1
|
Guarin G, Netzel A, Flores KM, Cumpelik A, Bose R. Sacituzumab-govitecan-induced severe acute tubulointerstitial nephritis requiring hemodialysis. BMC Nephrol 2024; 25:402. [PMID: 39522022 PMCID: PMC11550548 DOI: 10.1186/s12882-024-03828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Sacituzumab govitecan is an antibody-drug conjugate that is FDA approved for refractory metastatic triple-negative breast cancer. It targets the human trophoblastic cell-surface antigen 2 (Trop-2) with SN-38, a topoisomerase I inhibitor, attached to the antibody [1]. SN-38 breaks DNA strands and induces tumor apoptosis [2]. Acute kidney injury (AKI) is one of its adverse effects mainly prerenal due to gastrointestinal toxicity, but it has not been reported to cause acute tubulointerstitial nephritis (ATIN). CASE PRESENTATION This report describes a rare adverse effect of sacituzumab govitecan, the approach to diagnosing the etiology of the patient's AKI, and the mechanism by which sacituzumab govitecan causes ATIN. A woman with metastatic ER positive, PR positive, HER2 negative breast cancer who was initiated on sacituzumab govitecan presents with vomiting and diarrhea, and findings of nephrotic-range proteinuria, negative anti-PLA2R antibody, and severe AKI requiring hemodialysis. She underwent kidney biopsy and pathology showed ATIN characterized by patchy interstitial inflammation alongside tubular injury without glomerular and vascular involvement. With intermittent renal replacement therapy, furosemide challenge, and a course of prednisone, the patient's kidney function recovered. CONCLUSIONS Sacituzumab has a high affinity for Trop-2 protein which is also expressed within the collecting ducts, and to a lesser extent, the proximal tubule. Individuals, such as this patient, who express a homozygous genotype for UGT1A1*28 allele are at increased risk for AKI from sacituzumab govitecan due to decreased glucuronidation of SN-38.
Collapse
Affiliation(s)
- Geneva Guarin
- Department of Medicine, Division of Hospital Medicine, Washington University in St. Louis, Barnes-Jewish Hospital Plaza, St. Louis, MO, 63110, USA.
| | - Audrey Netzel
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Karen Marie Flores
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Arun Cumpelik
- Department of Medicine, Division of Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Ron Bose
- Department of Medicine, Division of Oncology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
2
|
Zhang J, Zhao H, Li X, Qian R, Gao P, Lu S, Ma Z. Efficacy of low-dose rituximab in minimal change disease and prevention of relapse. BMC Nephrol 2023; 24:112. [PMID: 37101300 PMCID: PMC10134665 DOI: 10.1186/s12882-023-03092-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 02/20/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Minimal change disease (MCD) is a major cause of nephrotic syndrome (NS) in children and a minority of adults. The higher tendency to relapse put patients at risk for prolonged exposure to steroids and other immunosuppressive agents. B cell depletion with rituximab (RTX) may be beneficial to the treatment and prevention of frequently relapsing MCD. Therefore, this study aimed to verify the therapeutic/preventive effects of low-dose RTX on the relapse in adult with MCD. METHODS A total of 33 adult patients were selected for the study, including 22 patients with relapsing MCD in relapse treatment group who were treated with low-dose RTX (200 mg per week × 4 following by 200 mg every 6 months) and 11 patients in relapse prevention group with complete remission (CR) after steroid therapy were treated with RTX (200 mg ×1 every 6 months) for preventing the relapse of MCD. RESULTS Of the 22 patients with MCD in relapse treatment group, there were 21 cases (95.45%) of remission [2 (9.09%) partial remission (PR), 19 (86.36%) CR], 1 (4.56%) no remission (NR) and 20 (90.90%) relapse-free. The Median duration of sustained remission was 16.3 months (3, 23.5 months, inter quartile range (IQR)). 11 patients in the relapse prevention group during a follow-up of 12 months (9-31 months) had no relapse. The average dose of prednisone in two groups after RTX treatment was significantly lower than before treatment. CONCLUSION The results of this study suggested low-dose RTX can significantly reduce relapse rate and steroid dose in adults with MCD with fewer side effects. Low-dose RTX regimens may be beneficial for the treatment of relapsing MCD in adults and may be the preferred regimen for patients at high risk for the development of adverse events from corticosteroids.
Collapse
Affiliation(s)
- Jian Zhang
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Hui Zhao
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Xiaoli Li
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Rui Qian
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Peijuan Gao
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Shouyan Lu
- Division of nephrology, Gansu Provincial Hospital, Lanzhou, 730001, China
| | - Zhigang Ma
- Department of nephrology, The Second Affiliated Hospital, School of Medcine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong, 518172, China.
| |
Collapse
|
3
|
Aydin Z, Yilmaz M, Sipahioglu M, Dervisoglu E, Aydemir N, Uzun S, Istemihan Z, Unsal O, Tatar E, Berktas HB, Ozdemir A, Sumnu A, Kumru G, Cetinkaya H, Kazan S, Kocyigit I, Gokalp C, Hasbal B, Artan AS, Ozelsancak R, Taymez D, Yadigar S, Alagoz S, Aslan BB, Yaylaci S, Jabrayilov J, Turgutalp K, Dursun B, Sahin G. Demographic, clinical and laboratory characteristics of adult-onset minimal change disease in Turkey: Turkish Society of Nephrology-Glomerular Diseases (TSN-GOLD) Working Group. Int Urol Nephrol 2023; 55:975-982. [PMID: 36178610 DOI: 10.1007/s11255-022-03382-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/24/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE In our study, diagnostic and demographic characteristics of patients diagnosed with minimal change disease (MCD) by biopsy, clinical and laboratory findings in our country were investigated. METHODS Data were obtained from the Turkish Society of Nephrology Glomerular Diseases (TSN-GOLD) Working Group database. Demographic characteristics, indications for biopsy, diagnosis of the glomerular diseases, comorbidities, laboratory and biopsy findings of all patients were recorded. The data presented are cross-sectional and includes application data for the biopsy period. RESULTS Of 3875 patients, 233 patients with MCD (median age 35.0 years) were included in the study, which constitutes 6.0% of the total glomerulonephritis database. Renal biopsy was performed in 196 (84.1%) patients due to nephrotic syndrome. Median serum creatinine was 0.7 (0.6-1.0) mg/dl, mean eGFR was 104 ± 33 ml/min/1.73 m2 and median proteinuria 6000 mg/day. The number of patients under the age of 40 years was 139 (59.7%) (Group A), and the number of patients aged 40 years and over was 94 (40.3%) (Group B). Compared to Group A, global sclerotic glomeruli (24 vs. 43, p < 0.001) interstitial inflammation (15 vs. 34, p < 0.001), interstitial fibrosis (20 vs. 31, p = 0.001, vascular changes (10 vs. 25, p < 0.001) and tubular atrophy (18 vs. 30, p < 0.001) were found to be significantly higher in Group B. There was no difference in immunofluorescent staining properties between the two groups. CONCLUSION Our data are generally compatible with the literature. Chronic histopathological changes were more common in patients aged 40 years and older than younger patients. Studies investigating the effects of these different features on renal survival are needed.
Collapse
Affiliation(s)
- Zeki Aydin
- Department of Nephrology, Darica Farabi Training and Research Hospital, University of Health Sciences, Fevziçakmak, Dr. Zeki Acar Ave. No: 62, 4170, Darica, Kocaeli, Turkey.
