TBC1D8B ortholog-induced proteinuria and that this phenotype was rescued by human TBC1D8B mRNA injection, but not by either of the two mutated mRNAs. We also showed an interaction between TBC1D8B and Rab11b, a key protein in vesicular recycling in cells. Interestingly, both internalization and recycling processes were dramatically decreased in affected individuals' podocytes and fibroblasts, confirming the crucial role of TBC1D8B in the cellular recycling processes, probably as a Rab11b GTPase-activating protein. Altogether, these results confirmed that pathogenic variations in TBC1D8B are involved in X-linked podocytopathy and points to alterations in recycling processes as a mechanism of SRNS.
Collapse
Affiliation(s)
- Guillaume Dorval
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France
| | - Valeryia Kuzmuk
- Bristol Renal, University of Bristol and Bristol Royal Hospital for Children, Bristol, UK
| | - Olivier Gribouval
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France
| | - Gavin I Welsh
- Bristol Renal, University of Bristol and Bristol Royal Hospital for Children, Bristol, UK
| | - Agnieszka Bierzynska
- Bristol Renal, University of Bristol and Bristol Royal Hospital for Children, Bristol, UK
| | - Alain Schmitt
- Inserm, U1016, Institut Cochin, 75014 Paris, France; Cnrs, UMR8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Stéphanie Miserey-Lenkei
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, 75005 Paris, France
| | - Ania Koziell
- Department of Children's Nephrology and Urology, Evelina London, London SE1 7EH, UK
| | - Shuman Haq
- Paediatric Nephrology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France
| | - Géraldine Mollet
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France
| | - Olivia Boyer
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France; Department of Pediatric Nephrology, Reference center for Hereditary Kidney Diseases (MARHEA), Necker Hospital, APHP, 75015 Paris, France
| | - Moin A Saleem
- Bristol Renal, University of Bristol and Bristol Royal Hospital for Children, Bristol, UK.
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases, Imagine Institute, INSERM U1163, Paris Descartes University, 75015 Paris, France; Department of Genetics, Reference center for Hereditary Kidney Diseases (MARHEA), Necker Hospital, APHP,75015 Paris, France.
| |
Collapse
|
53
|
Okabe M, Motojima M, Miyazaki Y, Pastan I, Yokoo T, Matsusaka T. Global polysome analysis of normal and injured podocytes. Am J Physiol Renal Physiol 2018; 316:F241-F252. [PMID: 30379099 DOI: 10.1152/ajprenal.00115.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Podocyte injury is a key event for progressive renal failure. We have previously established a mouse model of inducible podocyte injury (NEP25) that progressively develops glomerulosclerosis after immunotoxin injection. We performed polysome analysis of intact and injured podocytes utilizing the NEP25 and RiboTag transgenic mice, in which a hemagglutinin tag is attached to ribosomal protein L22 selectively in podocytes. Podocyte-specific polysomes were successfully obtained by immunoprecipitation with an antihemagglutinin antibody from glomerular homogenate and analyzed using a microarray. Compared with glomerular cells, 353 genes were highly expressed and enriched in podocytes; these included important podocyte genes and also heretofore uncharacterized genes, such as Dach1 and Foxd2. Podocyte injury by immunotoxin induced many genes to be upregulated, including inflammation-related genes despite no infiltration of inflammatory cells in the glomeruli. MafF and Egr-1, which structurally have the potential to antagonize MafB and WT1, respectively, were rapidly and markedly increased in injured podocytes before MafB and WT1 were decreased. We demonstrated that Maff and Egr1 knockdown increased the MafB targets Nphs2 and Ptpro and the WT1 targets Ptpro, Nxph3, and Sulf1, respectively. This indicates that upregulated MafF and Egr-1 may promote deterioration of podocytes by antagonizing MafB and WT1. Our systematic microarray study of the heretofore undescribed behavior of podocyte genes may open new insights into the understanding of podocyte pathophysiology.
Collapse
Affiliation(s)
- Masahiro Okabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine , Tokyo , Japan.,Department of Basic Medicine, Tokai University School of Medicine , Isehara , Japan
| | - Masaru Motojima
- Department of Clinical Pharmacology, Tokai University School of Medicine , Isehara , Japan
| | - Yoichi Miyazaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine , Tokyo , Japan
| | - Taiji Matsusaka
- Department of Basic Medicine, Tokai University School of Medicine , Isehara , Japan.,Institute of Medical Science, Tokai University School of Medicine , Isehara , Japan
| |
Collapse
|
54
|
Hudson NO, Buck-Koehntop BA. Zinc Finger Readers of Methylated DNA. Molecules 2018; 23:E2555. [PMID: 30301273 PMCID: PMC6222495 DOI: 10.3390/molecules23102555] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 01/07/2023] Open
Abstract
DNA methylation is a prevalent epigenetic modification involved in regulating a number of essential cellular processes, including genomic accessibility and transcriptional outcomes. As such, aberrant alterations in global DNA methylation patterns have been associated with a growing number of disease conditions. Nevertheless, the full mechanisms by which DNA methylation information is interpreted and translated into genomic responses is not yet fully understood. Methyl-CpG binding proteins (MBPs) function as important mediators of this essential process by selectively reading DNA methylation signals and translating this information into down-stream cellular outcomes. The Cys₂His₂ zinc finger scaffold is one of the most abundant DNA binding motifs found within human transcription factors, yet only a few zinc finger containing proteins capable of conferring selectivity for mCpG over CpG sites have been characterized. This review summarizes our current structural understanding for the mechanisms by which the zinc finger MBPs evaluated to date read this essential epigenetic mark. Further, some of the biological implications for mCpG readout elicited by this family of MBPs are discussed.
Collapse
Affiliation(s)
- Nicholas O Hudson
- Department of Chemistry, University of Utah, Salt Lake City, UT 84112-0850, USA.
| | | |
Collapse
|
55
|
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: 5.3] [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
|
56
|
Guo Y, Pace J, Li Z, Ma'ayan A, Wang Z, Revelo MP, Chen E, Gu X, Attalah A, Yang Y, Estrada C, Yang VW, He JC, Mallipattu SK. Podocyte-Specific Induction of Krüppel-Like Factor 15 Restores Differentiation Markers and Attenuates Kidney Injury in Proteinuric Kidney Disease. J Am Soc Nephrol 2018; 29:2529-2545. [PMID: 30143559 PMCID: PMC6171275 DOI: 10.1681/asn.2018030324] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/02/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Podocyte injury is the hallmark of proteinuric kidney diseases, such as FSGS and minimal change disease, and destabilization of the podocyte's actin cytoskeleton contributes to podocyte dysfunction in many of these conditions. Although agents, such as glucocorticoids and cyclosporin, stabilize the actin cytoskeleton, systemic toxicity hinders chronic use. We previously showed that loss of the kidney-enriched zinc finger transcription factor Krüppel-like factor 15 (KLF15) increases susceptibility to proteinuric kidney disease and attenuates the salutary effects of retinoic acid and glucocorticoids in the podocyte. METHODS We induced podocyte-specific KLF15 in two proteinuric murine models, HIV-1 transgenic (Tg26) mice and adriamycin (ADR)-induced nephropathy, and used RNA sequencing of isolated glomeruli and subsequent enrichment analysis to investigate pathways mediated by podocyte-specific KLF15 in Tg26 mice. We also explored in cultured human podocytes the potential mediating role of Wilms Tumor 1 (WT1), a transcription factor critical for podocyte differentiation. RESULTS In Tg26 mice, inducing podocyte-specific KLF15 attenuated podocyte injury, glomerulosclerosis, tubulointerstitial fibrosis, and inflammation, while improving renal function and overall survival; it also attenuated podocyte injury in ADR-treated mice. Enrichment analysis of RNA sequencing from the Tg26 mouse model shows that KLF15 induction activates pathways involved in stabilization of actin cytoskeleton, focal adhesion, and podocyte differentiation. Transcription factor enrichment analysis, with further experimental validation, suggests that KLF15 activity is in part mediated by WT1. CONCLUSIONS Inducing podocyte-specific KLF15 attenuates kidney injury by directly and indirectly upregulating genes critical for podocyte differentiation, suggesting that KLF15 induction might be a potential strategy for treating proteinuric kidney disease.
