1
|
Williamson CR, Pantic UV, Wang AY, Jones N. Revisiting nephrin signaling and its specialized effects on the uniquely adaptable podocyte. Biochem J 2025; 482:BCJ20230234. [PMID: 40457651 DOI: 10.1042/bcj20230234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 05/19/2025] [Indexed: 06/29/2025]
Abstract
Nephrin is a transmembrane Ig-like domain-containing protein that serves as a central structural and signaling scaffold in kidney filtration. First identified in 1998 as mutated in congenital nephrotic syndrome, the recent identification of nephrin autoantibodies in acquired kidney diseases has sparked renewed interest in nephrin biology. In specialized cells known as podocytes, nephrin helps establish and maintain the slit diaphragm (SD), a unique cell-cell junction formed between interdigitating cell projections known as foot processes (FPs). Together, the SD and FP are among the first stages of renal filtration, where they are subject to numerous biochemical and mechanical stressors. Although podocytes are highly adapted to this environment, over time and with injury, this elevated strain can lead to pathological structural changes, detachment, and proteinuria. As such, the complex set of signaling mechanisms provided by nephrin are essential for controlling podocyte adaptability. Herein, we provide a thorough and up-to-date review on nephrin signaling, including a focus on cross-talk between nephrin interactors and signaling regions across podocytes. We first highlight new findings regarding podocyte structure and function, followed by an emphasis on why nephrin is among the most critical proteins for maintaining these features. We then detail a comprehensive list of known nephrin interactors and describe several of their effects, including calcium regulation, cell survival, cell polarity, phase separation-mediated actin reorganization, and SD-focal adhesion dynamics. Collectively, our emerging understanding of the broader cellular context of nephrin signaling provides important insight for clinical strategies to mitigate podocyte injury and kidney disease progression.
Collapse
Affiliation(s)
- Casey R Williamson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Una V Pantic
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Alice Y Wang
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
2
|
Williamson CR, Jones N. Reduced Nephrin Tyrosine Phosphorylation Enhances Insulin Secretion and Increases Glucose Tolerance With Age. Endocrinology 2024; 165:bqae078. [PMID: 38954536 PMCID: PMC11247170 DOI: 10.1210/endocr/bqae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Nephrin is a transmembrane protein with well-established signaling roles in kidney podocytes, and a smaller set of secretory functions in pancreatic β cells are implicated in diabetes. Nephrin signaling is mediated in part through its 3 cytoplasmic YDxV motifs, which can be tyrosine phosphorylated by high glucose and β cell injuries. Although in vitro studies demonstrate these phosphorylated motifs can regulate β cell vesicle trafficking and insulin release, in vivo evidence of their role in this cell type remains to be determined. METHODS To further explore the role of nephrin YDxV phosphorylation in β cells, we used a mouse line with tyrosine to phenylalanine substitutions at each YDxV motif (nephrin-Y3F) to inhibit phosphorylation. We assessed islet function via primary islet glucose-stimulated insulin secretion assays and oral glucose tolerance tests. RESULTS Nephrin-Y3F mice successfully developed pancreatic endocrine and exocrine tissues with minimal structural differences. Unexpectedly, male and female nephrin-Y3F mice showed elevated insulin secretion, with a stronger increase observed in male mice. At 8 months of age, no differences in glucose tolerance were observed between wild-type (WT) and nephrin-Y3F mice. However, aged nephrin-Y3F mice (16 months of age) demonstrated more rapid glucose clearance compared to WT controls. CONCLUSION Taken together, loss of nephrin YDxV phosphorylation does not alter baseline islet function. Instead, our data suggest a mechanism linking impaired nephrin YDxV phosphorylation to improved islet secretory ability with age. Targeting nephrin phosphorylation could provide novel therapeutic opportunities to improve β cell function.
Collapse
Affiliation(s)
- Casey R Williamson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
3
|
Wang J, Casimiro-Garcia A, Johnson BG, Duffen J, Cain M, Savary L, Wang S, Nambiar P, Lech M, Zhao S, Xi L, Zhan Y, Olson J, Stejskal JA, Lin H, Zhang B, Martinez RV, Masek-Hammerman K, Schlerman FJ, Dower K. A protein kinase C α and β inhibitor blunts hyperphagia to halt renal function decline and reduces adiposity in a rat model of obesity-driven type 2 diabetes. Sci Rep 2023; 13:16919. [PMID: 37805649 PMCID: PMC10560236 DOI: 10.1038/s41598-023-43759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 09/28/2023] [Indexed: 10/09/2023] Open
Abstract
Type 2 diabetes (T2D) and its complications can have debilitating, sometimes fatal consequences for afflicted individuals. The disease can be difficult to control, and therapeutic strategies to prevent T2D-induced tissue and organ damage are needed. Here we describe the results of administering a potent and selective inhibitor of Protein Kinase C (PKC) family members PKCα and PKCβ, Cmpd 1, in the ZSF1 obese rat model of hyperphagia-induced, obesity-driven T2D. Although our initial intent was to evaluate the effect of PKCα/β inhibition on renal damage in this model setting, Cmpd 1 unexpectedly caused a marked reduction in the hyperphagic response of ZSF1 obese animals. This halted renal function decline but did so indirectly and indistinguishably from a pair feeding comparator group. However, above and beyond this food intake effect, Cmpd 1 lowered overall animal body weights, reduced liver vacuolation, and reduced inguinal adipose tissue (iWAT) mass, inflammation, and adipocyte size. Taken together, Cmpd 1 had strong effects on multiple disease parameters in this obesity-driven rodent model of T2D. Further evaluation for potential translation of PKCα/β inhibition to T2D and obesity in humans is warranted.
Collapse
Affiliation(s)
- Ju Wang
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| | | | - Bryce G Johnson
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Duffen
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Michael Cain
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Mediar Therapeutics, Boston, MA, USA
| | - Leigh Savary
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Instem Life Science Systems Ltd, Mount Ida College, South Hadley, MA, USA
| | - Stephen Wang
- Pharmacokinetics and Drug Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Novartis Gene Therapies, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Prashant Nambiar
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Strand Therapeutics, Cambridge, MA, USA
| | - Matthew Lech
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Shanrong Zhao
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Amunix Pharmaceuticals, San Francisco, CA, USA
| | - Li Xi
- Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Yutian Zhan
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Jennifer Olson
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
| | - James A Stejskal
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, CT, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Hank Lin
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Data Sciences, Biogen, Cambridge, MA, USA
| | - Robert V Martinez
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
- Center for Technological Innovation, Pfizer Worldwide Research and Development, San Francisco, CA, USA
| | | | - Franklin J Schlerman
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Ken Dower
- Inflammation and Immunology, Pfizer Worldwide Research and Development, Cambridge, MA, USA.
| |
Collapse
|
4
|
Yasuda H, Fukusumi Y, Zhang Y, Kawachi H. 14-3-3 Proteins stabilize actin and vimentin filaments to maintain processes in renal glomerular podocyte. FASEB J 2023; 37:e23168. [PMID: 37651095 DOI: 10.1096/fj.202300865r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
14-3-3 proteins are a ubiquitously expressed family of adaptor proteins. Despite exhibiting high sequence homology, several 14-3-3 isoforms have isoform-specific binding partners and roles. We reported that 14-3-3β interacts with FKBP12 and synaptopodin to maintain the structure of actin fibers in podocytes. However, the precise localization and differential role of 14-3-3 isoforms in kidneys are unclear. Herein, we showed that 14-3-3β in glomeruli was restricted in podocytes, and 14-3-3σ in glomeruli was expressed in podocytes and mesangial cells. Although 14-3-3β was dominantly co-localized with FKBP12 in the foot processes, a part of 14-3-3β was co-localized with Par3 at the slit diaphragm. 14-3-3β interacted with Par3, and FKBP12 bound to 14-3-3β competitively with Par3. Deletion of 14-3-3β enhanced the interaction of Par3 with Par6 in podocytes. Gene silencing for 14-3-3β altered the structure of actin fibers and process formation. 14-3-3β and synaptopodin expression was decreased in podocyte injury models. In contrast, 14-3-3σ in podocytes was expressed in the primary processes. 14-3-3σ interacted with vimentin but not with the actin-associated proteins FKBP12 and synaptopodin. Gene silencing for 14-3-3σ altered the structure of vimentin fibers and process formation. 14-3-3σ and vimentin expression was increased in the early phase of podocyte injury models but was decreased in the late stage. Together, the localization of 14-3-3β at actin cytoskeleton plays a role in maintaining the foot processes and the Par complex in podocytes. In contrast, 14-3-3σ at vimentin cytoskeleton is essential for maintaining primary processes.
Collapse
Affiliation(s)
- Hidenori Yasuda
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ying Zhang
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
5
|
Kostovska I, Trajkovska KT, Topuzovska S, Cekovska S, Labudovic D, Kostovski O, Spasovski G. Nephrinuria and podocytopathies. Adv Clin Chem 2022; 108:1-36. [PMID: 35659057 DOI: 10.1016/bs.acc.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The discovery of nephrin in 1998 has launched a new era in glomerular diseases research, emphasizing its crucial role in the structure and function of the glomerular filtration barrier. In the past 20 years, substantial advances have been made in understanding podocyte structure and function as well as the discovery of several podocyte-related proteins including nephrin. The glomerular filtration barrier is comprised of podocytes, the glomerular basement membrane and endothelial cells. Podocytes, with their specialized slit diaphragm, form the essential backbone of the glomerular filtration barrier. Nephrin is a crucial structural and functional feature of the slit diaphragm that prevents plasma protein, blood cell and macromolecule leakage into the urine. Podocyte damage results in nephrin release. Podocytopathies are kidney diseases in which podocyte damage drives proteinuria, i.e., nephrotic syndrome. Many kidney diseases involve podocytopathy including congenital nephrotic syndrome of Finnish type, diffuse mesangial sclerosis, minimal change disease, focal segmental glomerulosclerosis, collapsing glomerulonephropathy, diabetic nephropathy, lupus nephropathy, hypertensive nephropathy and preeclampsia. Recently, urinary nephrin measurement has become important in the early detection of podocytopathies. In this chapter, we elaborate the main structural and functional features of nephrin as a podocyte-specific protein, pathomechanisms of podocytopathies which result in nephrinuria, highlight the most commonly used methods for detecting urinary nephrin and investigate the diagnostic, prognostic and potential therapeutic relevance of urinary nephrin in primary and secondary proteinuric kidney diseases.