| | - Murvet Yilmaz
- Department of Nephrology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Murat Sipahioglu
- Division of Nephrology, Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey
| | - Erkan Dervisoglu
- Division of Nephrology, Department of Internal Medicine, Kocaeli University School of Medicine, Kocaeli, Turkey
| | - Nihal Aydemir
- Department of Nephrology, Ankara Numune Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Sami Uzun
- Department of Nephrology, Haseki Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Zulal Istemihan
- Division of Nephrology, Department of Internal Medicine, Istanbul University School of Medicine, Istanbul, Turkey
| | - Oktay Unsal
- Division of Nephrology, Department of Internal Medicine, Uludag University School of Medicine, Bursa, Turkey
| | - Erhan Tatar
- Department of Nephrology, Bozyaka Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Haci Bayram Berktas
- Division of Nephrology, Department of Internal Medicine, Inonu University School of Medicine, Malatya, Turkey
| | - Arzu Ozdemir
- Department of Nephrology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Abdullah Sumnu
- Department of Nephrology, Medipol University School of Medicine, Istanbul, Turkey
| | - Gizem Kumru
- Division of Nephrology, Department of Internal Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Hakki Cetinkaya
- Department of Nephrology, Sultan Abdülhamid Han Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Sinan Kazan
- Department of Nephrology, Afyonkarahisar School of Medicine, University of Health Sciences, Afyonkarahisar, Turkey
| | - Ismail Kocyigit
- Division of Nephrology, Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey
| | - Cenk Gokalp
- Division of Nephrology, Department of Internal Medicine, Trakya University School of Medicine, Edirne, Turkey
| | - Baris Hasbal
- Department of Nephrology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ayse Serra Artan
- Division of Nephrology, Department of Internal Medicine, Bezm-I Alem Vakif University School of Medicine, Istanbul, Turkey
| | - Ruya Ozelsancak
- Division of Nephrology, Department of Internal Medicine, Cukurova University School of Medicine, Adana, Turkey
| | - Dilek Taymez
- Department of Nephrology, Kocaeli State Hospital, Kocaeli, Turkey
| | - Serap Yadigar
- Department of Nephrology, Dr. Lutfi Kırdar Kartal Training and Research Hospital, Istanbul, Turkey
| | - Selma Alagoz
- Division of Nephrology, Department of Internal Medicine, Istanbul Cerrahpasa University School of Medicine, Istanbul, Turkey
| | - Bilal Burcak Aslan
- Department of Nephrology, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, Bursa, Turkey
| | - Selcuk Yaylaci
- Division of Nephrology, Department of Internal Medicine, Sakarya University School of Medicine, Sakarya, Turkey
| | - Jabrayil Jabrayilov
- Division of Nephrology, Department of Internal Medicine, Hacettepe University School of Medicine, Ankara, Turkey
| | - Kenan Turgutalp
- Division of Nephrology, Department of Internal Medicine, Mersin University School of Medicine, Mersin, Turkey
| | - Belda Dursun
- Division of Nephrology, Department of Internal Medicine, Pamukkale University School of Medicine, Denizli, Turkey
| | - Garip Sahin
- Division of Nephrology, Department of Internal Medicine, Osmangazi University School of Medicine, Eskisehir, Turkey
| |
Collapse
|
4
|
Manera M, Casciano F, Giari L. Ultrastructural Alterations of the Glomerular Filtration Barrier in Fish Experimentally Exposed to Perfluorooctanoic Acid. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:5253. [PMID: 37047869 PMCID: PMC10094651 DOI: 10.3390/ijerph20075253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 06/19/2023]
Abstract
Per- and polyfluoroalkyl substances can be referred to as the most critical group of contaminants of emerging concern. They can accumulate in high concentration in the kidney and are known to potentially affect its function. Nonetheless, there is a lack of knowledge about their morphopathological effect on the glomerular filtration barrier. Since previous research suggests perfluorooctanoic acid (PFOA) induces glomerular protein leakage, the glomerular filtration barrier of 30 carp from the same parental stock (10 unexposed; 10 exposed to 200 ng L-1 of PFOA; and 10 exposed to 2 mg L-1 of PFOA for 56 days) was screened for possible PFOA-induced ultrastructural lesions in order to shed light on the related pathophysiology. PFOA exposure affected the glomerular filtration barrier in carp experimentally exposed to 2 mg L-1, showing ultrastructural alterations compatible with glomerulonephrosis: podocyte effacement, reduction of filtration slits and filtration slit diaphragms, basement membrane disarrangement, and occurrence of proteinaceous material in the urinary space. The results of the present research confirm the glomerular origin of the PFOA-induced protein leakage and can contribute to the mechanistic comprehension of PFOA's impact on renal function and to the assessment of the exposure effect of environmental pollutants on animals and humans, according to the One Health approach.
Collapse
Affiliation(s)
- Maurizio Manera
- Department of Biosciences, Food and Environmental Technologies, University of Teramo, St. R. Balzarini 1, 64100 Teramo, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, St. Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Luisa Giari
- Department of Environmental and Prevention Sciences, University of Ferrara, St. Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
5
|
Circulating Permeability Factors in Focal Segmental Glomerulosclerosis: In V itro Detection. Kidney Int Rep 2022; 7:2691-2703. [PMID: 36506233 PMCID: PMC9727530 DOI: 10.1016/j.ekir.2022.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction The recurrence of proteinuria after kidney transplantation in patients with focal segmental glomerulosclerosis (FSGS) is considered proof of the presence of circulating permeability factors (CPFs). The aim of this study is to demonstrate the presence of plasma CPFs using series of in vitro assays. Methods Podocytes and endothelial cells (glomerular microvascular endothelial cells [GMVECs]) were incubated with plasma from FSGS patients with presumed CPFs in relapse and remission and from steroid-resistant nephrotic syndrome (SRNS), steroid-sensitive nephrotic syndrome (SSNS), membranous nephropathy (MN), and healthy controls (hCtrls). Cell viability, podocyte actin cytoskeleton architecture, and reactive oxygen species (ROS) formation with or without ROS scavenger were investigated by Cell Counting Kit-8 assay, immunofluorescence staining, and CM-H2DCFDA probing, respectively. Results Presumed CPF-containing plasma causes a series of events in podocytes but not in GMVECs. These events include actin cytoskeleton rearrangement and excessive formation of ROS, which results in podocyte loss. These effects were solely observed in response to CPF plasma collected during relapse, but not in response to plasma of hCtrls, or patients with SRNS, SSNS, and MN. The copresence of dimethylthiourea, a scavenger of ROS, abolished the aforementioned effects of CPF plasma. Conclusion We provide a panel of in vitro bioassays to measure podocyte injury and predict the presence of CPFs in plasma of patients with nephrotic syndrome (NS), providing a new framework for monitoring CPF activity that may contribute to future NS diagnostics or used for disease monitoring purposes. Moreover, our findings suggest that the inhibition of ROS formation or facilitating rapid ROS scavenging may exert beneficial effects in patients with CPFs.
Collapse
|
6
|
Ye Q, Lan B, Liu H, Persson PB, Lai EY, Mao J. A critical role of the podocyte cytoskeleton in the pathogenesis of glomerular proteinuria and autoimmune podocytopathies. Acta Physiol (Oxf) 2022; 235:e13850. [PMID: 35716094 DOI: 10.1111/apha.13850] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/23/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023]
Abstract
Selective glomerular filtration relies on the membrane separating the glomerular arterioles from the Bowman space. As a major component of the glomerular filtration barrier, podocytes form foot processes by the actin cytoskeleton, which dynamically adjusts in response to environmental changes to maintain filtration barrier integrity. The slit diaphragms bridge the filtration slits between neighboring foot processes and act as signaling hubs interacting with the actin cytoskeleton. Focal adhesions relay signals to regulate actin dynamics while allowing podocyte adherence to the basement membrane. Mutations in actin regulatory and signaling proteins may disrupt the actin cytoskeleton, resulting in foot process retraction, effacement, and proteinuria. Large-scale gene expression profiling platforms, transgenic animal models, and other in vivo gene delivery methods now enhance our understanding of the interactions among podocyte focal adhesions, slit diaphragms, and actin dynamics. In addition, our team found that at least 66% of idiopathic nephrotic syndrome (INS) children have podocyte autoantibodies, which was defined as a new disease subgroup-, autoimmune podocytopathies. This review outlines the pathophysiological mechanisms of podocyte cytoskeleton protein interactions in proteinuria and glomerular podocytopathy.
Collapse
Affiliation(s)
- Qing Ye
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Bing Lan
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Huihui Liu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Translational Physiology, Berlin, Germany
| | - En Yin Lai
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Translational Physiology, Berlin, Germany.,Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| |
Collapse
|
7
|
Shankland SJ, Wang Y, Shaw AS, Vaughan JC, Pippin JW, Wessely O. Podocyte Aging: Why and How Getting Old Matters. J Am Soc Nephrol 2021; 32:2697-2713. [PMID: 34716239 PMCID: PMC8806106 DOI: 10.1681/asn.2021050614] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/26/2021] [Indexed: 02/04/2023] Open
Abstract
The effects of healthy aging on the kidney, and how these effects intersect with superimposed diseases, are highly relevant in the context of the population's increasing longevity. Age-associated changes to podocytes, which are terminally differentiated glomerular epithelial cells, adversely affect kidney health. This review discusses the molecular and cellular mechanisms underlying podocyte aging, how these mechanisms might be augmented by disease in the aged kidney, and approaches to mitigate progressive damage to podocytes. Furthermore, we address how biologic pathways such as those associated with cellular growth confound aging in humans and rodents.