Collapse
Affiliation(s)
| | | | - Zhengzhe Li
- Division of Nephrology, Department of Medicine and
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zichen Wang
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Edward Chen
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | | - Vincent W Yang
- Gastroenterology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C He
- Division of Nephrology, Department of Medicine and.,Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York.,Renal Section, James J. Peters Veterans Affairs Medical Center, New York, New York; and
| | - Sandeep K Mallipattu
- Divisions of Nephrology and .,Renal Section, Northport Veterans Affairs Medical Center, Northport, New York
| |
Collapse
|
57
|
Karaiskos N, Rahmatollahi M, Boltengagen A, Liu H, Hoehne M, Rinschen M, Schermer B, Benzing T, Rajewsky N, Kocks C, Kann M, Müller RU. A Single-Cell Transcriptome Atlas of the Mouse Glomerulus. J Am Soc Nephrol 2018; 29:2060-2068. [PMID: 29794128 DOI: 10.1681/asn.2018030238] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/28/2018] [Indexed: 11/03/2022] Open
Abstract
Background Three different cell types constitute the glomerular filter: mesangial cells, endothelial cells, and podocytes. However, to what extent cellular heterogeneity exists within healthy glomerular cell populations remains unknown.Methods We used nanodroplet-based highly parallel transcriptional profiling to characterize the cellular content of purified wild-type mouse glomeruli.Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identified the three known glomerular cell types. We provide a comprehensive online atlas of gene expression in glomerular cells that can be queried and visualized using an interactive and freely available database. Novel marker genes for all glomerular cell types were identified and supported by immunohistochemistry images obtained from the Human Protein Atlas. Subclustering of endothelial cells revealed a subset of endothelium that expressed marker genes related to endothelial proliferation. By comparison, the podocyte population appeared more homogeneous but contained three smaller, previously unknown subpopulations.Conclusions Our study comprehensively characterized gene expression in individual glomerular cells and sets the stage for the dissection of glomerular function at the single-cell level in health and disease.
Collapse
Affiliation(s)
- Nikos Karaiskos
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Anastasiya Boltengagen
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Haiyue Liu
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Martin Hoehne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne
| | - Markus Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne.,Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, San Diego, California
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany; and
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany; and
| | - Nikolaus Rajewsky
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christine Kocks
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany;
| | - Martin Kann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany; and
| |
Collapse
|
58
|
Asfahani RI, Tahoun MM, Miller-Hodges EV, Bellerby J, Virasami AK, Sampson RD, Moulding D, Sebire NJ, Hohenstein P, Scambler PJ, Waters AM. Activation of podocyte Notch mediates early Wt1 glomerulopathy. Kidney Int 2018; 93:903-920. [PMID: 29398135 PMCID: PMC6169130 DOI: 10.1016/j.kint.2017.11.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 01/26/2023]
Abstract
The Wilms' tumor suppressor gene, WT1, encodes a zinc finger protein that regulates podocyte development and is highly expressed in mature podocytes. Mutations in the WT1 gene are associated with the development of renal failure due to the formation of scar tissue within glomeruli, the mechanisms of which are poorly understood. Here, we used a tamoxifen-based CRE-LoxP system to induce deletion of Wt1 in adult mice to investigate the mechanisms underlying evolution of glomerulosclerosis. Podocyte apoptosis was evident as early as the fourth day post-induction and increased during disease progression, supporting a role for Wt1 in mature podocyte survival. Podocyte Notch activation was evident at disease onset with upregulation of Notch1 and its transcriptional targets, including Nrarp. There was repression of podocyte FoxC2 and upregulation of Hey2 supporting a role for a Wt1/FoxC2/Notch transcriptional network in mature podocyte injury. The expression of cleaved Notch1 and HES1 proteins in podocytes of mutant mice was confirmed in early disease. Furthermore, induction of podocyte HES1 expression was associated with upregulation of genes implicated in epithelial mesenchymal transition, thereby suggesting that HES1 mediates podocyte EMT. Lastly, early pharmacological inhibition of Notch signaling ameliorated glomerular scarring and albuminuria. Thus, loss of Wt1 in mature podocytes modulates podocyte Notch activation, which could mediate early events in WT1-related glomerulosclerosis.
Collapse
Affiliation(s)
- Rowan I Asfahani
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Mona M Tahoun
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK; Clinical and Chemical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eve V Miller-Hodges
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Jack Bellerby
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Alex K Virasami
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Robert D Sampson
- Institute of Ophthalmology, University College of London, London, UK
| | - Dale Moulding
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Neil J Sebire
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Peter J Scambler
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Aoife M Waters
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
59
|
Ullmark T, Montano G, Gullberg U. DNA and RNA binding by the Wilms' tumour gene 1 (WT1) protein +KTS and −KTS isoforms-From initial observations to recent global genomic analyses. Eur J Haematol 2018; 100:229-240. [DOI: 10.1111/ejh.13010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Tove Ullmark
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| | - Giorgia Montano
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| | - Urban Gullberg
- Department of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| |
Collapse
|
60
|
Abstract
Ultimately, the common final pathway of any glomerular disease is podocyte effacement, podocyte loss, and, eventually, glomerular scarring. There has been a long-standing debate on the underlying mechanisms for podocyte depletion, ranging from necrosis and apoptosis to detachment of viable cells from the glomerular basement membrane. However, this debate still continues because additional pathways of programmed cell death have been reported in recent years. Interestingly, viable podocytes can be isolated out of the urine of proteinuric patients easily, emphasizing the importance of podocyte detachment in glomerular diseases. In contrast, detection of apoptosis and other pathways of programmed cell death in podocytes is technically challenging. In fact, we still are lacking direct evidence showing, for example, the presence of apoptotic bodies in podocytes, leaving the question unanswered as to whether podocytes undergo mechanisms of programmed cell death. However, understanding the mechanisms leading to podocyte depletion is of particular interest because future therapeutic strategies might interfere with these to prevent glomerular scarring. In this review, we summarize our current knowledge on podocyte cell death, the different molecular pathways and experimental approaches to study these, and, finally, focus on the mechanisms that prevent the onset of programmed cell death.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
61
|
Lal MA, Patrakka J. Understanding Podocyte Biology to Develop Novel Kidney Therapeutics. Front Endocrinol (Lausanne) 2018; 9:409. [PMID: 30083135 PMCID: PMC6065143 DOI: 10.3389/fendo.2018.00409] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/02/2018] [Indexed: 11/13/2022] Open
Abstract
Over the past two decades it has become increasing clear that injury and loss of podocytes is an early and common clinical observation presented in many forms of glomerulopathy and chronic kidney disease. Identification of disease-causing monogenic mutations in numerous podocyte-expressed genes as well as studies conducted using preclinical animal models have shown that the podocyte plays a central role in establishing kidney dysfunction. In this review, we summarize current knowledge regarding the potential for podocyte-targeted therapies and give our view on how a deeper understanding of the molecular makeup of the podocyte will enable future therapeutic interventions. Specifically, we recount some of the currently described podocentric strategies for therapy and summarize the status and evolution of various model systems used to facilitate our understanding of the molecular and functional underpinnings of podocyte biology.