Collapse
Affiliation(s)
- Irena Kostovska
- Department of Medical and Experimental Biochemistry, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia.
| | - Katerina Tosheska Trajkovska
- Department of Medical and Experimental Biochemistry, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Sonja Topuzovska
- Department of Medical and Experimental Biochemistry, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Svetlana Cekovska
- Department of Medical and Experimental Biochemistry, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Danica Labudovic
- Department of Medical and Experimental Biochemistry, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Ognen Kostovski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| | - Goce Spasovski
- University Clinic of Nephrology, Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, North Macedonia
| |
Collapse
|
6
|
PI(4,5)P2 controls slit diaphragm formation and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci 2022; 79:248. [PMID: 35437696 PMCID: PMC9016003 DOI: 10.1007/s00018-022-04273-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 12/03/2022]
Abstract
Drosophila nephrocytes are an emerging model system for mammalian podocytes and proximal tubules as well as for the investigation of kidney diseases. Like podocytes, nephrocytes exhibit characteristics of epithelial cells, but the role of phospholipids in polarization of these cells is yet unclear. In epithelia, phosphatidylinositol(4,5)bisphosphate (PI(4,5)P2) and phosphatidylinositol(3,4,5)-trisphosphate (PI(3,4,5)P3) are asymmetrically distributed in the plasma membrane and determine apical–basal polarity. Here, we demonstrate that both phospholipids are present in the plasma membrane of nephrocytes, but only PI(4,5)P2 accumulates at slit diaphragms. Knockdown of Skittles, a phosphatidylinositol(4)phosphate 5-kinase, which produces PI(4,5)P2, abolished slit diaphragm formation and led to strongly reduced endocytosis. Notably, reduction in PI(3,4,5)P3 by overexpression of PTEN or expression of a dominant-negative phosphatidylinositol-3-kinase did not affect nephrocyte function, whereas enhanced formation of PI(3,4,5)P3 by constitutively active phosphatidylinositol-3-kinase resulted in strong slit diaphragm and endocytosis defects by ectopic activation of the Akt/mTOR pathway. Thus, PI(4,5)P2 but not PI(3,4,5)P3 is essential for slit diaphragm formation and nephrocyte function. However, PI(3,4,5)P3 has to be tightly controlled to ensure nephrocyte development.
Collapse
|
7
|
Bali N, Lee HK(P, Zinn K. Sticks and Stones, a conserved cell surface ligand for the Type IIa RPTP Lar, regulates neural circuit wiring in Drosophila. eLife 2022; 11:e71469. [PMID: 35356892 PMCID: PMC9000958 DOI: 10.7554/elife.71469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Type IIa receptor-like protein tyrosine phosphatases (RPTPs) are essential for neural development. They have cell adhesion molecule (CAM)-like extracellular domains that interact with cell-surface ligands and coreceptors. We identified the immunoglobulin superfamily CAM Sticks and Stones (Sns) as a new partner for the Drosophila Type IIa RPTP Lar. Lar and Sns bind to each other in embryos and in vitro, and the human Sns ortholog, Nephrin, binds to human Type IIa RPTPs. Genetic analysis shows that Lar and Sns function together to regulate larval neuromuscular junction development, axon guidance in the mushroom body (MB), and innervation of the optic lobe (OL) medulla by R7 photoreceptors. In the neuromuscular system, Lar and Sns are both required in motor neurons, and may function as coreceptors. In the MB and OL, however, the relevant Lar-Sns interactions are in trans (between neurons), so Sns functions as a Lar ligand in these systems.
Collapse
Affiliation(s)
- Namrata Bali
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Hyung-Kook (Peter) Lee
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
8
|
van de Leemput J, Wen P, Han Z. Using Drosophila Nephrocytes to Understand the Formation and Maintenance of the Podocyte Slit Diaphragm. Front Cell Dev Biol 2022; 10:837828. [PMID: 35265622 PMCID: PMC8898902 DOI: 10.3389/fcell.2022.837828] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
The podocyte slit diaphragm (SD) is an essential component of the glomerular filtration barrier and its disruption is a common cause of proteinuria and many types of kidney disease. Therefore, better understanding of the pathways and proteins that play key roles in SD formation and maintenance has been of great interest. Podocyte and SD biology have been mainly studied using mouse and other vertebrate models. However, vertebrates are limited by inherent properties and technically challenging in vivo access to the podocytes. Drosophila is a relatively new alternative model system but it has already made great strides. Past the initial obvious differences, mammalian podocytes and fly nephrocytes are remarkably similar at the genetic, molecular and functional levels. This review discusses SD formation and maintenance, and their dependence on cell polarity, the cytoskeleton, and endo- and exocytosis, as learned from studies in fly nephrocytes and mammalian podocytes. In addition, it reflects on the remaining gaps in our knowledge, the physiological implications for glomerular diseases and how we can leverage the advantages Drosophila has to offer to further our understanding.
Collapse
Affiliation(s)
- Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Pei Wen
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Baskaran Y, Tay FPL, Ng EYW, Swa CLF, Wee S, Gunaratne J, Manser E. Proximity proteomics identifies PAK4 as a component of Afadin-Nectin junctions. Nat Commun 2021; 12:5315. [PMID: 34493720 PMCID: PMC8423818 DOI: 10.1038/s41467-021-25011-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Human PAK4 is an ubiquitously expressed p21-activated kinase which acts downstream of Cdc42. Since PAK4 is enriched in cell-cell junctions, we probed the local protein environment around the kinase with a view to understanding its location and substrates. We report that U2OS cells expressing PAK4-BirA-GFP identify a subset of 27 PAK4-proximal proteins that are primarily cell-cell junction components. Afadin/AF6 showed the highest relative biotin labelling and links to the nectin family of homophilic junctional proteins. Reciprocally >50% of the PAK4-proximal proteins were identified by Afadin BioID. Co-precipitation experiments failed to identify junctional proteins, emphasizing the advantage of the BioID method. Mechanistically PAK4 depended on Afadin for its junctional localization, which is similar to the situation in Drosophila. A highly ranked PAK4-proximal protein LZTS2 was immuno-localized with Afadin at cell-cell junctions. Though PAK4 and Cdc42 are junctional, BioID analysis did not yield conventional cadherins, indicating their spatial segregation. To identify cellular PAK4 substrates we then assessed rapid changes (12') in phospho-proteome after treatment with two PAK inhibitors. Among the PAK4-proximal junctional proteins seventeen PAK4 sites were identified. We anticipate mammalian group II PAKs are selective for the Afadin/nectin sub-compartment, with a demonstrably distinct localization from tight and cadherin junctions.
Collapse
Affiliation(s)
- Yohendran Baskaran
- sGSK Group, Institute of Molecular & Cell Biology, A*STAR, Singapore, Singapore
| | - Felicia Pei-Ling Tay
- FB Laboratory, Institute of Molecular & Cell Biology, A*STAR, Singapore, Singapore
| | - Elsa Yuen Wai Ng
- sGSK Group, Institute of Molecular & Cell Biology, A*STAR, Singapore, Singapore
| | - Claire Lee Foon Swa
- Quantitative Proteomics Group, Institute of Molecular & Cell Biology, Singapore, Singapore
| | - Sheena Wee
- Quantitative Proteomics Group, Institute of Molecular & Cell Biology, Singapore, Singapore
| | - Jayantha Gunaratne
- Quantitative Proteomics Group, Institute of Molecular & Cell Biology, Singapore, Singapore
| | - Edward Manser
- sGSK Group, Institute of Molecular & Cell Biology, A*STAR, Singapore, Singapore.
- Department of Pharmacology, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Rogacka D, Piwkowska A. Beneficial effects of metformin on glomerular podocytes in diabetes. Biochem Pharmacol 2021; 192:114687. [PMID: 34274355 DOI: 10.1016/j.bcp.2021.114687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 01/15/2023]
Abstract
Podocytes and their foot processes form an important cellular layer of the glomerular barrier involved in regulating glomerular permeability. Disturbances in podocyte function play a central role in the development of proteinuria in diabetic nephropathy. The retraction of podocyte foot processes forming a slit diaphragm is a common feature of proteinuria. Metformin is an oral antidiabetic agent of the biguanide class that is widely recommended for the treatment of high blood glucose in patients with type 2 diabetes mellitus. In addition to lowering glucose, several recent studies have reported potential beneficial effects of metformin on diabetic kidney function. Furthermore, a key molecule of the antidiabetic mechanism of action of metformin is adenosine 5'-monophospate-activated protein kinase (AMPK), as the metformin-induced activation of AMPK is well documented. The present review summarizes current knowledge on the protective effects of metformin against pathological changes in podocytes that are induced by hyperglycemia.
Collapse
Affiliation(s)
- Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland.
| |
Collapse
|
11
|
Mysh M, Poulton JS. The Basolateral Polarity Module Promotes Slit Diaphragm Formation in Drosophila Nephrocytes, a Model of Vertebrate Podocytes. J Am Soc Nephrol 2021; 32:1409-1424. [PMID: 33795424 PMCID: PMC8259641 DOI: 10.1681/asn.2020071050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/12/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocyte slit diaphragms (SDs) are intercellular junctions that function as size-selective filters, excluding most proteins from urine. Abnormalities in SDs cause proteinuria and nephrotic syndrome. Podocytes exhibit apicobasal polarity, which can affect fundamental aspects of cell biology, including morphology, intercellular junction formation, and asymmetric protein distribution along the plasma membrane. Apical polarity protein mutations cause nephrotic syndrome, and data suggest apical polarity proteins regulate SD formation. However, there is no evidence that basolateral polarity proteins regulate SDs. Thus, the role of apicobasal polarity in podocytes remains unclear. METHODS Genetic manipulations and transgenic reporters determined the effects of disrupting apicobasal polarity proteins in Drosophila nephrocytes, which have SDs similar to those of mammalian podocytes. Confocal and electron microscopy were used to characterize SD integrity after loss of basolateral polarity proteins, and genetic-interaction studies illuminated relationships among apicobasal polarity proteins. RESULTS The study identified four novel regulators of nephrocyte SDs: Dlg, Lgl, Scrib, and Par-1. These proteins comprise the basolateral polarity module and its effector kinase. The data suggest these proteins work together, with apical polarity proteins, to regulate SDs by promoting normal endocytosis and trafficking of SD proteins. CONCLUSIONS Given the recognized importance of apical polarity proteins and SD protein trafficking in podocytopathies, the findings connecting basolateral polarity proteins to these processes significantly advance our understanding of SD regulation.