Collapse
Affiliation(s)
- Stuart J. Shankland
- Division of Nephrology, University of Washington, Seattle, Washington
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington
| | - Yuliang Wang
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington
| | - Andrey S. Shaw
- Department of Research Biology, Genentech, South San Francisco, California
| | - Joshua C. Vaughan
- Department of Chemistry, University of Washington, Seattle, Washington
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Jeffrey W. Pippin
- Division of Nephrology, University of Washington, Seattle, Washington
| | - Oliver Wessely
- Lerner Research Institute, Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
8
|
Djenoune L, Tomar R, Dorison A, Ghobrial I, Schenk H, Hegermann J, Beverly-Staggs L, Hidalgo-Gonzalez A, Little MH, Drummond IA. Autonomous Calcium Signaling in Human and Zebrafish Podocytes Controls Kidney Filtration Barrier Morphogenesis. J Am Soc Nephrol 2021; 32:1697-1712. [PMID: 33911000 PMCID: PMC8425667 DOI: 10.1681/asn.2020101525] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/12/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocytes are critical to maintaining the glomerular filtration barrier, and mutations in nephrotic syndrome genes are known to affect podocyte calcium signaling. However, the role of calcium signaling during podocyte development remains unknown. METHODS We undertook live imaging of calcium signaling in developing podocytes, using zebrafish larvae and human kidney organoids. To evaluate calcium signaling during development and in response to channel blockers and genetic defects, the calcium biosensor GCaMP6s was expressed in zebrafish podocytes. We used electron microscopy to evaluate filtration barrier formation in zebrafish, and Fluo-4 to detect calcium signals in differentiating podocytes in human kidney organoids. RESULTS Immature zebrafish podocytes (2.5 days postfertilization) generated calcium transients that correlated with interactions with forming glomerular capillaries. Calcium transients persisted until 4 days postfertilization, and were absent after glomerular barrier formation was complete. We detected similar calcium transients in maturing human organoid glomeruli, suggesting a conserved mechanism. In both models, inhibitors of SERCA or IP3 receptor calcium-release channels blocked calcium transients in podocytes, whereas lanthanum was ineffective, indicating the calcium source is from intracellular podocyte endoplasmic-reticulum stores. Calcium transients were not affected by blocking heartbeat or by blocking development of endothelium or endoderm, and they persisted in isolated glomeruli, suggesting podocyte-autonomous calcium release. Inhibition of expression of phospholipase C-γ1, but not nephrin or phospholipase C-ε1, led to significantly decreased calcium activity. Finally, blocking calcium release affected glomerular shape and podocyte foot process formation, supporting the critical role of calcium signaling in glomerular morphogenesis. CONCLUSIONS These findings establish podocyte cell-autonomous calcium signaling as a prominent and evolutionarily conserved feature of podocyte differentiation and demonstrate its requirement for podocyte foot process formation.
Collapse
Affiliation(s)
- Lydia Djenoune
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Ritu Tomar
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Aude Dorison
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Irene Ghobrial
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany,Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Lynne Beverly-Staggs
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| | | | - Melissa H. Little
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Iain A. Drummond
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| |
Collapse
|
9
|
Ning L, Suleiman HY, Miner JH. Synaptopodin deficiency exacerbates kidney disease in a mouse model of Alport syndrome. Am J Physiol Renal Physiol 2021; 321:F12-F25. [PMID: 34029143 DOI: 10.1152/ajprenal.00035.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Synaptopodin (Synpo) is an actin-associated protein in podocyte foot processes. By generating mice that completely lack Synpo, we previously showed that Synpo is dispensable for normal kidney function. However, lack of Synpo worsened adriamycin-induced nephropathy, indicating a protective role for Synpo in injured podocytes. Here, we investigated whether lack of Synpo directly impacts a genetic disease, Alport syndrome (AS), because Synpo is reduced in podocytes of affected humans and mice; whether this is merely an association or pathogenic is unknown. We used collagen type IV-α5 (Col4a5) mutant mice, which model X-linked AS, showing glomerular basement membrane (GBM) abnormalities, eventual foot process effacement, and progression to end-stage kidney disease. We intercrossed mice carrying mutations in Synpo and Col4a5 to produce double-mutant mice. Urine and tissue were taken at select time points to evaluate albuminuria, histopathology, and glomerular capillary wall composition and ultrastructure. Lack of Synpo in Col4a5-/Y, Col4a5-/-, or Col4a5+/- Alport mice led to the acceleration of disease progression, including more severe proteinuria and glomerulosclerosis. Absence of Synpo attenuated the shift of myosin IIA from the podocyte cell body and major processes to actin cables near the GBM in the areas of effacement. We speculate that this is mechanistically associated with enhanced loss of podocytes due to easier detachment from the GBM. We conclude that Synpo deletion exacerbates the disease phenotype in Alport mice, revealing the podocyte actin cytoskeleton as a target for therapy in patients with AS.NEW & NOTEWORTHY Alport syndrome (AS) is a hereditary disease of the glomerular basement with hematuria and proteinuria. Podocytes eventually exhibit foot process effacement, indicating actin cytoskeletal changes. To investigate how cytoskeletal changes impact podocytes, we generated Alport mice lacking synaptopodin, an actin-binding protein in foot processes. Analysis showed a more rapid disease progression, demonstrating that synaptopodin is protective. This suggests that the actin cytoskeleton is a target for therapy in AS and perhaps other glomerular diseases.
Collapse
Affiliation(s)
- Liang Ning
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hani Y Suleiman
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
10
|
Rogg M, Maier JI, Dotzauer R, Artelt N, Kretz O, Helmstädter M, Abed A, Sammarco A, Sigle A, Sellung D, Dinse P, Reiche K, Yasuda-Yamahara M, Biniossek ML, Walz G, Werner M, Endlich N, Schilling O, Huber TB, Schell C. SRGAP1 Controls Small Rho GTPases To Regulate Podocyte Foot Process Maintenance. J Am Soc Nephrol 2021; 32:563-579. [PMID: 33514561 PMCID: PMC7920176 DOI: 10.1681/asn.2020081126] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/15/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Previous research demonstrated that small Rho GTPases, modulators of the actin cytoskeleton, are drivers of podocyte foot-process effacement in glomerular diseases, such as FSGS. However, a comprehensive understanding of the regulatory networks of small Rho GTPases in podocytes is lacking. METHODS We conducted an analysis of podocyte transcriptome and proteome datasets for Rho GTPases; mapped in vivo, podocyte-specific Rho GTPase affinity networks; and examined conditional knockout mice and murine disease models targeting Srgap1. To evaluate podocyte foot-process morphology, we used super-resolution microscopy and electron microscopy; in situ proximity ligation assays were used to determine the subcellular localization of the small GTPase-activating protein SRGAP1. We performed functional analysis of CRISPR/Cas9-generated SRGAP1 knockout podocytes in two-dimensional and three-dimensional cultures and quantitative interaction proteomics. RESULTS We demonstrated SRGAP1 localization to podocyte foot processes in vivo and to cellular protrusions in vitro. Srgap1fl/fl*Six2Cre but not Srgap1fl/fl*hNPHS2Cre knockout mice developed an FSGS-like phenotype at adulthood. Podocyte-specific deletion of Srgap1 by hNPHS2Cre resulted in increased susceptibility to doxorubicin-induced nephropathy. Detailed analysis demonstrated significant effacement of podocyte foot processes. Furthermore, SRGAP1-knockout podocytes showed excessive protrusion formation and disinhibition of the small Rho GTPase machinery in vitro. Evaluation of a SRGAP1-dependent interactome revealed the involvement of SRGAP1 with protrusive and contractile actin networks. Analysis of glomerular biopsy specimens translated these findings toward human disease by displaying a pronounced redistribution of SRGAP1 in FSGS. CONCLUSIONS SRGAP1, a podocyte-specific RhoGAP, controls podocyte foot-process architecture by limiting the activity of protrusive, branched actin networks. Therefore, elucidating the complex regulatory small Rho GTPase affinity network points to novel targets for potentially precise intervention in glomerular diseases.
Collapse
Affiliation(s)
- Manuel Rogg
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany,Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Jasmin I. Maier
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Robert Dotzauer
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Nadine Artelt
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Ahmed Abed
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Alena Sammarco
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - August Sigle
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany,Department of Urology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Dominik Sellung
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany,Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Patrick Dinse
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Karoline Reiche
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Mako Yasuda-Yamahara
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany,Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Martin L. Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schell
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany,Berta-Ottenstein Program, Medical Faculty, Medical Center – University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
da Silva CA, Monteiro MLGDR, Araújo LS, Urzedo MG, Rocha LB, dos Reis MA, Machado JR. In situ evaluation of podocytes in patients with focal segmental glomerulosclerosis and minimal change disease. PLoS One 2020; 15:e0241745. [PMID: 33147279 PMCID: PMC7641434 DOI: 10.1371/journal.pone.0241745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022] Open
Abstract
Podocyte injury in focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD) results from the imbalance between adaptive responses that maintain homeostasis and cellular dysfunction that can culminate in cell death. Therefore, an in situ analysis was performed to detect morphological changes related to cell death and autophagy in renal biopsies from adult patients with podocytopathies. Forty-nine renal biopsies from patients with FSGS (n = 22) and MCD (n = 27) were selected. In situ expression of Wilms Tumor 1 protein (WT1), light chain microtubule 1-associated protein (LC3) and caspase-3 protein were evaluated by immunohistochemistry. The foot process effacement and morphological alterations related to podocyte cell death and autophagy were analyzed with transmission electronic microscopy. Reduction in the density of WT1-labeled podocytes was observed for FSGS and MCD cases as compared to controls. Foot process width (FPW) in control group was lower than in cases of podocytopathies. In FSGS group, FPW was significantly higher than in MCD group and correlated with proteinuria. A density of LC3-labeled podocytes and the number of autophagosomes in podocytes/ pedicels were higher in the MCD group than in the FSGS group. The number of autophagosomes correlated positively with the estimated glomerular filtration rate in cases of MCD. The density of caspase-3-labeled podocytes in FSGS and MCD was higher than control group, and a higher number of podocytes with an evidence of necrosis was detected in FSGS cases than in MCD and control cases. Podocytes from patients diagnosed with FSGS showed more morphological and functional alterations resulting from a larger number of lesions and reduced cell adaptation.