Collapse
Affiliation(s)
- Mark A. Lal
- Bioscience, Cardiovascular, Renal and Metabolism, Innovative Medicines Biotech Unit, AstraZeneca, Gothenburg, Sweden
- *Correspondence: Mark A. Lal
| | - Jaakko Patrakka
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Center, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Jaakko Patrakka
| |
Collapse
|
62
|
O’Brien LL, Guo Q, Bahrami-Samani E, Park JS, Hasso SM, Lee YJ, Fang A, Kim AD, Guo J, Hong TM, Peterson KA, Lozanoff S, Raviram R, Ren B, Fogelgren B, Smith AD, Valouev A, McMahon AP. Transcriptional regulatory control of mammalian nephron progenitors revealed by multi-factor cistromic analysis and genetic studies. PLoS Genet 2018; 14:e1007181. [PMID: 29377931 PMCID: PMC5805373 DOI: 10.1371/journal.pgen.1007181] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/08/2018] [Accepted: 01/01/2018] [Indexed: 12/12/2022] Open
Abstract
Nephron progenitor number determines nephron endowment; a reduced nephron count is linked to the onset of kidney disease. Several transcriptional regulators including Six2, Wt1, Osr1, Sall1, Eya1, Pax2, and Hox11 paralogues are required for specification and/or maintenance of nephron progenitors. However, little is known about the regulatory intersection of these players. Here, we have mapped nephron progenitor-specific transcriptional networks of Six2, Hoxd11, Osr1, and Wt1. We identified 373 multi-factor associated 'regulatory hotspots' around genes closely associated with progenitor programs. To examine their functional significance, we deleted 'hotspot' enhancer elements for Six2 and Wnt4. Removal of the distal enhancer for Six2 leads to a ~40% reduction in Six2 expression. When combined with a Six2 null allele, progeny display a premature depletion of nephron progenitors. Loss of the Wnt4 enhancer led to a significant reduction of Wnt4 expression in renal vesicles and a mildly hypoplastic kidney, a phenotype also enhanced in combination with a Wnt4 null mutation. To explore the regulatory landscape that supports proper target gene expression, we performed CTCF ChIP-seq to identify insulator-boundary regions. One such putative boundary lies between the Six2 and Six3 loci. Evidence for the functional significance of this boundary was obtained by deep sequencing of the radiation-induced Brachyrrhine (Br) mutant allele. We identified an inversion of the Six2/Six3 locus around the CTCF-bound boundary, removing Six2 from its distal enhancer regulation, but placed next to Six3 enhancer elements which support ectopic Six2 expression in the lens where Six3 is normally expressed. Six3 is now predicted to fall under control of the Six2 distal enhancer. Consistent with this view, we observed ectopic Six3 in nephron progenitors. 4C-seq supports the model for Six2 distal enhancer interactions in wild-type and Br/+ mouse kidneys. Together, these data expand our view of the regulatory genome and regulatory landscape underpinning mammalian nephrogenesis.
Collapse
Affiliation(s)
- Lori L. O’Brien
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Department of Preventative Medicine, Division of Bioinformatics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Emad Bahrami-Samani
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| | - Joo-Seop Park
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Sean M. Hasso
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Young-Jin Lee
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Alan Fang
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Albert D. Kim
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Trudy M. Hong
- Department of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | | | - Scott Lozanoff
- Department of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Ramya Raviram
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, Moores Cancer Center, University of California San Diego La Jolla, California, United States of America
| | - Bing Ren
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, Moores Cancer Center, University of California San Diego La Jolla, California, United States of America
| | - Ben Fogelgren
- Department of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Andrew D. Smith
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| | - Anton Valouev
- Department of Preventative Medicine, Division of Bioinformatics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
63
|
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: 14.2] [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
|
64
|
Transcriptomes of major renal collecting duct cell types in mouse identified by single-cell RNA-seq. Proc Natl Acad Sci U S A 2017; 114:E9989-E9998. [PMID: 29089413 DOI: 10.1073/pnas.1710964114] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prior RNA sequencing (RNA-seq) studies have identified complete transcriptomes for most renal epithelial cell types. The exceptions are the cell types that make up the renal collecting duct, namely intercalated cells (ICs) and principal cells (PCs), which account for only a small fraction of the kidney mass, but play critical physiological roles in the regulation of blood pressure, extracellular fluid volume, and extracellular fluid composition. To enrich these cell types, we used FACS that employed well-established lectin cell surface markers for PCs and type B ICs, as well as a newly identified cell surface marker for type A ICs, c-Kit. Single-cell RNA-seq using the IC- and PC-enriched populations as input enabled identification of complete transcriptomes of A-ICs, B-ICs, and PCs. The data were used to create a freely accessible online gene-expression database for collecting duct cells. This database allowed identification of genes that are selectively expressed in each cell type, including cell-surface receptors, transcription factors, transporters, and secreted proteins. The analysis also identified a small fraction of hybrid cells expressing aquaporin-2 and anion exchanger 1 or pendrin transcripts. In many cases, mRNAs for receptors and their ligands were identified in different cells (e.g., Notch2 chiefly in PCs vs. Jag1 chiefly in ICs), suggesting signaling cross-talk among the three cell types. The identified patterns of gene expression among the three types of collecting duct cells provide a foundation for understanding physiological regulation and pathophysiology in the renal collecting duct.
Collapse
|
65
|
Nathan A, Reinhardt P, Kruspe D, Jörß T, Groth M, Nolte H, Habenicht A, Herrmann J, Holschbach V, Toth B, Krüger M, Wang ZQ, Platzer M, Englert C. The Wilms tumor protein Wt1 contributes to female fertility by regulating oviductal proteostasis. Hum Mol Genet 2017; 26:1694-1705. [PMID: 28334862 PMCID: PMC5411738 DOI: 10.1093/hmg/ddx075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/23/2017] [Indexed: 11/16/2022] Open
Abstract
Although the zinc finger transcription factor Wt1 has been linked to female fertility, its precise role in this process has not yet been understood. We have sequenced the WT1 exons in a panel of patients with idiopathic infertility and have identified a missense mutation in WT1 in one patient out of eight. This mutation leads to an amino acid change within the zinc finger domain and results in reduced DNA binding. We utilized Wt1+/- mice as a model to mechanistically pinpoint the consequences of reduced Wt1 levels for female fertility. Our results indicate that subfertility in Wt1+/- female mice is a maternal effect caused by the Wt1-dependent de-regulation of Prss29, encoding a serine protease. Notably, blocking Prss29 activity was sufficient to rescue subfertility in Wt1+/- mice indicating Prss29 as a critical factor in female fertility. Molecularly, Wt1 represses expression of Prss29. De-repression and precocious expression of Prss29 in the oviduct of Wt1+/- mice interferes with pre-implantation development. Our study reveals a novel role for Wt1 in early mammalian development and identifies proteases as critical mediators of the maternal-embryonic interaction. Our data also suggest that the role of Wt1 in regulating fertility is conserved in mammals.