Collapse
Affiliation(s)
- Michael Mysh
- Department of Biology, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John S. Poulton
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
12
|
Heiden S, Siwek R, Lotz ML, Borkowsky S, Schröter R, Nedvetsky P, Rohlmann A, Missler M, Krahn MP. Apical-basal polarity regulators are essential for slit diaphragm assembly and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci 2021; 78:3657-3672. [PMID: 33651172 PMCID: PMC8038974 DOI: 10.1007/s00018-021-03769-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/11/2021] [Accepted: 01/16/2021] [Indexed: 12/02/2022]
Abstract
Apical-basal polarity is a key feature of most epithelial cells and it is regulated by highly conserved protein complexes. In mammalian podocytes, which emerge from columnar epithelial cells, this polarity is preserved and the tight junctions are converted to the slit diaphragms, establishing the filtration barrier. In Drosophila, nephrocytes show several structural and functional similarities with mammalian podocytes and proximal tubular cells. However, in contrast to podocytes, little is known about the role of apical-basal polarity regulators in these cells. In this study, we used expansion microscopy and found the apical polarity determinants of the PAR/aPKC and Crb-complexes to be predominantly targeted to the cell cortex in proximity to the nephrocyte diaphragm, whereas basolateral regulators also accumulate intracellularly. Knockdown of PAR-complex proteins results in severe endocytosis and nephrocyte diaphragm defects, which is due to impaired aPKC recruitment to the plasma membrane. Similar, downregulation of most basolateral polarity regulators disrupts Nephrin localization but had surprisingly divergent effects on endocytosis. Our findings suggest that morphology and slit diaphragm assembly/maintenance of nephrocytes is regulated by classical apical-basal polarity regulators, which have distinct functions in endocytosis.
Collapse
Affiliation(s)
- Stefanie Heiden
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Rebecca Siwek
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Marie-Luise Lotz
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Sarah Borkowsky
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Rita Schröter
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Pavel Nedvetsky
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany
| | - Astrid Rohlmann
- Institute of Anatomy and Molecular Neurobiology, University of Münster, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, University of Münster, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Michael P Krahn
- Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Albert-Schweitzer Campus 1-A14, 48149, Münster, Germany.
| |
Collapse
|
13
|
Egerman MA, Wong JS, Runxia T, Mosoyan G, Chauhan K, Reyes-Bahamonde J, Anandakrishnan N, Wong NJ, Bagiella E, Salem F, Meliambro K, Li H, Azeloglu EU, Coca SG, Campbell KN, Raij L. Plasminogenuria is associated with podocyte injury, edema, and kidney dysfunction in incident glomerular disease. FASEB J 2020; 34:16191-16204. [PMID: 33070369 PMCID: PMC7686123 DOI: 10.1096/fj.202000413r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 01/07/2023]
Abstract
Urinary plasminogen/plasmin, or plasmin (ogen) uria, has been demonstrated in proteinuric patients and exposure of cultured podocytes to plasminogen results in injury via oxidative stress pathways. A causative role for plasmin (ogen) as a "second hit" in kidney disease progression has yet to have been demonstrated in vivo. Additionally, association between plasmin (ogen) uria and kidney function in glomerular diseases remains unclear. We performed comparative studies in a puromycin aminonucleoside (PAN) nephropathy rat model treated with amiloride, an inhibitor of plasminogen activation, and measured changes in plasmin (ogen) uria. In a glomerular disease biorepository cohort (n = 128), we measured time-of-biopsy albuminuria, proteinuria, and plasmin (ogen) uria for correlations with kidney outcomes. In cultured human podocytes, plasminogen treatment was associated with decreased focal adhesion marker expression with rescue by amiloride. Increased glomerular plasmin (ogen) was found in PAN rats and focal segmental glomerulosclerosis (FSGS) patients. PAN nephropathy was associated with increases in plasmin (ogen) uria and proteinuria. Amiloride was protective against PAN-induced glomerular injury, reducing CD36 scavenger receptor expression and oxidative stress. In patients, we found associations between plasmin (ogen) uria and edema status as well as eGFR. Our study demonstrates a role for plasmin (ogen)-induced podocyte injury in the PAN nephropathy model, with amiloride having podocyte-protective properties. In one of the largest glomerular disease cohorts to study plasminogen, we validated previous findings while suggesting a potentially novel relationship between plasmin (ogen) uria and estimated glomerular filtration rate (eGFR). Together, these findings suggest a role for plasmin (ogen) in mediating glomerular injury and as a viable targetable biomarker for podocyte-sparing treatments.
Collapse
Affiliation(s)
- Marc A. Egerman
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Jenny S. Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Tian Runxia
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine
| | - Gohar Mosoyan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Kinsuk Chauhan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | | | | | - Nicholas J. Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Emilia Bagiella
- Center for Biostatistics, Department of Population health Science and Policy, Icahn School of Medicine at Mount Sinai
| | - Fadi Salem
- Department of Pathology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Kristin Meliambro
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Hong Li
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School
| | - Evren U. Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai
| | - Steven G. Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Kirk N. Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai
| | - Leopoldo Raij
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine
| |
Collapse
|
14
|
Wang Y, Eng DG, Kaverina NV, Loretz CJ, Koirala A, Akilesh S, Pippin JW, Shankland SJ. Global transcriptomic changes occur in aged mouse podocytes. Kidney Int 2020; 98:1160-1173. [PMID: 32592814 PMCID: PMC7606654 DOI: 10.1016/j.kint.2020.05.052] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/17/2020] [Accepted: 05/28/2020] [Indexed: 01/15/2023]
Abstract
Glomerular podocytes undergo structural and functional changes with advanced age, that increase susceptibility of aging kidneys to worse outcomes following superimposed glomerular diseases. To delineate transcriptional changes in podocytes in aged mice, RNA-seq was performed on isolated populations of reporter-labeled (tdTomato) podocytes from multiple young (two to three months) and advanced aged mice (22 to 24 months, equivalent to 70 plus year old humans). Of the 2,494 differentially expressed genes, 1,219 were higher and 1,275 were lower in aged podocytes. Pathway enrichment showed that major biological processes increased in aged podocytes included immune responses, non-coding RNA metabolism, gene silencing and MAP kinase signaling. Conversely, aged podocytes showed downregulation of developmental, morphogenesis and metabolic processes. Canonical podocyte marker gene expression decreased in aged podocytes, with increases in apoptotic and senescence genes providing a mechanism for the progressive loss of podocytes seen with aging. In addition, we revealed aberrations in the podocyte autocrine signaling network, identified the top transcription factors perturbed in aged podocytes, and uncovered candidate gene modulations that might promote healthy aging in podocytes. The transcriptional signature of aging is distinct from other kidney diseases. Thus, our study provides insights into biomarker discovery and molecular targeting of the aging process itself within podocytes.
Collapse
Affiliation(s)
- Yuliang Wang
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Natalya V Kaverina
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Carol J Loretz
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Abbal Koirala
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Shreeram Akilesh
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
15
|
Kawachi H, Fukusumi Y. New insight into podocyte slit diaphragm, a therapeutic target of proteinuria. Clin Exp Nephrol 2020; 24:193-204. [PMID: 32020343 PMCID: PMC7040068 DOI: 10.1007/s10157-020-01854-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/15/2020] [Indexed: 12/26/2022]
Abstract
Dysfunction of slit diaphragm, a cell–cell junction of glomerular podocytes, is involved in the development of proteinuria in several glomerular diseases. Slit diaphragm should be a target of a novel therapy for proteinuria. Nephrin, NEPH1, P-cadherin, FAT, and ephrin-B1 were reported to be extracellular components forming a molecular sieve of the slit diaphragm. Several cytoplasmic proteins such as ZO-1, podocin, CD2AP, MAGI proteins and Par-complex molecules were identified as scaffold proteins linking the slit diaphragm to the cytoskeleton. In this article, new insights into these molecules and the pathogenic roles of the dysfunction of these molecules were introduced. The slit diaphragm functions not only as a barrier but also as a signaling platform transfer the signal to the inside of the cell. For maintaining the slit diaphragm function properly, the phosphorylation level of nephrin is strictly regulated. The recent studies on the signaling pathway from nephrin, NEPH1, and ephrin-B1 were reviewed. Although the mechanism regulating the function of the slit diaphragm had remained unclear, recent studies revealed TRPC6 and angiotensin II-regulating mechanisms play a critical role in regulating the barrier function of the slit diaphragm. In this review, recent investigations on the regulation of the slit diaphragm function were reviewed, and a strategy for the establishment of a novel therapy for proteinuria was proposed.
Collapse
Affiliation(s)
- Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510, Japan.
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510, Japan
| |
Collapse
|
16
|
BENZING THOMAS. MOLECULAR DESIGN OF THE KIDNEY FILTRATION BARRIER. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2020; 131:125-139. [PMID: 32675853 PMCID: PMC7358502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Kidneys are the central regulators of organismal homeostasis. These organs filter enormous amounts of fluid from plasma; excrete toxic waste products; maintain salt, water, and volume balance; coordinate blood pressure regulation; and maintain the acid-base equilibrium essential for life. Although it has been known for decades that renal glomeruli serve as the site of plasma ultrafiltration and urine production, both the molecular design and function of the kidney filtration barrier have remained elusive. Indeed, the past two decades have witnessed enormous breakthroughs in our fundamental understanding of kidney filtration and the critical role that podocytes, specialized terminally differentiated epithelial cells at the glomerular capillaries, fulfill in the function of the kidney filtration barrier. Here we discuss recent advances in this field that will change the way we think about plasma ultrafiltration in health and proteinuria as a manifestation of glomerular diseases.
Collapse
Affiliation(s)
- THOMAS BENZING
- Correspondence and reprint requests: Thomas Benzing, MD, Department II of Internal Medicine, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany+ 49 221 4784480, 49 221 4785959
| |
Collapse
|
17
|
Takamura S, Fukusumi Y, Zhang Y, Narita I, Kawachi H. Partitioning-Defective-6-Ephrin-B1 Interaction Is Regulated by Nephrin-Mediated Signal and Is Crucial in Maintaining Slit Diaphragm of Podocyte. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:333-346. [PMID: 31837290 DOI: 10.1016/j.ajpath.2019.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/04/2019] [Accepted: 10/18/2019] [Indexed: 01/15/2023]
Abstract
Ephrin-B1 plays a critical role at slit diaphragm. Partitioning-defective (Par)-6 is down-regulated in podocyte of ephrin-B1 knockout mouse, suggesting that Par-6 is associated with ephrin-B1. Par polarity complex, consisting of Par-6, Par-3, and atypical protein kinase C, is essential for tight junction formation. In this study, the expression of Par-6 was analyzed in the normal and nephrotic syndrome model rats, and the molecular association of Par-6, Par-3, ephrin-B1, and nephrin was assessed with the human embryonic kidney 293 cell expression system. Par-6 was concentrated at slit diaphragm. Par 6 interacted with ephrin-B1 but not with nephrin, and Par-3 interacted with nephrin but not with ephrin-B1. The complexes of Par-6-ephrin-B1 and Par-3-nephrin were linked via extracellular sites of ephrin-B1 and nephrin. The Par-6-ephrin-B1 complex was delinked from the Par-3-nephrin complex, and Par-6 and ephrin-B1 were clearly down-regulated already at early phase of nephrotic model. The alteration of Par-6/ephrin-B1 advanced that of Par-3/nephrin. Stimulation to nephrin phosphorylated not only nephrin but also ephrin-B1, and consequently inhibited the interaction between ephrin-B1 and Par-6. Par-6 appeared at presumptive podocyte of early developmental stage and moved to basal area at capillary loop stage to participate in slit diaphragm formation at the final stage. Par-6-ephrin-B1 interaction is crucial for formation and maintenance of slit diaphragm of podocyte.