Collapse
Affiliation(s)
- Crislaine Aparecida da Silva
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Maria Luíza Gonçalves dos Reis Monteiro
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Liliane Silvano Araújo
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Monise Gini Urzedo
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Lenaldo Branco Rocha
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia dos Reis
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
12
|
Blaine J, Dylewski J. Regulation of the Actin Cytoskeleton in Podocytes. Cells 2020; 9:cells9071700. [PMID: 32708597 PMCID: PMC7408282 DOI: 10.3390/cells9071700] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Podocytes are an integral part of the glomerular filtration barrier, a structure that prevents filtration of large proteins and macromolecules into the urine. Podocyte function is dependent on actin cytoskeleton regulation within the foot processes, structures that link podocytes to the glomerular basement membrane. Actin cytoskeleton dynamics in podocyte foot processes are complex and regulated by multiple proteins and other factors. There are two key signal integration and structural hubs within foot processes that regulate the actin cytoskeleton: the slit diaphragm and focal adhesions. Both modulate actin filament extension as well as foot process mobility. No matter what the initial cause, the final common pathway of podocyte damage is dysregulation of the actin cytoskeleton leading to foot process retraction and proteinuria. Disruption of the actin cytoskeleton can be due to acquired causes or to genetic mutations in key actin regulatory and signaling proteins. Here, we describe the major structural and signaling components that regulate the actin cytoskeleton in podocytes as well as acquired and genetic causes of actin dysregulation.
Collapse
Affiliation(s)
- Judith Blaine
- Renal Division, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - James Dylewski
- Renal Division, University of Colorado Anschutz Medical Campus and Denver Health Medical Center, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +303-724-4841
| |
Collapse
|
13
|
Sipovsky VG, Nevorotin AI, Avsiewitsch IV, Smirnov AV. [Ultrastructural study of podocyte alterations in non proliferative glomerulopathy]. Arkh Patol 2019; 81:51-58. [PMID: 31317931 DOI: 10.17116/patol20198103151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ultrastructural changes in podocytes are an important diagnostic and prognostic marker for nephropathies. However, the biomedical understanding of detected submicroscopic changes in podocytes remains controversial. OBJECTIVE To investigate the relationship between the ultrastructural changes of podocytes (fusion of cytopodia and denudation of the basement membrane as a result of their desquamation) with a number of clinical and laboratory indicators of kidney dysfunction in case of non-proliferative glomerulopathies (NPGP). Thirty-seven patients (23 men, 14 women) with NPGP, including 8 with focal segmental glomerulosclerosis (FSGS), 17 with membranous nephropathy (MN), and 12 with minimal change disease (MCD), were examined. SUBJECT AND METHODS All the patients underwent standard laboratory and instrumental studies: determinations of the levels of total serum cholesterol (mmol/l), total serum protein (g/l); serum albumin (g/l); CKD-EPI glomerular filtration rate (GFR) (ml/min/1.73 m2), and daily protein loss (g/day). Light optical changes were measured; completely sclerotic and/or focally segmentally sclerotic glomeruli were taken into account. Quantitative ultrastructural stereological analysis was carried out estimating the cytopodium width (CPW) and the degree of glomerular basement membrane denudation (GBMD) (%). RESULTS NPGP cases showed the largest number of sclerotic glomeruli in FSGS, which was accompanied by the lowest level of daily proteinuria and GFR. Quantitative values of CPW were associated with the level of daily protein loss (r=0.47; p < 0.05) and serum albumin (r=-0.57; p <0.05) in patients with nephrotic syndrome. In MN, the absolute value of CPW was larger than that in the other two patient groups. A correlation analysis of CPW and GBMD values among patients with NPGP revealed a statistically insignificant negative relation between these morphometric parameters. However, when a subgroup of patients with podocytopathies (only MCD and FSGS) was identified in the study group, this relationship was found to be significant (r=-0.54; p=0.012). CONCLUSION The patients with NPGP exhibited a relationship between the severity of nephrotic syndrome and proteinuria/hypoalbuminemia, on the one hand, and CPW, on the other. The established negative relationship between CPW and the percentage of GBMD in the subgroup of patients with podocytopathies may be due to the early stages of podocyte injury, which are accompanied by transient GBMD.
Collapse
Affiliation(s)
- V G Sipovsky
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - A I Nevorotin
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - I V Avsiewitsch
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - A V Smirnov
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| |
Collapse
|
14
|
Nilsson D, Heglind M, Arani Z, Enerbäck S. Foxc2 is essential for podocyte function. Physiol Rep 2019; 7:e14083. [PMID: 31062503 PMCID: PMC6503019 DOI: 10.14814/phy2.14083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/29/2022] Open
Abstract
Foxc2 is one of the earliest podocyte markers during glomerular development. To circumvent embryonic lethal effects of global deletion of Foxc2, and to specifically investigate the role of Foxc2 in podocytes, we generated mice with a podocyte-specific Foxc2 deletion. Mice carrying the homozygous deletion developed early proteinuria which progressed rapidly into end stage kidney failure and death around postnatal day 10. Conditional loss of Foxc2 in podocytes caused typical characteristics of podocyte injury, such as podocyte foot process effacement and podocyte microvillus transformation, probably caused by disruption of the slit diaphragm. These effects were accompanied by a redistribution of several proteins known to be necessary for correct podocyte structure. One target gene that showed reduced glomerular expression was Nrp1, the gene encoding neuropilin 1, a protein that has been linked to diabetic nephropathy and proteinuria. We could show that NRP1 was regulated by Foxc2 in vitro, but podocyte-specific ablation of Nrp1 in mice did not generate any phenotype in terms of proteinuria, suggesting that the gene might have more important roles in endothelial cells than in podocytes. Taken together, this study highlights a critical role for Foxc2 as an important gene for podocyte function.
Collapse
Affiliation(s)
- Daniel Nilsson
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Mikael Heglind
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Zahra Arani
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
15
|
Musah S, Dimitrakakis N, Camacho DM, Church GM, Ingber DE. Directed differentiation of human induced pluripotent stem cells into mature kidney podocytes and establishment of a Glomerulus Chip. Nat Protoc 2019; 13:1662-1685. [PMID: 29995874 DOI: 10.1038/s41596-018-0007-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protocols have been established to direct the differentiation of human induced pluripotent stem (iPS) cells into nephron progenitor cells and organoids containing many types of kidney cells, but it has been difficult to direct the differentiation of iPS cells to form specific types of mature human kidney cells with high yield. Here, we describe a detailed protocol for the directed differentiation of human iPS cells into mature, post-mitotic kidney glomerular podocytes with high (>90%) efficiency within 26 d and under chemically defined conditions, without genetic manipulations or subpopulation selection. We also describe how these iPS cell-derived podocytes may be induced to form within a microfluidic organ-on-a-chip (Organ Chip) culture device to build a human kidney Glomerulus Chip that mimics the structure and function of the kidney glomerular capillary wall in vitro within 35 d (starting with undifferentiated iPS cells). The podocyte differentiation protocol requires skills for culturing iPS cells, and the development of a Glomerulus Chip requires some experience with building and operating microfluidic cell culture systems. This method could be useful for applications in nephrotoxicity screening, therapeutic development, and regenerative medicine, as well as mechanistic study of kidney development and disease.