Collapse
Affiliation(s)
| | | | | | | | - Marco Groth
- Genome Analysis Lab, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hendrik Nolte
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andreas Habenicht
- Institute for Vascular Medicine, Jena University Hospital, Jena, Germany
| | - Jörg Herrmann
- Department of Gynecology and Obstetrics, Hufeland Klinikum, 99425 Weimar, Germany
| | - Verena Holschbach
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Bettina Toth
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marcus Krüger
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | - Matthias Platzer
- Genome Analysis Lab, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Christoph Englert
- Molecular Genetics Lab.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| |
Collapse
|
66
|
Abstract
The study of genes mutated in human disease often leads to new insights into biology as well as disease mechanisms. One such gene is Wilms' tumour 1 (WT1), which plays multiple roles in development, tissue homeostasis and disease. In this Primer, I summarise how this multifaceted gene functions in various mammalian tissues and organs, including the kidney, gonads, heart and nervous system. This is followed by a discussion of our current understanding of the molecular mechanisms by which WT1 and its two major isoforms regulate these processes at the transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- Nicholas D Hastie
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road (S), Edinburgh, EH4 2XU, UK
| |
Collapse
|
67
|
Kim YK, Refaeli I, Brooks CR, Jing P, Gulieva RE, Hughes MR, Cruz NM, Liu Y, Churchill AJ, Wang Y, Fu H, Pippin JW, Lin LY, Shankland SJ, Vogl AW, McNagny KM, Freedman BS. Gene-Edited Human Kidney Organoids Reveal Mechanisms of Disease in Podocyte Development. Stem Cells 2017; 35:2366-2378. [PMID: 28905451 DOI: 10.1002/stem.2707] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/04/2017] [Indexed: 11/07/2022]
Abstract
A critical event during kidney organogenesis is the differentiation of podocytes, specialized epithelial cells that filter blood plasma to form urine. Podocytes derived from human pluripotent stem cells (hPSC-podocytes) have recently been generated in nephron-like kidney organoids, but the developmental stage of these cells and their capacity to reveal disease mechanisms remains unclear. Here, we show that hPSC-podocytes phenocopy mammalian podocytes at the capillary loop stage (CLS), recapitulating key features of ultrastructure, gene expression, and mutant phenotype. hPSC-podocytes in vitro progressively establish junction-rich basal membranes (nephrin+ podocin+ ZO-1+ ) and microvillus-rich apical membranes (podocalyxin+ ), similar to CLS podocytes in vivo. Ultrastructural, biophysical, and transcriptomic analysis of podocalyxin-knockout hPSCs and derived podocytes, generated using CRISPR/Cas9, reveals defects in the assembly of microvilli and lateral spaces between developing podocytes, resulting in failed junctional migration. These defects are phenocopied in CLS glomeruli of podocalyxin-deficient mice, which cannot produce urine, thereby demonstrating that podocalyxin has a conserved and essential role in mammalian podocyte maturation. Defining the maturity of hPSC-podocytes and their capacity to reveal and recapitulate pathophysiological mechanisms establishes a powerful framework for studying human kidney disease and regeneration. Stem Cells 2017;35:2366-2378.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Ido Refaeli
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Craig R Brooks
- Division of Nephrology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Peifeng Jing
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Ramila E Gulieva
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael R Hughes
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nelly M Cruz
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Yannan Liu
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Angela J Churchill
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington, USA
| | - Hongxia Fu
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Bioengineering, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jeffrey W Pippin
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Lih Y Lin
- Department of Electrical Engineering, University of Washington, Seattle, Washington, USA
| | - Stuart J Shankland
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - A Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelly M McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Benjamin S Freedman
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Kidney Research Institute, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Department of Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
68
|
Kaverina NV, Eng DG, Largent AD, Daehn I, Chang A, Gross KW, Pippin JW, Hohenstein P, Shankland SJ. WT1 Is Necessary for the Proliferation and Migration of Cells of Renin Lineage Following Kidney Podocyte Depletion. Stem Cell Reports 2017; 9:1152-1166. [PMID: 28966119 PMCID: PMC5639431 DOI: 10.1016/j.stemcr.2017.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
Wilms' tumor suppressor 1 (WT1) plays an important role in cell proliferation and mesenchymal-epithelial balance in normal development and disease. Here, we show that following podocyte depletion in three experimental models, and in patients with focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, WT1 increased significantly in cells of renin lineage (CoRL). In an animal model of FSGS in RenWt1fl/fl reporter mice with inducible deletion of WT1 in CoRL, CoRL proliferation and migration to the glomerulus was reduced, and glomerular disease was worse compared with wild-type mice. To become podocytes, CoRL undergo mesenchymal-to-epithelial transformation (MET), typified by reduced staining for mesenchymal markers (MYH11, SM22, αSMA) and de novo expression of epithelial markers (E-cadherin and cytokeratin18). Evidence for changes in MET markers was barely detected in RenWt1fl/fl mice. Our results show that following podocyte depletion, WT1 plays essential roles in CoRL proliferation and migration toward an adult podocyte fate.
Collapse
Affiliation(s)
- Natalya V Kaverina
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Andrea D Largent
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Ilse Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA
| | - Anthony Chang
- Department of Pathology, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
69
|
Glucocorticoid therapy regulates podocyte motility by inhibition of Rac1. Sci Rep 2017; 7:6725. [PMID: 28751734 PMCID: PMC5532274 DOI: 10.1038/s41598-017-06810-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/19/2017] [Indexed: 02/03/2023] Open
Abstract
Nephrotic syndrome (NS) occurs when the glomerular filtration barrier becomes excessively permeable leading to massive proteinuria. In childhood NS, immune system dysregulation has been implicated and increasing evidence points to the central role of podocytes in the pathogenesis. Children with NS are typically treated with an empiric course of glucocorticoid (Gc) therapy; a class of steroids that are activating ligands for the glucocorticoid receptor (GR) transcription factor. Although Gc-therapy has been the cornerstone of NS management for decades, the mechanism of action, and target cell, remain poorly understood. We tested the hypothesis that Gc acts directly on the podocyte to produce clinically useful effects without involvement of the immune system. In human podocytes, we demonstrated that the basic GR-signalling mechanism is intact and that Gc induced an increase in podocyte barrier function. Defining the GR-cistrome identified Gc regulation of motility genes. These findings were functionally validated with live-cell imaging. We demonstrated that treatment with Gc reduced the activity of the pro-migratory small GTPase regulator Rac1. Furthermore, Rac1 inhibition had a direct, protective effect on podocyte barrier function. Our studies reveal a new mechanism for Gc action directly on the podocyte, with translational relevance to designing new selective synthetic Gc molecules.