Collapse
Affiliation(s)
- Sayuri Takamura
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ying Zhang
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
18
|
Wang X, Adegoke EO, Ma M, Huang F, Zhang H, Adeniran SO, Zheng P, Zhang G. Influence of Wilms' tumor suppressor gene WT1 on bovine Sertoli cells polarity and tight junctions via non-canonical WNT signaling pathway. Theriogenology 2019; 138:84-93. [PMID: 31302435 DOI: 10.1016/j.theriogenology.2019.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Sertoli cells (SCs) are polarized epithelial cells and provide a microenvironment for the development of germ cells (GCs). The Wilms' tumor suppressor gene WT1 which support spermatogenesis is expressed explicitly in SCs. This study investigated the effect of WT1 on the polarity and blood-testis barrier (BTB) formation of bovine SCs in order to provide theoretical and practical bases for the spermatogenic process in mammals. In this study, newborn calf SCs were used as research material, and the RNAi technique was used to knockdown the endogenous WT1. The results show that WT1 knockdown did not affect the proliferation ability of SCs, but down-regulated the expression of polarity-associated proteins (Par3, Par6b, and E-cadherin), junction-associated protein (occludin) and inhibits transcription of downstream genes (WNT4, JNK, αPKC, and CDC42) in non-canonical WNT signaling pathway. WT1 also altered ZO-1 and occludin protein distribution. Overexpression of WNT1 did not affect the expression of Par6b, E-cadherin, and occludin, whereas the non-canonical WNT signaling pathway inhibitors wnt-c59, CCG-1423, and GO-6983 down-regulated the expression of Par6b, E-cadherin, and occludin respectively. This study indicates that WT1 mediates the regulation of several proteins involved in bovine SCs polarity maintenance and intercellular tight junctions (TJ) by non-canonical WNT signaling pathway.
Collapse
Affiliation(s)
- Xue Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - E O Adegoke
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Mingjun Ma
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Fushuo Huang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Han Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - S O Adeniran
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Peng Zheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Guixue Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China.
| |
Collapse
|
19
|
Dlugos CP, Picciotto C, Lepa C, Krakow M, Stöber A, Eddy ML, Weide T, Jeibmann A, P Krahn M, Van Marck V, Klingauf J, Ricker A, Wedlich-Söldner R, Pavenstädt H, Klämbt C, George B. Nephrin Signaling Results in Integrin β1 Activation. J Am Soc Nephrol 2019; 30:1006-1019. [PMID: 31097607 DOI: 10.1681/asn.2018040362] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with certain mutations in the gene encoding the slit diaphragm protein Nephrin fail to develop functional slit diaphragms and display severe proteinuria. Many adult-onset glomerulopathies also feature alterations in Nephrin expression and function. Nephrin signals from the podocyte slit diaphragm to the Actin cytoskeleton by recruiting proteins that can interact with C3G, a guanine nucleotide exchange factor of the small GTPase Rap1. Because Rap activity affects formation of focal adhesions, we hypothesized that Nephrin transmits signals to the Integrin receptor complex, which mediates podocyte adhesion to the extracellular matrix. METHODS To investigate Nephrin's role in transmitting signals to the Integrin receptor complex, we conducted genetic studies in Drosophila nephrocytes and validated findings from Drosophila in a cultured human podocyte model. RESULTS Drosophila nephrocytes form a slit diaphragm-like filtration barrier and express the Nephrin ortholog Sticks and stones (Sns). A genetic screen identified c3g as necessary for nephrocyte function. In vivo, nephrocyte-specific gene silencing of sns or c3g compromised nephrocyte filtration and caused nephrocyte diaphragm defects. Nephrocytes with impaired Sns or C3G expression displayed an altered localization of Integrin and the Integrin-associated protein Talin. Furthermore, gene silencing of c3g partly rescued nephrocyte diaphragm defects of an sns overexpression phenotype, pointing to genetic interaction of sns and c3g in nephrocytes. We also found that activated Nephrin recruited phosphorylated C3G and resulted in activation of Integrin β1 in cultured podocytes. CONCLUSIONS Our findings suggest that Nephrin can mediate a signaling pathway that results in activation of Integrin β1 at focal adhesions, which may affect podocyte attachment to the extracellular matrix.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christian Klämbt
- Neurobiology, Westfälische-Wilhelms University Münster, Münster, Germany
| | | |
Collapse
|
20
|
Loyer N, Januschke J. The last-born daughter cell contributes to division orientation of Drosophila larval neuroblasts. Nat Commun 2018; 9:3745. [PMID: 30218051 PMCID: PMC6138640 DOI: 10.1038/s41467-018-06276-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 08/17/2018] [Indexed: 01/27/2023] Open
Abstract
Controlling the orientation of cell division is important in the context of cell fate choices and tissue morphogenesis. However, the mechanisms providing the required positional information remain incompletely understood. Here we use stem cells of the Drosophila larval brain that stably maintain their axis of polarity and division between cell cycles to identify cues that orient cell division. Using live cell imaging of cultured brains, laser ablation and genetics, we reveal that division axis maintenance relies on their last-born daughter cell. We propose that, in addition to known intrinsic cues, stem cells in the developing fly brain are polarized by an extrinsic signal. We further find that division axis maintenance allows neuroblasts to maximize their contact area with glial cells known to provide protective and proliferative signals to neuroblasts.
Collapse
Affiliation(s)
- Nicolas Loyer
- Cell & Developmental Biology, School of Life Sciences, University of Dundee, MSI/WTB3 Complex, Dow Street, Dundee, DD1 5EH, UK
| | - Jens Januschke
- Cell & Developmental Biology, School of Life Sciences, University of Dundee, MSI/WTB3 Complex, Dow Street, Dundee, DD1 5EH, UK.
| |
Collapse
|
21
|
Sivakumar A, Kurpios NA. Transcriptional regulation of cell shape during organ morphogenesis. J Cell Biol 2018; 217:2987-3005. [PMID: 30061107 PMCID: PMC6122985 DOI: 10.1083/jcb.201612115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The emerging field of transcriptional regulation of cell shape changes aims to address the critical question of how gene expression programs produce a change in cell shape. Together with cell growth, division, and death, changes in cell shape are essential for organ morphogenesis. Whereas most studies of cell shape focus on posttranslational events involved in protein organization and distribution, cell shape changes can be genetically programmed. This review highlights the essential role of transcriptional regulation of cell shape during morphogenesis of the heart, lungs, gastrointestinal tract, and kidneys. We emphasize the evolutionary conservation of these processes across different model organisms and discuss perspectives on open questions and research avenues that may provide mechanistic insights toward understanding birth defects.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
22
|
Weng Z, Shang Y, Ji Z, Ye F, Lin L, Zhang R, Zhu J. Structural Basis of Highly Specific Interaction between Nephrin and MAGI1 in Slit Diaphragm Assembly and Signaling. J Am Soc Nephrol 2018; 29:2362-2371. [PMID: 30006415 PMCID: PMC6115659 DOI: 10.1681/asn.2017121275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 06/19/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The slit diaphragm is a specialized adhesion junction between opposing podocytes, establishing the final filtration barrier that prevents passage of proteins from the capillary lumen into the urinary space. Nephrin, the key structural and signaling adhesion molecule expressed in the slit diaphragm, contains an evolutionally conserved, atypical PDZ-binding motif (PBM) reported to bind to a variety of proteins in the slit diaphragm. Several mutations in NPHS1 (the gene encoding nephrin) that result in nephrin lacking an intact PBM are associated with glomerular diseases. However, the molecular basis of nephrin-PBM-mediated protein complexes is still unclear. METHODS Using a combination of biochemic, biophysic, and cell biologic approaches, we systematically investigated the interactions between nephrin-PBM and PDZ domain-containing proteins in the slit diaphragm. RESULTS We found that nephrin-PBM specifically binds to one member of the membrane-associated guanylate kinase family of scaffolding proteins, MAGI1, but not to another, MAGI2. The complex structure of MAGI1-PDZ3/nephrin-PBM reveals that the Gly at the -3 position of nephrin-PBM is the determining feature for MAGI1-PDZ3 recognition, which sharply contrasts with the typical PDZ/PBM binding mode. A single gain-of-function mutation within MAGI2 enabled nephrin-PBM binding. In addition, using our structural analysis, we developed a highly efficient inhibitory peptide capable of specifically blocking the nephrin/MAGI1 interaction. CONCLUSIONS MAGI1 interacts with nephrin-PBM with exquisite specificity. A newly developed, potent inhibitory peptide that blocks this interaction may be useful for future functional investigations in vivo. Our findings also provide possible explanations for the diseases caused by NPHS1 mutations.
Collapse
Affiliation(s)
- Zhuangfeng Weng
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China;,School of Life Science and Technology, ShanghaiTech University, Shanghai, China; and
| | - Yuan Shang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and
| | - Zeyang Ji
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and
| | - Fei Ye
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and,Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Lin Lin
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Rongguang Zhang
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China;,School of Life Science and Technology, ShanghaiTech University, Shanghai, China; and
| | - Jinwei Zhu
- State Key Laboratory of Molecular Biology, Shanghai Science Research Center, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Association of crumbs homolog-2 with mTORC1 in developing podocyte. PLoS One 2018; 13:e0202400. [PMID: 30125302 PMCID: PMC6101391 DOI: 10.1371/journal.pone.0202400] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
The evidence that gene mutations in the polarity determinant Crumbs homologs-2 (CRB2) cause congenital nephrotic syndrome suggests the functional importance of this gene product in podocyte development. Because another isoform, CRB3, was reported to repress the mechanistic/mammalian target of the rapamycin complex 1 (mTORC1) pathway, we examined the role of CRB2 function in developing podocytes in relation to mTORC1. In HEK-293 and MDCK cells constitutively expressing CRB2, we found that the protein localized to the apicolateral side of the cell plasma membrane and that this plasma membrane assembly required N-glycosylation. Confocal microscopy of the neonate mouse kidney revealed that both the tyrosine-phosphorylated form and non-phosphorylated form of CRB2 commence at the S-shaped body stage at the apicolateral side of podocyte precursor cells and move to foot processes in a capillary tuft pattern. The pattern of phosphorylated mTOR in developing podocytes was similar to that of CRB2 tyrosine phosphorylation. Additionally, the lack of a tyrosine phosphorylation site on CRB2 led to the reduced sensitivity of mTORC1 activation in response to energy starvation. CRB2 may play an important role in the mechanistic pathway of developing podocytes through tyrosine phosphorylation by associating with mTORC1 activation.