Collapse
Affiliation(s)
- Samira Musah
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA. .,Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
16
|
Ichimura K, Miyaki T, Kawasaki Y, Kinoshita M, Kakuta S, Sakai T. Morphological Processes of Foot Process Effacement in Puromycin Aminonucleoside Nephrosis Revealed by FIB/SEM Tomography. J Am Soc Nephrol 2018; 30:96-108. [PMID: 30514724 DOI: 10.1681/asn.2018020139] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 11/07/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Foot process effacement is one of the pathologic indicators of podocyte injury. However, the morphologic changes associated with it remain unclear. METHODS To clarify the developmental process, we analyzed puromycin nephrotic podocytes reconstructed from serial focused-ion beam/scanning electron microscopy (FIB/SEM) images. RESULTS Intact podocytes consisted of four subcellular compartments: cell body, primary process, ridge-like prominence (RLP), and foot process. The RLP, a longitudinal protrusion from the basal surface of the cell body and primary process, served as an adhesive apparatus for the cell body and primary process to attach to the glomerular basement membrane. Foot processes protruded from both sides of the RLP. In puromycin nephrotic podocytes, foot process effacement occurred in two ways: by type-1 retraction, where the foot processes retracted while maintaining their rounded tips; or type-2 retraction, where they narrowed across their entire lengths, tapering toward the tips. Puromycin nephrotic podocytes also exhibited several alterations associated with foot process effacement, such as deformation of the cell body, retraction of RLPs, and cytoplasmic fragmentation. Finally, podocytes were reorganized into a broad, flattened shape. CONCLUSIONS The three-dimensional reconstruction of podocytes by serial FIB/SEM images revealed the morphologic changes involved in foot process effacement in greater detail than previously described.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Anatomy and Life Structure and .,Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | | | | | | | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | | |
Collapse
|
17
|
Chebotareva NV, Bobkova IN, Lysenko LV. The role of podocytes dysfunction in chronic glomerulonephritis progression. TERAPEVT ARKH 2018; 90:92-97. [PMID: 30701911 DOI: 10.26442/terarkh201890692-97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the review, the mechanisms of podocytes damage underlying the development of proteinuria and progression of glomerulosclerosis in chronic glomerulonephritis are discussed in detail. The results of experimental and clinical studies are presented. Under the different immune and non-immune factors the podocytes form a stereotyped response to damage consisting in the reorganization of the actin cytoskeleton, foot process effacement, the detachment of podocytes from the glomerular basement membrane, and the appearance of specific podocyte proteins and whole cells (podocyturia) in the urine. Massive podocyturia in a limited proliferative capacity of podocytes leads to reduce their total count in the glomerulus (podocytopenia) and the development of glomerulosclerosis. The authors describe the line of markers of the podocyte injury and invasive and non-invasive methods of their assessment. In addition, the relationship of podocyturia level with proteinuria and renal dysfunction are discussed, the prospects of assessment the podocyte proteins in urine for assessing of glomerular damage severity and glomerulosclerosis risk are examined.
Collapse
Affiliation(s)
- N V Chebotareva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| | - I N Bobkova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| | - L V Lysenko
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
18
|
Suleiman HY, Roth R, Jain S, Heuser JE, Shaw AS, Miner JH. Injury-induced actin cytoskeleton reorganization in podocytes revealed by super-resolution microscopy. JCI Insight 2017; 2:94137. [PMID: 28814668 DOI: 10.1172/jci.insight.94137] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/07/2017] [Indexed: 02/06/2023] Open
Abstract
The architectural integrity of tissues requires complex interactions, both between cells and between cells and the extracellular matrix. Fundamental to cell and tissue homeostasis are the specific mechanical forces conveyed by the actomyosin cytoskeleton. Here we used super-resolution imaging methods to visualize the actin cytoskeleton in the kidney glomerulus, an organized collection of capillaries that filters the blood to make the primary urine. Our analysis of both mouse and human glomeruli reveals a network of myosin IIA-containing contractile actin cables within podocyte cell bodies and major processes at the outer aspects of the glomerular tuft. These likely exert force on an underlying network of myosin IIA-negative, noncontractile actin fibers present within podocyte foot processes that function to both anchor the cells to the glomerular basement membrane and stabilize the slit diaphragm against the pressure of fluid flow. After injuries that disrupt the kidney filtration barrier and cause foot process effacement, the podocyte's contractile actomyosin network relocates to the basolateral surface of the cell, manifesting as sarcomere-like structures juxtaposed to the basement membrane. Our findings suggest a new model of the podocyte actin cytoskeleton in health and disease and suggest the existence of novel mechanisms that regulate podocyte architecture.
Collapse
Affiliation(s)
- Hani Y Suleiman
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robyn Roth
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John E Heuser
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, Japan
| | | | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Ha TS, Park HY, Seong SB, Ahn HY. Puromycin aminonucleoside increases podocyte permeability by modulating ZO-1 in an oxidative stress-dependent manner. Exp Cell Res 2015; 340:139-49. [PMID: 26683996 DOI: 10.1016/j.yexcr.2015.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/17/2015] [Accepted: 12/08/2015] [Indexed: 11/30/2022]
Abstract
Puromycin aminonucleoside (PAN)-induced nephrosis is a widely studied animal model of human idiopathic nephrotic syndrome because PAN injection into rats results in increased glomerular permeability with the characteristic ultrastructural changes in podocytes similar to human nephrosis. To investigate the role of zonula occludens (ZO)-1 and oxidative stress on PAN-induced podocyte phenotypical changes and hyperpermeability in vitro, we cultured rat and mouse podocytes and treated with various concentrations of PAN. PAN treatment increased oxidative stress level of podocytes significantly with the induction of Nox4. In addition, PAN changed the ultrastructure of podocytes, such as shortening and fusion of microvilli, and the separation of intercellular gaps, which were improved by anti-oxidative vitamin C and Nox4 siRNA. PAN also disrupted the intercellular linear ZO-1 staining and induced inner cytoplasmic re-localization of ZO-1 protein, resulting in increased podocyte intercellular permeability. PAN reduced ZO-1 protein amount and mRNA expression in a dose-dependent manner, which means that PAN could also modulate ZO-1 protein transcriptionally. However, the decreased ZO-1 protein of podocytes by PAN was improved by Nox4 siRNA transfection. Furthermore, vitamin C mitigated the quantitative and distributional disturbances of ZO-1 protein caused by PAN. Our results demonstrate that the phenotypical changes of intercellular ZO-1 by oxidative stress via Nox4 likely contribute to the glomerular hyperpermeability caused by PAN.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, South Korea.
| | - Hye-Young Park
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Su-Bin Seong
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Hee Yul Ahn
- Department of Pharmacology, College of Medicine, Chungbuk National University, Cheongju, South Korea
| |
Collapse
|
20
|
Minimal Change Disease as a Secondary and Reversible Event of a Renal Transplant Case with Systemic Lupus Erythematosus. Case Rep Nephrol 2015; 2015:987212. [PMID: 26351598 PMCID: PMC4550805 DOI: 10.1155/2015/987212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/30/2015] [Accepted: 08/02/2015] [Indexed: 11/23/2022] Open
Abstract
Secondary causes of minimal change disease (MCD) account for a minority of cases compared to its primary or idiopathic form and provide ground for consideration of common mechanisms of pathogenesis. In this paper we report a case of a 27-year-old Latina woman, a renal transplant recipient with systemic lupus erythematosus (SLE), who developed nephrotic range proteinuria 6 months after transplantation. The patient had recurrent acute renal failure and multiple biopsies were consistent with MCD. However, she lacked any other features of the typical nephrotic syndrome. An angiogram revealed a right external iliac vein stenosis in the region of renal vein anastomosis, which when restored resulted in normalization of creatinine and relief from proteinuria. We report a rare case of MCD developing secondary to iliac vein stenosis in a renal transplant recipient with SLE. Additionally we suggest that, in the event of biopsy-proven MCD presenting as an atypical nephrotic syndrome, alternative or secondary, potentially reversible, causes should be considered and explored.
Collapse
|
21
|
Three-dimensional architecture of podocytes revealed by block-face scanning electron microscopy. Sci Rep 2015; 5:8993. [PMID: 25759085 PMCID: PMC4355681 DOI: 10.1038/srep08993] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/12/2015] [Indexed: 12/31/2022] Open
Abstract
Block-face imaging is a scanning electron microscopic technique which enables easier acquisition of serial ultrastructural images directly from the surface of resin-embedded biological samples with a similar quality to transmission electron micrographs. In the present study, we analyzed the three-dimensional architecture of podocytes using serial block-face imaging. It was previously believed that podocytes are divided into three kinds of subcellular compartment: cell body, primary process, and foot process, which are simply aligned in this order. When the reconstructed podocytes were viewed from their basal side, the foot processes were branched from a ridge-like prominence, which was formed on the basal surface of the primary process and was similar to the usual foot processes in structure. Moreover, from the cell body, the foot processes were also emerged via the ridge-like prominence, as found in the primary process. The ridge-like prominence anchored the cell body and primary process to the glomerular basement membrane, and connected the foot processes to the cell body and primary process. In conclusion, serial block-face imaging is a powerful tool for clear understanding the three-dimensional architecture of podocytes through its ability to reveal novel structures which were difficult to determine by conventional transmission and scanning electron microscopes alone.