Collapse
|
70
|
Lu Y, Ye Y, Bao W, Yang Q, Wang J, Liu Z, Shi S. Genome-wide identification of genes essential for podocyte cytoskeletons based on single-cell RNA sequencing. Kidney Int 2017; 92:1119-1129. [PMID: 28709640 DOI: 10.1016/j.kint.2017.04.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/06/2017] [Accepted: 04/13/2017] [Indexed: 12/21/2022]
Abstract
Gene expression differs substantially among individual cells of the same type. We speculate that genes that are expressed in all but a portion of cells of a given cell type would be likely essential and required for either the cell survival (housekeeping) or for the cell type's unique structure and function, enabling the organism to survive. Here, we performed RNA-seq of 20 mouse podocytes using the Fluidigm C1 system and identified 335 genes that were expressed in all of them. Among them, 239 genes were also expressed in mesangial and endothelial cells and were involved in energy metabolism, protein synthesis, etc., as housekeeping genes. In contrast, 92 genes were preferentially expressed in podocytes (over five-fold versus expression in mesangial and endothelial cells) and are, therefore, the essential candidate genes specific for podocytes. Assessments by bioinformatics, conserved expression in human podocytes, and association with injury/disease all support the essentiality of these genes for podocytes. Factually, 27 of the 92 genes are already known to be essential for podocyte structure and function. Thirty-seven novel genes were functionally analyzed by siRNA silencing, and we found that a deficiency of 30 genes led to either cytoskeletal injury (FGFR1, AOX1, AIF1L, HAUS8, RAB3B, LPIN2, GOLIM4, CERS6, ARHGEF18, ARPC1A, SRGAP1, ITGB5, ILDR2, MPP5, TSC22D1, DNAJC11, SEPT10, MOCS2, FNBP1L, and TMOD3) or significant downregulation of CD2AP and synaptopodin (IFT80, MYOM2, ANXA4, CYB5R4, GPC1, ZNF277, NSF, ITGAV, CRYAB, and MTSS1). Thus, the list of genes essential for podocyte cytoskeletons is expanded by single-cell RNA sequencing. It appears that podocyte-specific essential genes are mainly associated with podocyte cytoskeletons.
Collapse
Affiliation(s)
- Yuqiu Lu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Yuting Ye
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Wenduona Bao
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Qianqian Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jinquan Wang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China.
| |
Collapse
|
71
|
Bharathavikru R, Dudnakova T, Aitken S, Slight J, Artibani M, Hohenstein P, Tollervey D, Hastie N. Transcription factor Wilms' tumor 1 regulates developmental RNAs through 3' UTR interaction. Genes Dev 2017; 31:347-352. [PMID: 28289143 PMCID: PMC5358755 DOI: 10.1101/gad.291500.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/15/2017] [Indexed: 12/23/2022]
Abstract
Bharathavikru et al. show that Wilms’ tumour 1 (WT1) binds preferentially to 3′ UTRs of developmental targets, which are down-regulated upon WT1 depletion in cell culture and developing kidney mesenchyme. Combining experimental and computational analyses, they propose that WT1 influences key developmental and disease processes in part through regulating mRNA turnover. Wilms’ tumor 1 (WT1) is essential for the development and homeostasis of multiple mesodermal tissues. Despite evidence for post-transcriptional roles, no endogenous WT1 target RNAs exist. Using RNA immunoprecipitation and UV cross-linking, we show that WT1 binds preferentially to 3′ untranslated regions (UTRs) of developmental targets. These target mRNAs are down-regulated upon WT1 depletion in cell culture and developing kidney mesenchyme. Wt1 deletion leads to rapid turnover of specific mRNAs. WT1 regulates reporter gene expression through interaction with 3′ UTR-binding sites. Combining experimental and computational analyses, we propose that WT1 influences key developmental and disease processes in part through regulating mRNA turnover.
Collapse
Affiliation(s)
- Ruthrothaselvi Bharathavikru
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Tatiana Dudnakova
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Stuart Aitken
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Joan Slight
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Mara Artibani
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Peter Hohenstein
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom.,Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - David Tollervey
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Nick Hastie
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
72
|
Ahmed AA, Mohamed AD, Gener M, Li W, Taboada E. YAP and the Hippo pathway in pediatric cancer. Mol Cell Oncol 2017; 4:e1295127. [PMID: 28616573 DOI: 10.1080/23723556.2017.1295127] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/01/2017] [Accepted: 02/10/2017] [Indexed: 12/17/2022]
Abstract
The Hippo pathway is an important signaling pathway that controls cell proliferation and apoptosis. It is evolutionarily conserved in mammals and is stimulated by cell-cell contact, inhibiting cell proliferation in response to increased cell density. During early embryonic development, the Hippo signaling pathway regulates organ development and size, and its functions result in the coordinated balance between proliferation, apoptosis, and differentiation. Its principal effectors, YAP and TAZ, regulate signaling by the embryonic stem cells and determine cell fate and histogenesis. Dysfunction of this pathway contributes to cancer development in adults and children. Emerging studies have shed light on the upregulation of Hippo pathway members in several pediatric cancers and may offer prognostic information on rhabdomyosarcoma, osteosarcoma, Wilms tumor, neuroblastoma, medulloblastoma, and other brain gliomas. We review the results of such published studies and highlight the potential clinical application of this pathway in pediatric oncologic and pathologic studies. These studies support targeting this pathway as a novel treatment strategy.
Collapse
Affiliation(s)
- Atif A Ahmed
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | | | - Melissa Gener
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Weijie Li
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Eugenio Taboada
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| |
Collapse
|
73
|
Cuevas VD, Anta L, Samaniego R, Orta-Zavalza E, Vladimir de la Rosa J, Baujat G, Domínguez-Soto Á, Sánchez-Mateos P, Escribese MM, Castrillo A, Cormier-Daire V, Vega MA, Corbí ÁL. MAFB Determines Human Macrophage Anti-Inflammatory Polarization: Relevance for the Pathogenic Mechanisms Operating in Multicentric Carpotarsal Osteolysis. THE JOURNAL OF IMMUNOLOGY 2017; 198:2070-2081. [PMID: 28093525 DOI: 10.4049/jimmunol.1601667] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022]
Abstract
Macrophage phenotypic and functional heterogeneity derives from tissue-specific transcriptional signatures shaped by the local microenvironment. Most studies addressing the molecular basis for macrophage heterogeneity have focused on murine cells, whereas the factors controlling the functional specialization of human macrophages are less known. M-CSF drives the generation of human monocyte-derived macrophages with a potent anti-inflammatory activity upon stimulation. We now report that knockdown of MAFB impairs the acquisition of the anti-inflammatory profile of human macrophages, identify the MAFB-dependent gene signature in human macrophages and illustrate the coexpression of MAFB and MAFB-target genes in CD163+ tissue-resident and tumor-associated macrophages. The contribution of MAFB to the homeostatic/anti-inflammatory macrophage profile is further supported by the skewed polarization of monocyte-derived macrophages from multicentric carpotarsal osteolysis (Online Mendelian Inheritance in Man #166300), a pathology caused by mutations in the MAFB gene. Our results demonstrate that MAFB critically determines the acquisition of the anti-inflammatory transcriptional and functional profiles of human macrophages.