Collapse
|
24
|
Wang P, Zhao F, Nie X, Liu J, Yu Z. Knockdown of NUP160 inhibits cell proliferation, induces apoptosis, autophagy and cell migration, and alters the expression and localization of podocyte associated molecules in mouse podocytes. Gene 2018; 664:12-21. [PMID: 29704630 DOI: 10.1016/j.gene.2018.04.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 12/22/2022]
Abstract
Genetic mutations in dozens of monogenic genes can lead to serious podocyte dysfunction, which is a major cause of steroid-resistant nephrotic syndrome (SRNS). The NUP160 gene is expressed in both human kidney and mouse kidney. However, whether knockdown of NUP160 impairs podocytes has not yet been established. Therefore, we knocked down NUP160 by targeted short hairpin RNA (shRNA) in conditionally immortalized mouse podocytes and observed the effect of NUP160 knockdown on the proliferation, apoptosis, autophagy and cell migration of podocytes. We also investigated the effect of NUP160 knockdown on the expression and localization of podocyte associated molecules, such as nephrin, podocin, CD2AP and α-actinin-4. The knockdown of NUP160 significantly inhibited the proliferation of podocytes by decreasing the expression of both cyclin D1 and CDK4, increasing the expression of p27, and inducing S phase arrest. The knockdown of NUP160 promoted the apoptosis and autophagy of podocytes, and enhanced cell migration. The knockdown of NUP160 decreased the expression of nephrin, podocin and CD2AP in podocytes, and increased the expression of α-actinin-4. The knockdown of NUP160 also altered the subcellular localization of nephrin, podocin and CD2AP in podocytes. These results suggest that the knockdown of NUP160 impairs mouse podocytes, i.e. inhibiting cell proliferation, inducing apoptosis, autophagy and cell migration of mouse podocytes, and altering the expression and localization of podocyte associated molecules, including nephrin, podocin, CD2AP and α-actinin-4.
Collapse
Affiliation(s)
- Ping Wang
- Department of Pediatrics, Fuzhou Dongfang Hospital, Second Military Medical University, Fuzhou 350025, Fujian, China; The Military Hospital of 92435 Unit of the People's Liberation Army, Ningde 352103, Fujian, China
| | - Feng Zhao
- Department of Pediatrics, Dongfang Hospital, Xiamen University, Fuzhou 350025, Fujian, China
| | - Xiaojing Nie
- Department of Pediatrics, Dongfang Hospital, Xiamen University, Fuzhou 350025, Fujian, China
| | - Jiewei Liu
- Department of Pediatrics, Fuzhou Dongfang Hospital, Second Military Medical University, Fuzhou 350025, Fujian, China
| | - Zihua Yu
- Department of Pediatrics, Fuzhou Dongfang Hospital, Second Military Medical University, Fuzhou 350025, Fujian, China; Department of Pediatrics, Dongfang Hospital, Xiamen University, Fuzhou 350025, Fujian, China; Department of Pediatrics, Fuzhou Clinical Medical College, Fujian Medical University, Fuzhou 350025, Fujian, China.
| |
Collapse
|
25
|
Jing P, Liu Y, Keeler EG, Cruz NM, Freedman BS, Lin LY. Optical tweezers system for live stem cell organization at the single-cell level. BIOMEDICAL OPTICS EXPRESS 2018; 9:771-779. [PMID: 29552411 PMCID: PMC5854077 DOI: 10.1364/boe.9.000771] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 05/04/2023]
Abstract
Cell manipulation is one of the most impactful applications for optical tweezers, and derived from this promise, we demonstrate a new optical tweezers system for the study of cell adhesion and organization. This method utilizes photonic-crystal-enhanced optical tweezers to manipulate cells with low laser intensities. By doing so, it enables effective cell patterning and culturing within the conditions necessary for successful differentiation and colony formation of human pluripotent stem cells. To this end, the biocompatibility of plasma-treated parylene-C for cell culturing was studied, and a thorough characterization of cellular interactive forces was performed using this system. Furthermore, this study also demonstrates construction of patterned cell arrays at arbitrary positions with micrometer-scale precision.
Collapse
Affiliation(s)
- Peifeng Jing
- Department of Electrical Engineering, University of Washington, 185 Stevens Way, Seattle, WA 98195, USA
| | - Yannan Liu
- Department of Electrical Engineering, University of Washington, 185 Stevens Way, Seattle, WA 98195, USA
| | - Ethan G Keeler
- Department of Electrical Engineering, University of Washington, 185 Stevens Way, Seattle, WA 98195, USA
| | - Nelly M Cruz
- Division of Nephrology, Kidney Research Institute, and Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, 850 Republican St., Seattle, WA 98109, USA
| | - Benjamin S Freedman
- Division of Nephrology, Kidney Research Institute, and Institute for Stem Cell and Regenerative Medicine, Department of Medicine, University of Washington School of Medicine, 850 Republican St., Seattle, WA 98109, USA
| | - Lih Y Lin
- Department of Electrical Engineering, University of Washington, 185 Stevens Way, Seattle, WA 98195, USA
| |
Collapse
|
26
|
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: 96] [Impact Index Per Article: 13.7] [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
|
27
|
Hochapfel F, Denk L, Mendl G, Schulze U, Maaßen C, Zaytseva Y, Pavenstädt H, Weide T, Rachel R, Witzgall R, Krahn MP. Distinct functions of Crumbs regulating slit diaphragms and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci 2017; 74:4573-4586. [PMID: 28717874 PMCID: PMC11107785 DOI: 10.1007/s00018-017-2593-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 06/26/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
Abstract
Mammalian podocytes, the key determinants of the kidney's filtration barrier, differentiate from columnar epithelial cells and several key determinants of apical-basal polarity in the conventional epithelia have been shown to regulate podocyte morphogenesis and function. However, little is known about the role of Crumbs, a conserved polarity regulator in many epithelia, for slit-diaphragm formation and podocyte function. In this study, we used Drosophila nephrocytes as model system for mammalian podocytes and identified a conserved function of Crumbs proteins for cellular morphogenesis, nephrocyte diaphragm assembly/maintenance, and endocytosis. Nephrocyte-specific knock-down of Crumbs results in disturbed nephrocyte diaphragm assembly/maintenance and decreased endocytosis, which can be rescued by Drosophila Crumbs as well as human Crumbs2 and Crumbs3, which were both expressed in human podocytes. In contrast to the extracellular domain, which facilitates nephrocyte diaphragm assembly/maintenance, the intracellular FERM-interaction motif of Crumbs is essential for regulating endocytosis. Moreover, Moesin, which binds to the FERM-binding domain of Crumbs, is essential for efficient endocytosis. Thus, we describe here a new mechanism of nephrocyte development and function, which is likely to be conserved in mammalian podocytes.
Collapse
Affiliation(s)
- Florian Hochapfel
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
- Medizinische Klinik und Poliklinik D, Universitätsklinikum Münster, Domagkstr. 3a, 48149, Münster, Germany
| | - Lucia Denk
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Gudrun Mendl
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Ulf Schulze
- Medizinische Klinik und Poliklinik D, Universitätsklinikum Münster, Domagkstr. 3a, 48149, Münster, Germany
| | - Christine Maaßen
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Yulia Zaytseva
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Hermann Pavenstädt
- Medizinische Klinik und Poliklinik D, Universitätsklinikum Münster, Domagkstr. 3a, 48149, Münster, Germany
| | - Thomas Weide
- Medizinische Klinik und Poliklinik D, Universitätsklinikum Münster, Domagkstr. 3a, 48149, Münster, Germany
| | - Reinhard Rachel
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Ralph Witzgall
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Michael P Krahn
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany.
- Medizinische Klinik und Poliklinik D, Universitätsklinikum Münster, Domagkstr. 3a, 48149, Münster, Germany.
| |
Collapse
|
28
|
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: 97] [Impact Index Per Article: 12.1] [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
|
29
|
Targeting Neph1 and ZO-1 protein-protein interaction in podocytes prevents podocyte injury and preserves glomerular filtration function. Sci Rep 2017; 7:12047. [PMID: 28935902 PMCID: PMC5608913 DOI: 10.1038/s41598-017-12134-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/05/2017] [Indexed: 11/08/2022] Open
Abstract
Targeting protein-protein interaction (PPI) is rapidly becoming an attractive alternative for drug development. While drug development commonly involves inhibiting a PPI, in this study, we show that stabilizing PPI may also be therapeutically beneficial. Junctional proteins Neph1 and ZO-1 and their interaction is an important determinant of the structural integrity of slit diaphragm, which is a critical component of kidney's filtration system. Since injury induces loss of this interaction, we hypothesized that strengthening this interaction may protect kidney's filtration barrier and preserve kidney function. In this study, Neph1-ZO-1 structural complex was screened for the presence of small druggable pockets formed from contributions from both proteins. One such pocket was identified and screened using a small molecule library. Isodesmosine (ISD) a rare naturally occurring amino acid and a biomarker for pulmonary arterial hypertension was selected as the best candidate and to establish the proof of concept, its ability to enhance Neph1-CD and ZO-1 binding was tested. Results from biochemical binding analysis showed that ISD enhanced Neph1 and ZO-1 interaction under in vitro and in vivo conditions. Importantly, ISD treated podocytes were resistant to injury-induced loss of transepithelial permeability. Finally, mouse and zebrafish studies show that ISD protects from injury-induced renal damage.
Collapse
|
30
|
Tsuji K, Suleiman H, Miner JH, Daley JM, Capen DE, Păunescu TG, Lu HAJ. Ultrastructural Characterization of the Glomerulopathy in Alport Mice by Helium Ion Scanning Microscopy (HIM). Sci Rep 2017; 7:11696. [PMID: 28916834 PMCID: PMC5601433 DOI: 10.1038/s41598-017-12064-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/01/2017] [Indexed: 01/19/2023] Open
Abstract
The glomerulus exercises its filtration barrier function by establishing a complex filtration apparatus consisting of podocyte foot processes, glomerular basement membrane and endothelial cells. Disruption of any component of the glomerular filtration barrier leads to glomerular dysfunction, frequently manifested as proteinuria. Ultrastructural studies of the glomerulus by transmission electron microscopy (TEM) and conventional scanning electron microscopy (SEM) have been routinely used to identify and classify various glomerular diseases. Here we report the application of newly developed helium ion scanning microscopy (HIM) to examine the glomerulopathy in a Col4a3 mutant/Alport syndrome mouse model. Our study revealed unprecedented details of glomerular abnormalities in Col4a3 mutants including distorted podocyte cell bodies and disorganized primary processes. Strikingly, we observed abundant filamentous microprojections arising from podocyte cell bodies and processes, and presence of unique bridging processes that connect the primary processes and foot processes in Alport mice. Furthermore, we detected an altered glomerular endothelium with disrupted sub-endothelial integrity. More importantly, we were able to clearly visualize the complex, three-dimensional podocyte and endothelial interface by HIM. Our study demonstrates that HIM provides nanometer resolution to uncover and rediscover critical ultrastructural characteristics of the glomerulopathy in Col4a3 mutant mice.