Collapse
|
22
|
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: 71] [Impact Index Per Article: 7.1] [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
|
23
|
Lal MA, Andersson AC, Katayama K, Xiao Z, Nukui M, Hultenby K, Wernerson A, Tryggvason K. Rhophilin-1 is a key regulator of the podocyte cytoskeleton and is essential for glomerular filtration. J Am Soc Nephrol 2014; 26:647-62. [PMID: 25071083 DOI: 10.1681/asn.2013111195] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Rhophilin-1 is a Rho GTPase-interacting protein, the biologic function of which is largely unknown. Here, we identify and describe the functional role of Rhophilin-1 as a novel podocyte-specific protein of the kidney glomerulus. Rhophilin-1 knockout mice were phenotypically normal at birth but developed albuminuria at about 2 weeks of age. Kidneys from severely albuminuric mice revealed widespread podocyte foot process effacement, thickening of the glomerular basement membrane, and FSGS-like lesions. The absence of any overt changes in the expression of podocyte proteins at the onset of proteinuria suggested that the primary cause of podocyte abnormalities in Rhpn1-null mice was the result of cell-autonomous, Rhophilin-1-dependent signaling events. In culture, Rhophilin-1 was detected at the plasma membrane leading edge of primary podocytes, where it elicited remodeling of the actin cytoskeleton network. This effect of Rhophilin-1 on actin cytoskeleton organization associated with inhibitory effects on Rho-dependent phosphorylation of the myosin regulatory light chain and stress fiber formation. Conversely, phosphorylation of myosin regulatory light chain increased in podocyte foot processes of Rhpn1(-/-) mice, implicating altered actinomyosin contractility in foot process effacement and compromised filtration capacity. Targeted deletion of RhoA in podocytes of Rhophilin-1 knockout mice exacerbated the renal injury. Taken together, our results indicate that Rhophilin-1 is essential for the integrity of the glomerular filtration barrier and that this protein is a key determinant of podocyte cytoskeleton architecture.
Collapse
Affiliation(s)
- Mark A Lal
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | | | - Kan Katayama
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Ziejie Xiao
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Masatoshi Nukui
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Kjell Hultenby
- Department of Laboratory Medicine, Division of Clinical Research Center, and
| | - Annika Wernerson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics,
| |
Collapse
|
24
|
Ichimura K, Powell R, Nakamura T, Kurihara H, Sakai T, Obara T. Podocalyxin regulates pronephric glomerular development in zebrafish. Physiol Rep 2013; 1. [PMID: 24224085 PMCID: PMC3819108 DOI: 10.1002/phy2.74] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Vertebrate glomerular podocytes possess a highly sialylated transmembrane glycoprotein, Podocalyxin. In mammals, the sialic acid of Podocalyxin plays a crucial role in the formation of the characteristic podocyte architecture required for glomerular filtration. We examined the function of Podocalyxin in the developing zebrafish pronephros by disrupting the expression of podocalyxin through the use of morpholino antisense oligonucleotides. Podocalyxin was localized at the apical membrane of podocytes throughout pronephric glomerular development in zebrafish. Translational blocking of podocalyxin expression resulted in pericardial edema and a hypoplastic glomerulus. Whereas regular foot processes with a slit diaphragm covered 66.7 ± 7.8% of the urinary surface of glomerular basement membrane in control fish, only 14.4 ± 7.5% of this area was covered with regular foot processes in the translationally blocked morphants. Splice blocking of podocalyxin exon 2, which partially encodes the bulky mucin domain with extensive sialic acid-containing sugar chains, resulted in the deletion of 53% of mucin domain-coding sequence from podocalyxin mRNA. Approximately 40% of these splice-blocked morphants had mild pericardial edema. Although the pronephric glomerulus in the splice-blocked morphants exhibited almost normal appearance with developed glomerular capillaries and mesangium, they had only 36.3 ± 6.9% of the area covered with regular foot processes. In conclusion, Podocalyxin is predominantly expressed in the podocytes and plays a distinct role in the formation of the podocyte foot processes with a slit diaphragm during zebrafish pronephric development.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA ; Department of Anatomy and Life Structure, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Kim EY, Anderson M, Wilson C, Hagmann H, Benzing T, Dryer SE. NOX2 interacts with podocyte TRPC6 channels and contributes to their activation by diacylglycerol: essential role of podocin in formation of this complex. Am J Physiol Cell Physiol 2013; 305:C960-71. [PMID: 23948707 DOI: 10.1152/ajpcell.00191.2013] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Canonical transient receptor potential-6 (TRPC6) channels have been implicated in the pathophysiology of glomerular diseases. TRPC6 channels are typically activated by diacylglycerol (DAG) during PLC-dependent transduction cascades. TRPC6 channels can also be activated by reactive oxygen species (ROS). We previously showed that podocin is required for DAG analogs to produce robust activation of TRPC6 channels in podocytes. Here we show that endogenous TRPC6 channels in immortalized podocytes reciprocally coimmunoprecipitate with the catalytic subunit of the NADPH oxidase NOX2 (gp91(phox)). The NOX2-TRPC6 interaction was not detected in cells stably expressing a short hairpin RNA targeting podocin, although NOX2 and TRPC6 were present at normal levels. Application of a membrane-permeable DAG analog [1-oleoyl-2-acetyl-sn-glycerol (OAG)] increased generation of ROS in podocytes, but this effect was not detected in podocin knockdown cells. OAG also increased steady-state surface expression of the NOX2 regulatory subunit p47(phox). In whole cell recordings, TRPC6 activation by OAG was reduced in podocytes pretreated with the NOX2 inhibitor apocynin, by the pan-NOX inhibitor diphenylene iodonium, and by tempol, a ROS quencher. Cholesterol depletion and disruption of lipid rafts by methyl-β-cyclodextrin reduced activation of podocyte TRPC6 channels by OAG and also eliminated the NOX2-TRPC6 interaction as assessed by coimmunoprecipitation. These data suggest that active NOX2 assembles with TRPC6 at podocin-organized sterol-rich raft domains and becomes catalytically active in response to DAG. The localized production of ROS contributes to TRPC6 activation by chemical stimuli such as DAG. Podocin appears to be necessary for assembly of the NOX2-TRPC6 complex in lipid rafts.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | | | | | | | | |
Collapse
|
26
|
Kimura J, Ichii O, Otsuka S, Sasaki H, Hashimoto Y, Kon Y. Close relations between podocyte injuries and membranous proliferative glomerulonephritis in autoimmune murine models. Am J Nephrol 2013; 38:27-38. [PMID: 23817053 DOI: 10.1159/000353093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/10/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Membranous proliferative glomerulonephritis (MPGN) is a major primary cause of chronic kidney disease (CKD). Podocyte injury is crucial in the pathogenesis of glomerular disease with proteinuria, leading to CKD. To assess podocyte injuries in MPGN, the pathological features of spontaneous murine models were analyzed. METHODS The autoimmune-prone mice strains BXSB/MpJ-Yaa and B6.MRL-(D1Mit202-D1Mit403) were used as the MPGN models, and BXSB/MpJ-Yaa(+) and C57BL/6 were used as the respective controls. In addition to clinical parameters and glomerular histopathology, the protein and mRNA levels of podocyte functional markers were evaluated as indices for podocyte injuries. The relation between MPGN pathology and podocyte injuries was analyzed by statistical correlation. RESULTS Both models developed MPGN with albuminuria and elevated serum anti-double-strand DNA (dsDNA) antibody levels. BXSB/MpJ-Yaa and B6.MRL showed severe proliferative lesions with T and B cell infiltrations and membranous lesions with T cell infiltrations, respectively. Foot process effacement and microvillus-like structure formation were observed ultrastructurally in the podocytes of both MPGN models. Furthermore, both MPGN models showed a decrease in immune-positive areas of nephrin, podocin and synaptopodin in the glomerulus, and in the mRNA expression of Nphs1, Nphs2, Synpo, Actn4, Cd2ap, and Podxl in the isolated glomerulus. Significant negative correlations were detected between serum anti-dsDNA antibody levels and glomerular Nphs1 expression, and between urinary albumin-to-creatinine ratio and glomerular expression of Nphs1, Synpo, Actn4, Cd2ap, or Podxl. CONCLUSION MPGN models clearly developed podocyte injuries characterized by the decreased expression of podocyte functional markers with altered morphology. These data emphasized the importance of regulation of podocyte injuries in MPGN.