Collapse
Affiliation(s)
- Víctor D Cuevas
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Laura Anta
- Servicio de Cirugía Ortopédica y Traumatología, Complejo Hospitalario de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Rafael Samaniego
- Laboratorio de Inmuno-Oncología, Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - Emmanuel Orta-Zavalza
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Juan Vladimir de la Rosa
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain
| | - Geneviève Baujat
- Unidad de Biomedicina, Instituto de Investigaciones Biomédicas-Universidad de Las Palmas de Gran Canaria (ULPGC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias de la ULPGC, 35001 Las Palmas, Spain.,Département de Génétique, INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, 75015 Paris, France; and
| | - Ángeles Domínguez-Soto
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Paloma Sánchez-Mateos
- Laboratorio de Inmuno-Oncología, Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - María M Escribese
- Institute for Applied Molecular Medicine, School of Medicine, University CEU San Pablo, Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain
| | - Valérie Cormier-Daire
- Unidad de Biomedicina, Instituto de Investigaciones Biomédicas-Universidad de Las Palmas de Gran Canaria (ULPGC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias de la ULPGC, 35001 Las Palmas, Spain.,Département de Génétique, INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, 75015 Paris, France; and
| | - Miguel A Vega
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain;
| | - Ángel L Corbí
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain;
| |
Collapse
|
74
|
Dysregulation of WTI (-KTS) is Associated with the Kidney-Specific Effects of the LMX1B R246Q Mutation. Sci Rep 2017; 7:39933. [PMID: 28059119 PMCID: PMC5216339 DOI: 10.1038/srep39933] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/28/2016] [Indexed: 01/11/2023] Open
Abstract
Mutations in the LIM homeobox transcription factor 1-beta (LMX1B) are a cause of nail patellar syndrome, a condition characterized by skeletal changes, glaucoma and focal segmental glomerulosclerosis. Recently, a missense mutation (R246Q) in LMX1B was reported as a cause of glomerular pathologies without extra-renal manifestations, otherwise known as nail patella-like renal disease (NPLRD). We have identified two additional NPLRD families with the R246Q mutation, though the mechanisms by which LMX1BR246Q causes a renal-specific phenotype is unknown. In this study, using human podocyte cell lines overexpressing either myc-LMX1BWT or myc-LMX1BR246Q, we observed dominant negative and haploinsufficiency effects of the mutation on the expression of podocyte genes such as NPHS1, GLEPP1, and WT1. Specifically, we observed a novel LMX1BR246Q-mediated downregulation of WT1(−KTS) isoforms in podocytes. In conclusion, we have shown that the renal-specific phenotype associated with the LMX1BR246Q mutation may be due to a dominant negative effect on WT1(−KTS) isoforms that may cause a disruption of the WT1 (−KTS):(+KTS) isoform ratio and a decrease in the expression of podocyte genes. Full delineation of the LMX1B gene regulon is needed to define its role in maintenance of glomerular filtration barrier integrity.
Collapse
|
75
|
Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnol Adv 2016; 35:150-167. [PMID: 28017905 DOI: 10.1016/j.biotechadv.2016.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/12/2016] [Accepted: 12/17/2016] [Indexed: 02/09/2023]
Abstract
The global rise in the numbers of kidney patients and the shortage in transplantable organs have led to an increasing interest in kidney-specific regenerative therapies, renal disease modelling and bioartificial kidneys. Sources for large quantities of high-quality renal cells and tissues would be required, also for applications in in vitro platforms for compound safety and efficacy screening. Stem cell-based approaches for the generation of renal-like cells and tissues would be most attractive, but such methods were not available until recently. This situation has drastically changed since 2013, and various protocols for the generation of renal-like cells and precursors from pluripotent stem cells (PSC) have been established. The most recent breakthroughs were related to the establishment of various protocols for the generation of PSC-derived kidney organoids. In combination with recent advances in genome editing, bioprinting and the establishment of predictive renal screening platforms this results in exciting new possibilities. This review will give a comprehensive overview over current PSC-based protocols for the generation of renal-like cells, precursors and organoids, and their current and potential applications in regenerative medicine, compound screening, disease modelling and bioartificial organs.
Collapse
Affiliation(s)
- Jacqueline Kai Chin Chuah
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| |
Collapse
|
76
|
Saito R, Rocanin-Arjo A, You YH, Darshi M, Van Espen B, Miyamoto S, Pham J, Pu M, Romoli S, Natarajan L, Ju W, Kretzler M, Nelson R, Ono K, Thomasova D, Mulay SR, Ideker T, D'Agati V, Beyret E, Belmonte JCI, Anders HJ, Sharma K. Systems biology analysis reveals role of MDM2 in diabetic nephropathy. JCI Insight 2016; 1:e87877. [PMID: 27777973 DOI: 10.1172/jci.insight.87877] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To derive new insights in diabetic complications, we integrated publicly available human protein-protein interaction (PPI) networks with global metabolic networks using metabolomic data from patients with diabetic nephropathy. We focused on the participating proteins in the network that were computationally predicted to connect the urine metabolites. MDM2 had the highest significant number of PPI connections. As validation, significant downregulation of MDM2 gene expression was found in both glomerular and tubulointerstitial compartments of kidney biopsy tissue from 2 independent cohorts of patients with diabetic nephropathy. In diabetic mice, chemical inhibition of MDM2 with Nutlin-3a led to reduction in the number of podocytes, increased blood urea nitrogen, and increased mortality. Addition of Nutlin-3a decreased WT1+ cells in embryonic kidneys. Both podocyte- and tubule-specific MDM2-knockout mice exhibited severe glomerular and tubular dysfunction, respectively. Interestingly, the only 2 metabolites that were reduced in both podocyte and tubule-specific MDM2-knockout mice were 3-methylcrotonylglycine and uracil, both of which were also reduced in human diabetic kidney disease. Thus, our bioinformatics tool combined with multi-omics studies identified an important functional role for MDM2 in glomeruli and tubules of the diabetic nephropathic kidney and links MDM2 to a reduction in 2 key metabolite biomarkers.
Collapse
Affiliation(s)
- Rintaro Saito
- Institute of Metabolomic Medicine.,Center for Renal Translational Medicine, Division of Nephrology-Hypertension.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Anaïs Rocanin-Arjo
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Young-Hyun You
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Manjula Darshi
- Institute of Metabolomic Medicine.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Benjamin Van Espen
- Institute of Metabolomic Medicine.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Satoshi Miyamoto
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Jessica Pham
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Minya Pu
- Institute of Metabolomic Medicine.,Department of Family Medicine and Epidemiology, UCSD, San Diego, California, USA
| | - Simone Romoli
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Loki Natarajan
- Institute of Metabolomic Medicine.,Department of Family Medicine and Epidemiology, UCSD, San Diego, California, USA
| | - Wenjun Ju
- Department of Internal Medicine, Nephrology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Nephrology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Keiichiro Ono
- Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Dana Thomasova
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Shrikant R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Trey Ideker
- Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA
| | - Vivette D'Agati
- Renal Pathology Laboratory, Columbia University, College of Physicians and Surgeons, Department of Pathology, New York, New York, USA
| | - Ergin Beyret
- Salk Institute for Biological Studies, San Diego, California, USA
| | | | - Hans Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Kumar Sharma
- Institute of Metabolomic Medicine.,Center for Renal Translational Medicine, Division of Nephrology-Hypertension.,Division of Medical Genetics, Department of Medicine, UCSD, San Diego, California, USA.,Veterans Affairs Health Systems, San Diego, California, USA
| |
Collapse
|
77
|
Pereira EM, Labilloy A, Eshbach ML, Roy A, Subramanya AR, Monte S, Labilloy G, Weisz OA. Characterization and phosphoproteomic analysis of a human immortalized podocyte model of Fabry disease generated using CRISPR/Cas9 technology. Am J Physiol Renal Physiol 2016; 311:F1015-F1024. [PMID: 27681560 DOI: 10.1152/ajprenal.00283.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/20/2016] [Indexed: 01/22/2023] Open
Abstract
Fabry nephropathy is a major cause of morbidity and premature death in patients with Fabry disease (FD), a rare X-linked lysosomal storage disorder. Gb3, the main substrate of α-galactosidase A (α-Gal A), progressively accumulates within cells in a variety of tissues. Establishment of cell models has been useful as a tool for testing hypotheses of disease pathogenesis. We applied CRISPR/Cas9 genome editing technology to the GLA gene to develop human kidney cell models of FD in human immortalized podocytes, which are the main affected renal cell type. Our podocytes lack detectable α-Gal A activity and have increased levels of Gb3. To explore different pathways that could have distinct patterns of activation under conditions of α-gal A deficiency, we used a high-throughput antibody array to perform phosphorylation profiling of CRISPR/Cas9-edited and control podocytes. Changes in both total protein levels and in phosphorylation status per site were observed. Analysis of our candidate proteins suggests that multiple signaling pathways are impaired in FD.