Collapse
Affiliation(s)
- Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Hani Suleiman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - James M Daley
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Abstract
Podocytes exhibit a unique cytoskeletal architecture that is fundamentally linked to their function in maintaining the kidney filtration barrier. The cytoskeleton regulates podocyte shape, structure, stability, slit diaphragm insertion, adhesion, plasticity, and dynamic response to environmental stimuli. Genetic mutations demonstrate that even slight impairment of the podocyte cytoskeletal apparatus results in proteinuria and glomerular disease. Moreover, mechanisms underpinning all acquired glomerular pathologies converge on disruption of the cytoskeleton, suggesting that this subcellular structure could be targeted for therapeutic purposes. This review summarizes our current understanding of the function of the cytoskeleton in podocytes and the associated implications for pathophysiology.
Collapse
Affiliation(s)
- Christoph Schell
- Institute of Surgical Pathology and.,Department of Medicine IV, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias B Huber
- Department of Medicine IV, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; .,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and.,BIOSS Centre for Biological Signalling Studies and Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
32
|
Ganner A, Neumann-Haefelin E. Genetic kidney diseases: Caenorhabditis elegans as model system. Cell Tissue Res 2017; 369:105-118. [PMID: 28484847 DOI: 10.1007/s00441-017-2622-z] [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: 12/15/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022]
Abstract
Despite its apparent simplicity, the nematode Caenorhabditis elegans has a high rating as a model in molecular and developmental biology and biomedical research. C. elegans has no excretory system comparable with the mammalian kidney but many of the genes and molecular pathways involved in human kidney diseases are conserved in C. elegans. The plethora of genetic, molecular and imaging tools available in C. elegans has enabled major discoveries in renal research and advanced our understanding of the pathogenesis of genetic kidney diseases. In particular, studies in C. elegans have pioneered the fundamental role of cilia for cystic kidney diseases. In addition, proteins of the glomerular filtration barrier and podocytes are critical for cell recognition, assembly of functional neuronal circuits, mechanosensation and signal transduction in C. elegans. C. elegans has also proved tremendously valuable for aging research and the Von Hippel-Lindau tumor suppressor gene has been shown to modulate lifespan in the nematode. Further, studies of the excretory canal, membrane transport and ion channel function in C. elegans have provided insights into mechanisms of tubulogenesis and cellular homeostasis. This review recounts the way that C. elegans can be used to investigate various aspects of genetic and molecular nephrology. This model system opens up an exciting and new area of study of renal development and diseases.
Collapse
Affiliation(s)
- Athina Ganner
- Department of Nephrology, Medical Center, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Elke Neumann-Haefelin
- Department of Nephrology, Medical Center, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany.
| |
Collapse
|
33
|
Swiatecka-Urban A. Endocytic Trafficking at the Mature Podocyte Slit Diaphragm. Front Pediatr 2017; 5:32. [PMID: 28286744 PMCID: PMC5324021 DOI: 10.3389/fped.2017.00032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/03/2017] [Indexed: 12/16/2022] Open
Abstract
Endocytic trafficking couples cell signaling with the cytoskeletal dynamics by organizing a crosstalk between protein networks in different subcellular compartments. Proteins residing in the plasma membrane are internalized and transported as cargo in endocytic vesicles (i.e., endocytosis). Subsequently, cargo proteins can be delivered to lysosomes for degradation or recycled back to the plasma membrane. The slit diaphragm is a modified tight junction connecting foot processes of the glomerular epithelial cells, podocytes. Signaling at the slit diaphragm plays a critical role in the kidney while its dysfunction leads to glomerular protein loss (proteinuria), manifesting as nephrotic syndrome, a rare condition with an estimated incidence of 2-4 new cases per 100,000 each year. Relatively little is known about the role of endocytic trafficking in podocyte signaling and maintenance of the slit diaphragm integrity. This review will focus on the role of endocytic trafficking at the mature podocyte slit diaphragm.
Collapse
Affiliation(s)
- Agnieszka Swiatecka-Urban
- Department of Nephrology, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Akchurin O, Du Z, Ramkellawan N, Dalal V, Han SH, Pullman J, Müsch A, Susztak K, Reidy KJ. Partitioning-Defective 1a/b Depletion Impairs Glomerular and Proximal Tubule Development. J Am Soc Nephrol 2016; 27:3725-3737. [PMID: 27185860 DOI: 10.1681/asn.2014111124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The kidney is a highly polarized epithelial organ that develops from undifferentiated mesenchyme, although the mechanisms that regulate the development of renal epithelial polarity are incompletely understood. Partitioning-defective 1 (Par1) proteins have been implicated in cell polarity and epithelial morphogenesis; however, the role of these proteins in the developing kidney has not been established. Therefore, we studied the contribution of Par1a/b to renal epithelial development. We examined the renal phenotype of newborn compound mutant mice carrying only one allele of Par1a or Par1b. Loss of three out of four Par1a/b alleles resulted in severe renal hypoplasia, associated with impaired ureteric bud branching. Compared with kidneys of newborn control littermates, kidneys of newborn mutant mice exhibited dilated proximal tubules and immature glomeruli, and the renal proximal tubular epithelia lacked proper localization of adhesion complexes. Furthermore, Par1a/b mutants expressed low levels of renal Notch ligand Jag1, activated Notch2, and Notch effecter Hes1. Together, these data demonstrate that Par1a/b has a key role in glomerular and proximal tubule development, likely via modulation of Notch signaling.
Collapse
Affiliation(s)
- Oleh Akchurin
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Zhongfang Du
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Nadira Ramkellawan
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Vidhi Dalal
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James Pullman
- Department of Pathology, Montefiore Medical Center, Bronx, New York; and
| | - Anne Müsch
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kimberly J Reidy
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York; .,Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
35
|
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: 1.0] [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
|
36
|
Yaoita E, Nishimura H, Nameta M, Yoshida Y, Takimoto H, Fujinaka H, Kawachi H, Magdeldin S, Zhang Y, Xu B, Oyama T, Nakamura F, Yamamoto T. Avian Podocytes, Which Lack Nephrin, Use Adherens Junction Proteins at Intercellular Junctions. J Histochem Cytochem 2015; 64:67-76. [PMID: 26416242 DOI: 10.1369/0022155415611708] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/19/2015] [Indexed: 12/14/2022] Open
Abstract
Nephrin, a major intercellular junction (ICJ) molecule of mammalian podocytes in the renal glomerulus, is absent in the avian genome. We hypothesized that birds use ICJ molecules other than nephrin in their podocytes. Therefore, in the present study, we examined the possible involvement of adherens junction (AJ) proteins in the ICJs of avian podocytes. We found the AJ proteins N-cadherin and α- and β-catenins in podocytes of quail and chickens but not in those of rats, pigs or humans. The AJ proteins were prominent in avian glomerulus-rich fractions in immunoblot analyses, and in immunofluorescence microscopy analyses, they were localized along glomerular capillary walls appearing in at least two staining patterns: weakly diffuse and distinctly granular. Immunoelectron microscopy demonstrated that the significant accumulation of immunogold particles for the AJ proteins were especially evident in avian slit diaphragms and AJs. Furthermore, N-cadherin was found to be expressed in all nephron cells in the early developmental stage but became confined to podocytes during maturation. These results indicate that avian slit diaphragms clearly express AJ proteins as compared with that in the mammal-where AJ proteins are suppressed to an extremely low level-and that avian podocytes are interconnected by AJs per se in addition to slit diaphragms.
Collapse
Affiliation(s)
- Eishin Yaoita
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hiroko Nishimura
- Department of Health Informatics, Niigata University of Health and Welfare, Niigata, Japan (HN)
| | - Masaaki Nameta
- Cooperative Laboratory of Electron Microscopy, Niigata University, Niigata, Japan (MN)
| | - Yutaka Yoshida
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hiroki Takimoto
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hidehiko Fujinaka
- Institute for Clinical Research, Niigata National Hospital, Niigata, Japan (HF)
| | - Hiroshi Kawachi
- Department of Cell Biology (HK), Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sameh Magdeldin
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Ying Zhang
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Bo Xu
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Tomizo Oyama
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Fujio Nakamura
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Tadashi Yamamoto
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| |
Collapse
|
37
|
Abstract
The function of the kidney, filtering blood and concentrating metabolic waste into urine, takes place in an intricate and functionally elegant structure called the renal glomerulus. Normal glomerular function retains circulating cells and valuable macromolecular components of plasma in blood, resulting in urine with just trace amounts of proteins. Endothelial cells of glomerular capillaries, the podocytes wrapped around them, and the fused extracellular matrix these cells form altogether comprise the glomerular filtration barrier, a dynamic and highly selective filter that sieves on the basis of molecular size and electrical charge. Current understanding of the structural organization and the cellular and molecular basis of renal filtration draws from studies of human glomerular diseases and animal models of glomerular dysfunction.
Collapse
Affiliation(s)
- Rizaldy P Scott
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Susan E Quaggin
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
38
|
Genetic causes of proteinuria and nephrotic syndrome: impact on podocyte pathobiology. Pediatr Nephrol 2015; 30:221-33. [PMID: 24584664 PMCID: PMC4262721 DOI: 10.1007/s00467-014-2753-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/31/2013] [Accepted: 01/02/2014] [Indexed: 12/11/2022]
Abstract
In the past 20 years, multiple genetic mutations have been identified in patients with congenital nephrotic syndrome (CNS) and both familial and sporadic focal segmental glomerulosclerosis (FSGS). Characterization of the genetic basis of CNS and FSGS has led to the recognition of the importance of podocyte injury to the development of glomerulosclerosis. Genetic mutations induce injury due to effects on the podocyte's structure, actin cytoskeleton, calcium signaling, and lysosomal and mitochondrial function. Transgenic animal studies have contributed to our understanding of podocyte pathobiology. Podocyte endoplasmic reticulum stress response, cell polarity, and autophagy play a role in maintenance of podocyte health. Further investigations related to the effects of genetic mutations on podocytes may identify new pathways for targeting therapeutics for nephrotic syndrome.