Collapse
Affiliation(s)
- Junpei Kimura
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Kriz W, Shirato I, Nagata M, LeHir M, Lemley KV. The podocyte's response to stress: the enigma of foot process effacement. Am J Physiol Renal Physiol 2013; 304:F333-47. [DOI: 10.1152/ajprenal.00478.2012] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Progressive loss of podocytes is the most frequent cause accounting for end-stage renal failure. Podocytes are complex, terminally differentiated cells incapable of replicating. Thus lost podocytes cannot be replaced by proliferation of neighboring undamaged cells. Moreover, podocytes occupy a unique position as epithelial cells, adhering to the glomerular basement membrane (GBM) only by their processes, whereas their cell bodies float within the filtrate in Bowman's space. This exposes podocytes to the danger of being lost by detachment as viable cells from the GBM. Indeed, podocytes are continually excreted as viable cells in the urine, and the rate of excretion dramatically increases in glomerular diseases. Given this situation, it is likely that evolution has developed particular mechanisms whereby podocytes resist cell detachment. Podocytes respond to stress and injury by undergoing tremendous changes in shape. Foot process effacement is the most prominent and, yet in some ways, the most enigmatic of those changes. This review summarizes the various structural responses of podocytes to injury, focusing on foot process effacement and detachment. We raise the hypothesis that foot process effacement represents a protective response of podocytes to escape detachment from the GBM.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Anatomy and Developmental Biology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Isao Shirato
- Division of Nephrology, Department of Internal Medicine, Juntendo University, School of Medicine, Tokyo, Japan
| | - Michio Nagata
- Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-City, Japan
| | - Michel LeHir
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; and
| | - Kevin V. Lemley
- Division of Nephrology, Children's Hospital Los Angeles, Los Angeles, California
| |
Collapse
|
28
|
Kim EY, Anderson M, Dryer SE. Sustained activation of N-methyl-D-aspartate receptors in podoctyes leads to oxidative stress, mobilization of transient receptor potential canonical 6 channels, nuclear factor of activated T cells activation, and apoptotic cell death. Mol Pharmacol 2012; 82:728-37. [PMID: 22828802 DOI: 10.1124/mol.112.079376] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Atypical N-methyl-D-aspartate (NMDA) receptors are expressed in podocytes. Sustained (≥24 h) application of 50 to100 μM NMDA to immortalized mouse podocytes evoked a marked increase in the production of reactive oxygen species(ROS) such as H₂O₂. This effect of NMDA was associated with increased cell-surface expression of p47(phox), a cytosolic regulatory subunit of the NADPH oxidase NOX2. NMDA-evoked generation of ROS drove an increase in steady-state surface expression of transient receptor potential canonical (TRPC) 6 channels, which was blocked by the NMDA antagonist dizocilpine(MK-801) and by a membrane-permeable scavenger of ROS. The effect of NMDA on TRPC6 was observed using cell surface biotinylation assays and also with whole-cell recordings made under conditions designed to facilitate detection of current through TRPC6. NMDA mobilization of TRPC6 channels was blocked by concurrent treatment with the NMDA antagonist MK-801 and by a membrane-permeable scavenger ofROS. Mobilization of TRPC6 was also evoked by L-homocysteic acid. NMDA treatment also increased nuclear localization of endogenous nuclear factor of activated T cells, which could be blocked by MK-801, by scavenging ROS, by the calcineurin inhibitor cyclosporine, and by the TRPC channel inhibitor 1-[2-(4-methoxyphenyl)-2-[3-(4-methoxyphenyl)propoxy]ethyl]imidazole (SKF-96365). NMDA treatment also evoked robust activation of Rho but not Rac,consistent with previous studies of downstream effectors of TRPC6 activation. Exposing cells to NMDA for 24 h reduced total and cell surface expression of the podocyte markers nephrin and podocin, but there was no loss of cells. With longer NMDA exposure (72 h), we observed loss of cells associated with nuclear fragmentation and increased expression of caspase-3, caspase-6, and Bax, suggesting an apoptotic process.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | | | | |
Collapse
|
29
|
An overview of molecular mechanism of nephrotic syndrome. Int J Nephrol 2012; 2012:937623. [PMID: 22844593 PMCID: PMC3401527 DOI: 10.1155/2012/937623] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 06/20/2012] [Accepted: 06/20/2012] [Indexed: 12/13/2022] Open
Abstract
Podocytopathies (minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS)) together with membranous nephropathy are the main causes of nephrotic syndrome. Some changes on the expression of nephrin, podocin, TGF-β, and slit diaphragm components as well as transcription factors and transmembrane proteins have been demonstrated in podocytopathies. Considering the pathogenesis of proteinuria, some elucidations have been directed towards the involvement of epithelial-mesenchymal transition. Moreover, the usefulness of some markers such as TGF-β1, nephrin, synaptopodin, dystroglycans, and malondialdehyde have been determined in the differentiation between MCD and FSGS. Experimental models and human samples indicated an essential role of autoantibodies in membranous glomerulonephritis, kidney damage, and proteinuria events. Megalin and phospholipase-A2-receptor have been described as antigens responsible for the formation of the subepithelial immune complexes and renal disease occurrence. In addition, the complement system seems to play a key role in basal membrane damage and in the development of proteinuria in membranous nephropathy. This paper focuses on the common molecular changes involved in the development of nephrotic proteinuria.
Collapse
|
30
|
Grgic I, Brooks CR, Hofmeister AF, Bijol V, Bonventre JV, Humphreys BD. Imaging of podocyte foot processes by fluorescence microscopy. J Am Soc Nephrol 2012; 23:785-91. [PMID: 22362911 DOI: 10.1681/asn.2011100988] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Visualizing podocyte foot processes requires electron microscopy, a technique that depends on special equipment, requires immunogold for colabeling, and does not take advantage of the growing number of in vivo fluorophores available. To address these limitations, we developed a genetic strategy to allow detailed visualization of single podocytes and their foot processes by conventional fluorescence microscopy. We generated a transgenic mouse line expressing a GFP-Cre-ERT2 fusion protein under the control of the collagen α1(I) promoter with strong podocyte expression. Administration of submaximal tamoxifen allowed genetic labeling of single podocytes when crossed with a Cre-reporter line. Of three different reporter systems that we evaluated for the ability to reveal fine structural details of podocytes, bigenic Coll1α1GCE;Gt(ROSA)26Sor(tm9(CAG-tdTomato)) mice allowed podocyte labeling with a strong and homogeneous reporter signal that was easily observed by epifluorescence. We could easily detect anatomic features of podocytes down to tertiary foot processes, and we were able to visualize and quantitate ultrastructural changes to foot processes after podocyte injury. In summary, using this method of genetic labeling and conventional fluorescence microscopy to visualize podocyte foot processes will complement electron microscopy and facilitate the analysis of podocytes and their precursors in vivo.
Collapse
Affiliation(s)
- Ivica Grgic
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
31
|
Tyritzis SI, Zachariades M, Evangelou K, Gorgoulis VG, Kyroudi-Voulgari A, Pavlakis K, Troupis TG, Constantinides CA. Effects of Prolonged Warm and Cold Ischemia in a Solitary Kidney Animal Model after Partial Nephrectomy: An Ultrastructural Investigation. Ultrastruct Pathol 2011; 35:60-5. [DOI: 10.3109/01913123.2010.542880] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Cheng H, Harris RC. The glomerulus--a view from the outside--the podocyte. Int J Biochem Cell Biol 2010; 42:1380-7. [PMID: 20542138 DOI: 10.1016/j.biocel.2010.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 04/12/2010] [Accepted: 05/30/2010] [Indexed: 01/11/2023]
Abstract
In the past decade, podocyte research has been greatly aided by the development of powerful new molecular, cellular and animal tools, leading to elucidation of an increasing number of proteins involved in podocyte function and identification of mutated genes in hereditary glomerulopathies. Accumulating evidence indicates that podocyte disorders may not only underlie these hereditary glomerulopathies but also play crucial role in a broad spectrum of acquired glomerular diseases. Genetic susceptibility, environmental influence and systemic responses are all involved in the mediation of the pathogenesis of podocytopathies. Injured podocytes may predisopose to further injury of other podocytes and other adjacent/distant renal cells in a vicious cycle, leading to inexorable progression of glomerular injury. The classic view is that podocytes have a limited ability to proliferate in the normal mature kidney. However, recent research in rodents has provided suggestive evidence for podocyte regeneration resulting from differentiation of progenitor cells within Bowman's capsule.
Collapse
Affiliation(s)
- Huifang Cheng
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-2372, USA.
| | | |
Collapse
|
33
|
Ha TS. High-glucose and advanced glycosylation end products increased podocyte permeability via PI3-K/Akt signaling. J Mol Med (Berl) 2010; 88:391-400. [PMID: 20054520 DOI: 10.1007/s00109-009-0575-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 11/07/2009] [Accepted: 12/07/2009] [Indexed: 02/06/2023]
Abstract
Regardless of the underlying disease, the proteinuric condition demonstrates ultrastructural changes in podocytes with retraction and effacement of the highly specialized interdigitating foot processes. To investigate how high-glucose (HG) and advanced glycosylation end products (AGE) induce podocyte phenotypical changes, including quantitative and distributional changes of zonula occludens (ZO)-1 protein and search for the signaling mechanisms, we cultured rat glomerular epithelial cells (GEpC) and mouse podocytes under: (1) normal glucose (5 mM, control); (2) HG (30 mM); (3) AGE-added; or (4) HG plus AGE-added conditions. HG plus AGE increased the permeability of monolayered GEpCs and induced ultrastructural separation between confluent GEpCs. ZO-1 moved to inner actin filament complexes in both AGE- and/or HG by confocal imaging. HG plus AGE-added condition also decreased ZO-1 protein amount and mRNA expression compared to normal glucose or osmotic control conditions. We could also confirm the induction of RAGE (receptor for AGE) and PI3-K/Akt signaling pathway by AGE and HG. In addition, LY294002, a PI3-K inhibitor, could prevent the quantitative and distributional changes of ZO-1 and RAGE and the increased permeability induced by HG and AGE. These findings suggest that diabetic conditions induce the podocyte ZO-1 changes via RAGE and PI3-K/Akt signaling, leading to increased permeability.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, College of Medicine, Chungbuk National University, Gaeshin-dong 48, Heungdeok-gu, Cheongju, Chungbuk, 361-240, Korea.