Collapse
Affiliation(s)
- Ester M Pereira
- Laboratory of Immunogenetics and Molecular Biology, Federal University of Piaui, Teresina, Brazil
| | - Anatália Labilloy
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Megan L Eshbach
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Ankita Roy
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Semiramis Monte
- Laboratory of Immunogenetics and Molecular Biology, Federal University of Piaui, Teresina, Brazil;
| | - Guillaume Labilloy
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| |
Collapse
|
78
|
Repression of CMIP transcription by WT1 is relevant to podocyte health. Kidney Int 2016; 90:1298-1311. [PMID: 27650733 DOI: 10.1016/j.kint.2016.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 07/11/2016] [Accepted: 07/14/2016] [Indexed: 12/23/2022]
Abstract
The WT1 (Wilm's tumor suppressor) gene is expressed throughout life in podocytes and is essential for the functional integrity of the glomerular filtration barrier. We have previously shown that CMIP (C-Maf inducing protein) is overproduced in podocyte diseases and alters intracellular signaling. Here we isolated the proximal region of the human CMIP promoter and showed by chromatin immunoprecipitation assays and electrophoretic-mobility shift that Wilm's tumor protein (WT1) bound to 2 WT1 response elements, located at positions -290/-274 and -57/-41 relative to transcription start site. Unlike the human CMIP gene, only one Wt1 response element was identified in the mouse Cmip proximal promoter located at position -217/-206. Luciferase reporter assays indicated that WT1 dose-dependently inhibited the transcriptional induction of the CMIP promoter. Transfection of decoy oligonucleotides mimicking the WT1 response elements prevented the inhibition of WT1 on CMIP promoter activity. Furthermore, WT1 silencing promoted Cmip expression. In line with these findings, the abundance of Cmip was early and significantly increased at the transcript and protein level in podocytes displaying a primary defect in Wt1, including Denys-Drash syndrome and Frasier syndrome. Thus, WT1 is a major repressor of the CMIP gene in physiological situations, while conditional deletion of CMIP in the developing kidney did not affect the development of mature glomeruli.
Collapse
|
79
|
Ullmark T, Järvstråt L, Sandén C, Montano G, Jernmark-Nilsson H, Lilljebjörn H, Lennartsson A, Fioretos T, Drott K, Vidovic K, Nilsson B, Gullberg U. Distinct global binding patterns of the Wilms tumor gene 1 (WT1) -KTS and +KTS isoforms in leukemic cells. Haematologica 2016; 102:336-345. [PMID: 27612989 DOI: 10.3324/haematol.2016.149815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/05/2016] [Indexed: 12/29/2022] Open
Abstract
The zinc finger transcription factor Wilms tumor gene 1 (WT1) acts as an oncogene in acute myeloid leukemia. A naturally occurring alternative splice event between zinc fingers three and four, removing or retaining three amino acids (±KTS), is believed to change the DNA binding affinity of WT1, although there are conflicting data regarding the binding affinity and motifs of the different isoforms. Increased expression of the WT1 -KTS isoform at the expense of the WT1 +KTS isoform is associated with poor prognosis in acute myeloid leukemia. We determined the genome-wide binding pattern of WT1 -KTS and WT1 +KTS in leukemic K562 cells by chromatin immunoprecipitation and deep sequencing. We discovered that the WT1 -KTS isoform predominantly binds close to transcription start sites and to enhancers, in a similar fashion to other transcription factors, whereas WT1 +KTS binding is enriched within gene bodies. We observed a significant overlap between WT1 -KTS and WT1 +KTS target genes, despite the binding sites being distinct. Motif discovery revealed distinct binding motifs for the isoforms, some of which have been previously reported as WT1 binding sites. Additional analyses showed that both WT1 -KTS and WT1 +KTS target genes are more likely to be transcribed than non-targets, and are involved in cell proliferation, cell death, and development. Our study provides evidence that WT1 -KTS and WT1 +KTS share target genes yet still bind distinct locations, indicating isoform-specific regulation in transcription of genes related to cell proliferation and differentiation, consistent with the involvement of WT1 in acute myeloid leukemia.
Collapse
Affiliation(s)
- Tove Ullmark
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Linnea Järvstråt
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Carl Sandén
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Giorgia Montano
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Helena Jernmark-Nilsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Henrik Lilljebjörn
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Kristina Drott
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Karina Vidovic
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| | - Urban Gullberg
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Huddinge, Sweden
| |
Collapse
|
80
|
Rinschen MM, Schroeter CB, Koehler S, Ising C, Schermer B, Kann M, Benzing T, Brinkkoetter PT. Quantitative deep mapping of the cultured podocyte proteome uncovers shifts in proteostatic mechanisms during differentiation. Am J Physiol Cell Physiol 2016; 311:C404-17. [PMID: 27357545 DOI: 10.1152/ajpcell.00121.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/27/2016] [Indexed: 11/22/2022]
Abstract
The renal filtration barrier is maintained by the renal podocyte, an epithelial postmitotic cell. Immortalized mouse podocyte cell lines-both in the differentiated and undifferentiated state-are widely utilized tools to estimate podocyte injury and cytoskeletal rearrangement processes in vitro. Here, we mapped the cultured podocyte proteome at a depth of more than 8,800 proteins and quantified 7,240 proteins. Copy numbers of proteins mutated in forms of hereditary nephrotic syndrome or focal segmental glomerulosclerosis (FSGS) were assessed. We found that cultured podocytes express abundant copy numbers of endogenous receptors, such as tyrosine kinase membrane receptors, the G protein-coupled receptor (GPCR), NPR3 (ANP receptor), and several poorly characterized GPCRs. The data set was correlated with deep mapping mRNA sequencing ("mRNAseq") data from the native mouse podocyte, the native mouse podocyte proteome and staining intensities from the human protein atlas. The generated data set was similar to these previously published resources, but several native and high-abundant podocyte-specific proteins were not identified in the data set. Notably, this data set detected general perturbations in proteostatic mechanisms as a dominant alteration during podocyte differentiation, with high proteasome activity in the undifferentiated state and markedly increased expression of lysosomal proteins in the differentiated state. Phosphoproteomics analysis of mouse podocytes at a resolution of more than 3,000 sites suggested a preference of phosphorylation of actin filament-associated proteins in the differentiated state. The data set obtained here provides a resource and provides the means for deep mapping of the native podocyte proteome and phosphoproteome in a similar manner.