Collapse
|
39
|
Ebarasi L, Ashraf S, Bierzynska A, Gee H, McCarthy H, Lovric S, Sadowski C, Pabst W, Vega-Warner V, Fang H, Koziell A, Simpson M, Dursun I, Serdaroglu E, Levy S, Saleem M, Hildebrandt F, Majumdar A. Defects of CRB2 cause steroid-resistant nephrotic syndrome. Am J Hum Genet 2015; 96:153-61. [PMID: 25557779 DOI: 10.1016/j.ajhg.2014.11.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/21/2014] [Indexed: 01/15/2023] Open
Abstract
Nephrotic syndrome (NS), the association of gross proteinuria, hypoalbuminaemia, edema, and hyperlipidemia, can be clinically divided into steroid-sensitive (SSNS) and steroid-resistant (SRNS) forms. SRNS regularly progresses to end-stage renal failure. By homozygosity mapping and whole exome sequencing, we here identify recessive mutations in Crumbs homolog 2 (CRB2) in four different families affected by SRNS. Previously, we established a requirement for zebrafish crb2b, a conserved regulator of epithelial polarity, in podocyte morphogenesis. By characterization of a loss-of-function mutation in zebrafish crb2b, we now show that zebrafish crb2b is required for podocyte foot process arborization, slit diaphragm formation, and proper nephrin trafficking. Furthermore, by complementation experiments in zebrafish, we demonstrate that CRB2 mutations result in loss of function and therefore constitute causative mutations leading to NS in humans. These results implicate defects in podocyte apico-basal polarity in the pathogenesis of NS.
Collapse
|
40
|
Identification of novel Kirrel3 gene splice variants in adult human skeletal muscle. BMC PHYSIOLOGY 2014; 14:11. [PMID: 25488023 PMCID: PMC4269076 DOI: 10.1186/s12899-014-0011-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 11/19/2014] [Indexed: 01/08/2023]
Abstract
Background Multiple cell types including trophoblasts, osteoclasts and myoblasts require somatic cell fusion events as part of their physiological functions. In Drosophila Melanogaster the paralogus type 1 transmembrane receptors and members of the immunoglobulin superfamily Kin of Irre (Kirre) and roughest (Rst) regulate myoblast fusion during embryonic development. Present within the human genome are three homologs to Kirre termed Kin of Irre like (Kirrel) 1, 2 and 3. Currently it is unknown if Kirrel3 is expressed in adult human skeletal muscle. Results We investigated (using PCR and Western blot) Kirrel3 in adult human skeletal muscle samples taken at rest and after mild exercise induced muscle damage. Kirrel3 mRNA expression was verified by sequencing and protein presence via blotting with 2 different anti-Kirrel3 protein antibodies. Evidence for three alternatively spliced Kirrel3 mRNA transcripts in adult human skeletal muscle was obtained. Kirrel3 mRNA in adult human skeletal muscle was detected at low or moderate levels, or not at all. This sporadic expression suggests that Kirrel3 is expressed in a pulsatile manner. Several anti Kirrel3 immunoreactive proteins were detected in all adult human skeletal muscle samples analysed and results suggest the presence of different isoforms or posttranslational modification, or both. Conclusion The results presented here demonstrate for the first time that there are at least 3 splice variants of Kirrel3 expressed in adult human skeletal muscle, two of which have never previously been identified in human muscle. Importantly, mRNA of all splice variants was not always present, a finding with potential physiological relevance. These initial discoveries highlight the need for more molecular and functional studies to understand the role of Kirrel3 in human skeletal muscle.
Collapse
|
41
|
Hulkko J, Patrakka J, Lal M, Tryggvason K, Hultenby K, Wernerson A. Neph1 is reduced in primary focal segmental glomerulosclerosis, minimal change nephrotic syndrome, and corresponding experimental animal models of adriamycin-induced nephropathy and puromycin aminonucleoside nephrosis. NEPHRON EXTRA 2014; 4:146-54. [PMID: 25404935 PMCID: PMC4202611 DOI: 10.1159/000365091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND/AIMS The transmembrane proteins Neph1 and nephrin form a complex in the slit diaphragm (SD) of podocytes. As recent studies indicate an involvement of this complex in the polymerization of the actin cytoskeleton and proteinuria, we wanted to study the subcellular localization of Neph1 in the normal human kidney and its expression in focal segmental glomerulosclerosis (FSGS), minimal change nephrotic syndrome (MCNS), and the corresponding experimental models of Adriamycin-induced nephropathy (ADR) and puromycin aminonucleoside nephrosis (PAN). All these disorders are characterized by substantial foot process effacement (FPE) and proteinuria. MATERIALS AND METHODS Kidney biopsies from patients with primary FSGS (perihilar type) and MCNS were compared to normal renal tissue. Mouse and rat kidney cortices from days 7 and 14 after Adriamycin injection and days 2 and 4 after puromycin aminonucleoside injection, respectively, were compared to control mouse and rat kidney. Polyclonal antibodies against Neph1 and nephrin were used for immunoelectron microscopy, and semiquantification was performed. RESULTS We localized Neph1 mainly to, and in close proximity to, the SD. Double staining of Neph1 and nephrin showed the proteins to be in close connection in the SD. The total amount of Neph1 in the podocytes was significantly reduced in FSGS, MCNS, ADR, and PAN. The reduction of Neph1 was also seen in areas with and without FPE. Nephrin was reduced in MCNS and PAN but unchanged in FSGS. CONCLUSION With nephrin (but not Neph1) unchanged in FSGS, there might be a disruption of the complex and an involvement of Neph1 in its pathogenesis.
Collapse
Affiliation(s)
- Jenny Hulkko
- Department of Clinical Science, Intervention and Technology, Stockholm, Sweden
| | - Jaakko Patrakka
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mark Lal
- Department of Medical Biochemistry and Biophysics, Stockholm, Sweden
| | - Karl Tryggvason
- Department of Medical Biochemistry and Biophysics, Stockholm, Sweden
| | - Kjell Hultenby
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika Wernerson
- Department of Clinical Science, Intervention and Technology, Stockholm, Sweden
| |
Collapse
|
42
|
Rocque BL, Babayeva S, Li J, Leung V, Nezvitsky L, Cybulsky AV, Gros P, Torban E. Deficiency of the planar cell polarity protein Vangl2 in podocytes affects glomerular morphogenesis and increases susceptibility to injury. J Am Soc Nephrol 2014; 26:576-86. [PMID: 25145929 DOI: 10.1681/asn.2014040340] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The planar cell polarity (PCP) signaling pathway is crucial for tissue morphogenesis. Van Gogh-like protein 2 (Vangl2) is central in the PCP pathway; in mice, Vangl2 loss is embryonically lethal because of neural tube defects, and mutations in Vangl2 are associated with human neural tube defects. In the kidney, PCP signaling may be important for tubular morphogenesis and organization of glomerular epithelial cells (podocytes) along the glomerular basement membrane. Podocyte cell protrusions (foot processes) are critical for glomerular permselectivity; loss of foot process architecture results in proteinuria and FSGS. Previously, we showed a profound effect of PCP signaling on podocyte shape, actin rearrangement, cell motility, and nephrin endocytosis. To test our hypothesis that the PCP pathway is involved in glomerular development and function and circumvent lethality of the ubiquitous Vangl2 mutation in the Looptail mouse, we generated a mouse model with a podocyte-specific ablation of the Vangl2 gene. We report here that podocyte-specific deletion of Vangl2 leads to glomerular maturation defects in fetal kidneys. In adult mice, we detected significantly smaller glomeruli, but it did not affect glomerular permselectivity in aging animals. However, in the context of glomerular injury induced by injection of antiglomerular basement membrane antibody, deletion of Vangl2 resulted in exacerbation of injury and accelerated progression to chronic segmental and global glomerular sclerosis. Our results indicate that Vangl2 function in podocytes is important for glomerular development and protects against glomerular injury in adult animals.
Collapse
Affiliation(s)
- Brittany L Rocque
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| | - Sima Babayeva
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| | - Jane Li
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| | - Vicki Leung
- Departments of Human Genetics and Complex Traits Group, McGill University, Montreal, Quebec, Canada
| | - Lisa Nezvitsky
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| | - Andrey V Cybulsky
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| | - Philippe Gros
- Complex Traits Group, McGill University, Montreal, Quebec, Canada Biochemistry and
| | - Elena Torban
- Departments of Medicine and Physiology, McGill University and McGill University Health Centre, Montreal, Quebec, Canada; and
| |
Collapse
|
43
|
Glomerular development--shaping the multi-cellular filtration unit. Semin Cell Dev Biol 2014; 36:39-49. [PMID: 25153928 DOI: 10.1016/j.semcdb.2014.07.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 01/09/2023]
Abstract
The glomerulus represents a highly structured filtration unit, composed of glomerular endothelial cells, mesangial cells, podocytes and parietal epithelial cells. During glomerulogenesis an intricate network of signaling pathways involving transcription factors, secreted factors and cell-cell communication is required to guarantee accurate evolvement of a functional, complex 3-dimensional glomerular architecture. Here, we want to provide an overview on the critical steps and relevant signaling cascades of glomerular development.
Collapse
|
44
|
Costa MSA, Machado MCR, Vieceli FM, Amistá L, Baroneza JE, Yan CYI, Ramos RGP. The Rst-Neph family of cell adhesion molecules in Gallus gallus. J Neurogenet 2014; 28:270-81. [PMID: 24914768 DOI: 10.3109/01677063.2014.933220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Rst-Neph family comprises an evolutionarily conserved group of single-pass transmembrane glycoproteins that belong to the immunoglobulin superfamily and participate in a wide range of cell adhesion and recognition events in both vertebrates and invertebrates. In mammals and fish, three Rst-Neph members, named Neph1-3, are present. Besides being widely expressed in the embryo, particularly in the developing nervous system, they also contribute to the formation and integrity of the urine filtration apparatus in the slit diaphragm of kidney glomerular podocytes, where they form homodimers, as well as heterodimers with Nephrin, another immunoglobulin-like cell adhesion molecule. In mice, absence of Neph1 causes severe proteinuria, podocyte effacement and perinatal death, while in humans, a mutated form of Nephrin leads to congenital nephrotic syndrome of the Finnish type. Intriguingly, neither Nephrin nor Neph3 are present in birds, which nevertheless have typical vertebrate kidneys with mammalian-like slit diaphragms. These characteristics make, in principle, avian systems very helpful for understanding the evolution and functional significance of the complex interactions displayed by Rst-Neph proteins. To this end we have started a systematic study of chicken Neph embryonic and post-embryonic expression, both at mRNA and protein level. RT-qPCR mRNA quantification of the two Neph paralogues in adult tissues showed that both are expressed in heart, brain, and retina. Neph1 is additionally present in kidney, liver, pancreas, lungs, and testicles, while Neph2 mRNA is barely detected in kidney, testicles, pancreas and absent in liver and lungs. In embryos, mRNA from both genes can already be detected at as early as stage HH14, and remain expressed until at least HH28. Finally, we used a specific antibody to examine the spatial dynamics and subcellular distribution of ggNeph2 between stages HH20-28, particularly in the mesonephros, dermomyotomes, developing heart, and retina.
Collapse
Affiliation(s)
- Mara Silvia A Costa
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | | | | | | | | | | | | |
Collapse
|
45
|
Satoh D, Hirose T, Harita Y, Daimon C, Harada T, Kurihara H, Yamashita A, Ohno S. aPKCλ maintains the integrity of the glomerular slit diaphragm through trafficking of nephrin to the cell surface. J Biochem 2014; 156:115-28. [PMID: 24700503 PMCID: PMC4112437 DOI: 10.1093/jb/mvu022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The slit diaphragm (SD), the specialized intercellular junction between renal glomerular epithelial cells (podocytes), provides a selective-filtration barrier in renal glomeruli. Dysfunction of the SD results in glomerular diseases that are characterized by disappearance of SD components, such as nephrin, from the cell surface. Although the importance of endocytosis and degradation of SD components for the maintenance of SD integrity has been suggested, the dynamic nature of the turnover of intact cell-surface SD components remained unclear. Using isolated rat glomeruli we show that the turnover rates of cell-surface SD components are relatively high; they almost completely disappear from the cell surface within minutes. The exocytosis, but not endocytosis, of heterologously expressed nephrin requires the kinase activity of the cell polarity regulator atypical protein kinase C (aPKC). Consistently, we demonstrate that podocyte-specific deletion of aPKCλ resulted in a decrease of cell-surface localization of SD components, causing massive proteinuria. In conclusion, the regulation of SD turnover by aPKC is crucial for the maintenance of SD integrity and defects in aPKC signalling can lead to proteinuria. These findings not only reveal the pivotal importance of the dynamic turnover of cell-surface SD components but also suggest a novel pathophysiological basis in glomerular disease.
Collapse
Affiliation(s)
- Daisuke Satoh
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Tomonori Hirose
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Yutaka Harita
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, JapanDepartment of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Chikara Daimon
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Tomonori Harada
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hidetake Kurihara
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, JapanDepartment of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo; Department of Pediatrics, Yokohama City University, Yokohama; Department of Anatomy, Juntendo University, School of Medicine, Bunkyo, Tokyo; and Advanced Medical Research Center, Yokohama City University, Yokohama, Kanagawa, Japan
| |
Collapse
|
46
|
Rinschen MM, Wu X, König T, Pisitkun T, Hagmann H, Pahmeyer C, Lamkemeyer T, Kohli P, Schnell N, Schermer B, Dryer S, Brooks BR, Beltrao P, Krueger M, Brinkkoetter PT, Benzing T. Phosphoproteomic analysis reveals regulatory mechanisms at the kidney filtration barrier. J Am Soc Nephrol 2014; 25:1509-22. [PMID: 24511133 DOI: 10.1681/asn.2013070760] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Diseases of the kidney filtration barrier are a leading cause of ESRD. Most disorders affect the podocytes, polarized cells with a limited capacity for self-renewal that require tightly controlled signaling to maintain their integrity, viability, and function. Here, we provide an atlas of in vivo phosphorylated, glomerulus-expressed proteins, including podocyte-specific gene products, identified in an unbiased tandem mass spectrometry-based approach. We discovered 2449 phosphorylated proteins corresponding to 4079 identified high-confidence phosphorylated residues and performed a systematic bioinformatics analysis of this dataset. We discovered 146 phosphorylation sites on proteins abundantly expressed in podocytes. The prohibitin homology domain of the slit diaphragm protein podocin contained one such site, threonine 234 (T234), located within a phosphorylation motif that is mutated in human genetic forms of proteinuria. The T234 site resides at the interface of podocin dimers. Free energy calculation through molecular dynamic simulations revealed a role for T234 in regulating podocin dimerization. We show that phosphorylation critically regulates formation of high molecular weight complexes and that this may represent a general principle for the assembly of proteins containing prohibitin homology domains.
Collapse
Affiliation(s)
- Markus M Rinschen
- Department of Internal Medicine II, Center for Molecular Medicine, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Systems Biology of Ageing Cologne
| | - Xiongwu Wu
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Tim König
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Trairak Pisitkun
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Henning Hagmann
- Department of Internal Medicine II, Center for Molecular Medicine
| | | | - Tobias Lamkemeyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases
| | - Priyanka Kohli
- Department of Internal Medicine II, Center for Molecular Medicine, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases
| | - Nicole Schnell
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases
| | - Bernhard Schermer
- Department of Internal Medicine II, Center for Molecular Medicine, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Systems Biology of Ageing Cologne
| | - Stuart Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases
| | - Bernard R Brooks
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Pedro Beltrao
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, Cambridge, United Kingdom; and
| | - Marcus Krueger
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Thomas Benzing
- Department of Internal Medicine II, Center for Molecular Medicine, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Systems Biology of Ageing Cologne,
| |
Collapse
|
47
|
Teng B, Duong M, Tossidou I, Yu X, Schiffer M. Role of protein kinase C in podocytes and development of glomerular damage in diabetic nephropathy. Front Endocrinol (Lausanne) 2014; 5:179. [PMID: 25414693 PMCID: PMC4220730 DOI: 10.3389/fendo.2014.00179] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/06/2014] [Indexed: 12/13/2022] Open
Abstract
The early glomerular changes in diabetes include a podocyte phenotype with loss of slit diaphragm proteins, changes in the actin cytoskeleton and foot process architecture. This review focuses on the role of the protein kinase C (PKC) family in podocytes and points out the differential roles of classical, novel, and atypical PKCs in podocytes. Some PKC isoforms are indispensable for proper glomerular development and slit diaphragm maintenance, whereas others might be harmful when activated in the diabetic milieu. Therefore, some might be interesting treatment targets in the early phase of diabetes.
Collapse
Affiliation(s)
- Beina Teng
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
| | - Michelle Duong
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
| | - Irini Tossidou
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
| | - Xuejiao Yu
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
| | - Mario Schiffer
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- *Correspondence: Mario Schiffer, Department of Medicine/Nephrology, Hannover Medical School, Carl Neuberg Street 1-OE6840, Hannover 30625, Germany e-mail:
| |
Collapse
|
48
|
Worthmann K, Leitges M, Teng B, Sestu M, Tossidou I, Samson T, Haller H, Huber TB, Schiffer M. Def-6, a novel regulator of small GTPases in podocytes, acts downstream of atypical protein kinase C (aPKC) λ/ι. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1945-1959. [PMID: 24096077 PMCID: PMC5707189 DOI: 10.1016/j.ajpath.2013.08.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 08/21/2013] [Accepted: 08/26/2013] [Indexed: 11/23/2022]
Abstract
The atypical protein kinase C (aPKC) isotypes PKCλ/ι and PKCζ are both expressed in podocytes; however, little is known about differences in their function. Previous studies in mice have demonstrated that podocyte-specific loss of PKCλ/ι leads to a severe glomerular phenotype, whereas mice deficient in PKCζ develop no renal phenotype. We analyzed various effects caused by PKCλ/ι and PKCζ deficiency in cultured murine podocytes. In contrast to PKCζ-deficient podocytes, PKCλ/ι-deficient podocytes exhibited a severe actin cytoskeletal phenotype, reduced cell size, decreased number of focal adhesions, and increased activation of small GTPases. Comparative microarray analysis revealed that the guanine nucleotide exchange factor Def-6 was specifically up-regulated in PKCλ/ι-deficient podocytes. In vivo Def-6 expression is significantly increased in podocytes of PKCλ/ι-deficient mice. Cultured PKCλ/ι-deficient podocytes exhibited an enhanced membrane association of Def-6, indicating enhanced activation. Overexpression of aPKCλ/ι in PKCλ/ι-deficient podocytes could reduce the membrane-associated expression of Def-6 and rescue the actin phenotype. In the present study, PKCλ/ι was identified as an important factor for actin cytoskeletal regulation in podocytes and Def-6 as a specific downstream target of PKCλ/ι that regulates the activity of small GTPases and subsequently the actin cytoskeleton of podocytes.
Collapse
Affiliation(s)
- Kirstin Worthmann
- Division of Nephrology, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Michael Leitges
- Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | - Beina Teng
- Division of Nephrology, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Marcello Sestu
- Faculty of Medicine, Interdisciplinary Centre for Clinical Research (IZKF) Leipzig, University of Leipzig, Leipzig, Germany
| | - Irini Tossidou
- Division of Nephrology, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Samson
- Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Hermann Haller
- Division of Nephrology, Department of Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias B Huber
- Renal Division, University Hospital Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University, Freiburg, Germany
| | - Mario Schiffer
- Division of Nephrology, Department of Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
49
|
Grahammer F, Schell C, Huber TB. Molecular understanding of the slit diaphragm. Pediatr Nephrol 2013; 28:1957-62. [PMID: 23233041 DOI: 10.1007/s00467-012-2375-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 10/16/2012] [Accepted: 10/17/2012] [Indexed: 11/30/2022]
Abstract
Glomerular filtration has always attracted the interest of nephrologists and renal researchers alike. Although several key questions on the structure and function of the kidney filter may have been answered within the last 40 years of intense research, there still remain crucial questions to be solved. The following article attempts to give a brief overview of recent developments in glomerular research highlighting particular advances in our understanding of the slit diaphragm.
Collapse
Affiliation(s)
- Florian Grahammer
- Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany
| | | | | |
Collapse
|
50
|
Grahammer F, Schell C, Huber TB. The podocyte slit diaphragm--from a thin grey line to a complex signalling hub. Nat Rev Nephrol 2013; 9:587-98. [PMID: 23999399 DOI: 10.1038/nrneph.2013.169] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The architectural design of our kidneys is amazingly complex, and culminates in the 3D structure of the glomerular filter. During filtration, plasma passes through a sieve consisting of a fenestrated endothelium and a broad basement membrane before it reaches the most unique part, the slit diaphragm, a specialized type of intercellular junction that connects neighbouring podocyte foot processes. When podocytes become stressed, irrespective of the causative stimulus, they undergo foot process effacement and loss of slit diaphragms--two key steps leading to proteinuria. Thus, proteinuria is the unifying denominator of a broad spectrum of podocytopathies. With the rising prevalence of chronic kidney disease and the fact that glomerular diseases account for the majority of patients with end-stage renal disease, further investigation and elucidation of this unique structure is of paramount importance. This Review recounts how perception of the slit diaphragm has changed over time as a result of intense research, from its first anatomical description as a thin intercellular connection, to an appreciation of its role as a dynamic signalling hub. These observations led to the introduction of novel concepts in podocyte biology, which could pave the way to development of highly desired, specific therapeutic strategies for glomerular diseases.
Collapse
Affiliation(s)
- Florian Grahammer
- Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany
| | | | | |
Collapse
|