Collapse
Affiliation(s)
- Markus M Rinschen
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Christina B Schroeter
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sybille Koehler
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Christina Ising
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Martin Kann
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; and Systems Biology of Ageing Cologne, SybaCol, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
81
|
Koehler S, Tellkamp F, Niessen CM, Bloch W, Kerjaschki D, Schermer B, Benzing T, Brinkkoetter PT. Par3A is dispensable for the function of the glomerular filtration barrier of the kidney. Am J Physiol Renal Physiol 2016; 311:F112-9. [PMID: 27122542 DOI: 10.1152/ajprenal.00171.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/22/2016] [Indexed: 01/08/2023] Open
Abstract
Polarity signaling through the atypical PKC (aPKC)-Par polarity complex is essential for the development and maintenance of the podocyte architecture and the function of the glomerular filtration barrier of the kidney. To study the contribution of Par3A in this complex, we generated a novel Pard3 podocyte-specific knockout mouse model by targeting exon 6 of the Pard3 gene. Genetic deletion of Pard3a did not impair renal function, neither at birth nor later in life. Even challenging the animals did not result in glomerular disease. Despite its well-established role in aPKC-mediated signaling, Par3A appears to be dispensable for the function of the glomerular filtration barrier. Moreover, its homolog Pard3b, and not Pard3a, is the dominant Par3 gene expressed in podocytes and found at the basis of the slit diaphragm, where it partially colocalizes with podocin. In conclusion, Par3A function is either dispensable for slit diaphragm integrity, or compensatory mechanisms and a high redundancy of the different polarity proteins, including Par3B, Lgl, or PALS1, maintain the function of the glomerular filtration barrier, even in the absence of Par3A.
Collapse
Affiliation(s)
- Sybille Koehler
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Frederik Tellkamp
- Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Dermatology, University Hospital of Cologne, Cologne, Germany
| | - Carien M Niessen
- Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Dermatology, University Hospital of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany, and
| | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Schermer
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine, University of Cologne, Cologne, Germany;
| |
Collapse
|
82
|
Keyvani Chahi A, Martin CE, Jones N. Nephrin Suppresses Hippo Signaling through the Adaptor Proteins Nck and WTIP. J Biol Chem 2016; 291:12799-12808. [PMID: 27033705 DOI: 10.1074/jbc.m116.724245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Indexed: 11/06/2022] Open
Abstract
Podocytes are key components of the kidney blood filtration barrier, and their ability to withstand hemodynamic strain is proposed to be closely tied to their unique and flexible cytoarchitecture. However, the mechanisms that control such mechanotransduction are poorly understood. We have previously established that tyrosine phosphorylation of the transmembrane protein nephrin promotes recruitment of the Nck1/2 cytoskeletal adaptor proteins and downstream actin remodeling. We now reveal that Nck integrates nephrin with the Hippo kinase cascade through association with the adaptor protein WTIP. Using mutational analysis, we show that Nck sequesters WTIP and its binding partner Lats1 to phosphorylated nephrin, resulting in decreased phospho-activation of Lats1. We further demonstrate that, coincident with nephrin dephosphorylation in a transient model of podocyte injury in mice, Lats1 is rapidly activated, and this precedes significant down-regulation of the transcription regulator Yap. Moreover, we show reduced levels of Yap protein in mice with chronic disruption of nephrin phospho-signaling. Together, these findings support the existence of a dynamic molecular link between nephrin signaling and the canonical Hippo pathway in podocytes, which may facilitate the conversion of mechanical cues to biochemical signals promoting podocyte viability.
Collapse
Affiliation(s)
- Ava Keyvani Chahi
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Claire E Martin
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Nina Jones
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
83
|
|
84
|
Dong L, Pietsch S, Englert C. Towards an understanding of kidney diseases associated with WT1 mutations. Kidney Int 2015; 88:684-90. [PMID: 26154924 PMCID: PMC4687464 DOI: 10.1038/ki.2015.198] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/12/2015] [Indexed: 12/23/2022]
Abstract
Mutations in Wilms' tumor 1 (WT1) cause a wide spectrum of renal manifestations, eventually leading to end-stage kidney failure. Insufficient understanding of WT1's molecular functions in kidney development has hampered efficient therapeutic applications for WT1-associated diseases. Recently, the generation and characterization of mouse models and application of multiple state-of-the-art approaches have significantly expanded our understanding of the molecular mechanisms of how WT1 mutations lead to kidney failure. Here, we discuss the WT1 binding consensus and illustrate the major roles of WT1 in different cell populations in kidney biology. WT1 controls metanephric mesenchyme (MM) self-renewal and proliferation mainly by regulating FGF and BMP-pSMAD signaling pathways as well as Sall1 and Pax2, encoding key transcription factors; WT1 drives MM differentiation and mesenchyme–epithelial transition by targeting Fgf8 and Wnt4; WT1 defines podocyte identity by activation of other podocyte-specific transcription factors, including Mafb, Lmx1b, FoxC2, and Tcf21. These factors potentially cooperate with WT1 regulating the expression of components and regulators of the cytoskeleton for establishing podocyte polarity, slit diaphragm structure, and focal adhesion to the glomerular basement membrane. Understanding of WT1's function in kidney biology including WT1-regulated pathways will give insights that will eventually help therapeutic applications.
Collapse
Affiliation(s)
- Lihua Dong
- Molecular Genetics, Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
| | - Stefan Pietsch
- Molecular Genetics, Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
| | - Christoph Englert
- Molecular Genetics, Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany.,Faculty of Biology and Pharmacy, Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
85
|
Wnt/β-catenin signalling and podocyte dysfunction in proteinuric kidney disease. Nat Rev Nephrol 2015; 11:535-45. [PMID: 26055352 DOI: 10.1038/nrneph.2015.88] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Podocytes are unique, highly specialized, terminally differentiated cells that are integral components of the kidney glomerular filtration barrier. Podocytes are vulnerable to a variety of injuries and in response they undergo a series of changes ranging from hypertrophy, autophagy, dedifferentiation, mesenchymal transition and detachment to apoptosis, depending on the nature and extent of the insult. Emerging evidence indicates that Wnt/β-catenin signalling has a central role in mediating podocyte dysfunction and proteinuria. Wnts are induced and β-catenin is activated in podocytes in various proteinuric kidney diseases. Genetic or pharmacologic activation of β-catenin is sufficient to impair podocyte integrity and causes proteinuria in healthy mice, whereas podocyte-specific ablation of β-catenin protects against proteinuria after kidney injury. Mechanistically, Wnt/β-catenin controls the expression of several key mediators implicated in podocytopathies, including Snail1, the renin-angiotensin system and matrix metalloproteinase 7. Wnt/β-catenin also negatively regulates Wilms tumour protein, a crucial transcription factor that safeguards podocyte integrity. Targeted inhibition of Wnt/β-catenin signalling preserves podocyte integrity and ameliorates proteinuria in animal models. This Review highlights advances in our understanding of the pathomechanisms of Wnt/β-catenin signalling in mediating podocyte injury, and describes the therapeutic potential of targeting this pathway for the treatment of proteinuric kidney disease.
Collapse
|