| |
Collapse
|
34
|
ASANUMA KATSUHIKO, YANAGIDA-ASANUMA ETSUKO, TAKAGI MIYUKI, KODAMA FUMIKO, TOMINO YASUHIKO. The role of podocytes in proteinuria. Nephrology (Carlton) 2007; 12 Suppl 3:S15-20. [DOI: 10.1111/j.1440-1797.2007.00876.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
35
|
Barisoni L, Schnaper HW, Kopp JB. A proposed taxonomy for the podocytopathies: a reassessment of the primary nephrotic diseases. Clin J Am Soc Nephrol 2007; 2:529-42. [PMID: 17699461 DOI: 10.2215/cjn.04121206] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A spectrum of proteinuric glomerular diseases results from podocyte abnormalities. The understanding of these podocytopathies has greatly expanded in recent years, particularly with the discovery of more than a dozen genetic mutations that are associated with loss of podocyte functional integrity. It is apparent that classification of the podocytopathies on the basis of morphology alone is inadequate to capture fully the complexity of these disorders. Herein is proposed a taxonomy for the podocytopathies that classifies along two dimensions: Histopathology, including podocyte phenotype and glomerular morphology (minimal-change nephropathy, focal segmental glomerulosclerosis, diffuse mesangial sclerosis, and collapsing glomerulopathy), and etiology (idiopathic, genetic, and reactive forms). A more complete understanding of the similarities and differences among podocyte diseases will help the renal pathologist and the nephrologist communicate more effectively about the diagnosis; this in turn will help the nephrologist provide more accurate prognostic information and select the optimal therapy for these often problematic diseases. It is proposed that final diagnosis of the podocytopathies should result from close collaboration between renal pathologists and nephrologists and should whenever possible include three elements: Morphologic entity, etiologic form, and specific pathogenic mechanism or association.
Collapse
Affiliation(s)
- Laura Barisoni
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
36
|
Yi F, dos Santos EA, Xia M, Chen QZ, Li PL, Li N. Podocyte injury and glomerulosclerosis in hyperhomocysteinemic rats. Am J Nephrol 2007; 27:262-8. [PMID: 17396029 DOI: 10.1159/000101471] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 03/02/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS We previously reported that increase in plasma homocysteine (Hcys) levels by a 6-week methionine treatment produced remarkable glomerular injury. However, the mechanism by which hyperhomocysteinemia (hHcys) produces glomerular injury remains unknown. The present study was to observe when glomerular injury happens during hHcys and to explore the possible role of podocyte injury in the progression of glomerulosclerosis associated with hHcys. METHODS Uninephrectomized Sprague-Dawley rats treated with methionine were used to examine the time course of glomerular injury induced by hHcys. RESULTS Creatinine clearance was not different until rats were treated with methionine for 6 weeks, although plasma Hcys levels significantly increased at the 1st week of methionine treatment. However, urinary albumin excretion increased at the 2nd week of methionine treatment. Morphological examinations showed that mesangial expansion occurred at the 2nd week and podocyte effacement was also observed as processed glomerular damage during hHcys. Immunofluorescence analyses demonstrated that podocin and nephrin expressions were reduced, while alpha-actinin-4 increased during hHcys. CONCLUSIONS Increased plasma Hcys level is an important pathogenic factor resulting in glomerular injury even in the very early time of hHcys. These pathogenic effects of Hcys are associated with podocyte injury and changed expression and distribution of podocyte-associated proteins.
Collapse
Affiliation(s)
- Fan Yi
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | | | |
Collapse
|
37
|
Morigi M, Buelli S, Zanchi C, Longaretti L, Macconi D, Benigni A, Moioli D, Remuzzi G, Zoja C. Shigatoxin-induced endothelin-1 expression in cultured podocytes autocrinally mediates actin remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:1965-75. [PMID: 17148661 PMCID: PMC1762486 DOI: 10.2353/ajpath.2006.051331] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Shigatoxin (Stx) is the offending agent of post-diarrheal hemolytic uremic syndrome, characterized by glomerular ischemic changes preceding microvascular thrombosis. Because podocytes are highly sensitive to Stx cytotoxicity and represent a source of vasoactive molecules, we studied whether Stx-2 modulated the production of endothelin-1 (ET-1), taken as candidate mediator of podocyte dysfunction. Stx-2 enhanced ET-1 mRNA and protein expression via activation of nuclear factor kappaB (NF-kappaB) and activator protein-1 (Ap-1) to the extent that transfection with the dominant-negative mutant of IkappaB-kinase 2 or with Ap-1 decoy oligodeoxynucleotides reduced ET-1 mRNA levels. We propose a role for p38 and p42/44 mitogen-activated protein kinases (MAPKs) in mediating NF-kappaB-dependent gene transcription induced by Stx-2, based on data that Stx-2 phosphorylated p38 and p42/44 MAPKs and that MAPK inhibitors reduced transcription of NF-kappaB promoter/luciferase reporter gene construct induced by Stx-2. Stx-2 caused F-actin redistribution and intercellular gaps via production of ET-1 acting on ETA receptor, because cytoskeleton changes were prevented by ETA receptor blockade. Exogenous ET-1 induced cytoskeleton rearrangement and intercellular gaps via phosphatidylinositol-3 kinase and Rho-kinase pathway and increased protein permeability across the podocyte monolayer. These data suggest that the podocyte is a target of Stx, a novel stimulus for the synthesis of ET-1, which may control cytoskeleton remodeling and glomerular permeability in an autocrine fashion.
Collapse
Affiliation(s)
- Marina Morigi
- Mario Negri Institute for Pharmacological Research, Unit of Nephrology and Dialysis, Azienda Ospedaliera, Ospedali Riuniti di Bergamo, Via Gavazzeni 11, 24125 Bergamo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ha TS. High glucose and advanced glycosylated end-products affect the expression of ?-actinin-4 in glomerular epithelial cells. Nephrology (Carlton) 2006; 11:435-41. [PMID: 17014558 DOI: 10.1111/j.1440-1797.2006.00668.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM To investigate the molecular basis for the phenotypic alterations of glomerular epithelial cells (GEpC, podocytes), involving cytoskeletal changes especially on alpha-actinin-4 as a candidate regulating the barrier to protein filtration and the podocyte actin cytoskeleton. METHODS To examine the effects of glucose and advanced glycosylated end-products (AGE) on alpha-actinin-4, the author cultured rat GEpC on AGE- or BSA-coated plates under normal (5 mmol) and high glucose (30 mmol) conditions and examined the distribution of alpha-actinin by confocal microscope and measured the change in alpha-actinin-4 production by western blotting and reverse transcription-polymerase chain reaction. RESULTS Confocal microscopy indicated that alpha-actinin-4 moved from the peripheral cytoplasm to inner actin filament complexes in the presence of AGE and high glucose. These changes might be related to the fusion of microvilli of cell surface examined by electron microscopy. In western blot analysis, AGE significantly decreased the amount of alpha-actinin by 28.1%. Furthermore, the combination of high glucose and AGE decreased the amount of alpha-actinin more significantly by 53.6% compared with that of the control. The mRNA expression for alpha-actinin-4 was not changed with high glucose or AGE-coated surfaces; however, when added, the combination of high glucose and AGE significantly decreased the expression of alpha-actinin-4 mRNA by 15.7% compared with that of the control. CONCLUSION The author suggests that both high glucose and AGE (either individually or in combination) induce the cytoplasmic translocation and the combination suppresses the production of alpha-actinin-4 at the transcriptional level with post-translational modification and these in vitro changes may explain the cytoskeletal changes of GEpC in diabetic conditions.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, Korea.
| |
Collapse
|
39
|
Abstract
The terminally differentiated podocyte, also called glomerular visceral epithelial cell, are highly specialized cells. They function as a critical size and charge barrier to prevent proteinuria. Podocytes are injured in diabetic and non-diabetic renal diseases. The clinical signature of podocyte injury is proteinuria, with or without loss of renal function owing to glomerulosclerosis. There is an exciting and expanding literature showing that hereditary, congenital, or acquired abnormalities in the molecular anatomy of podocytes leads to proteinuria, and at times, glomerulosclerosis. The change in podocyte shape, called effacement, is not simply a passive process following injury, but is owing to a complex interplay of proteins that comprise the molecular anatomy of the different protein domains of podocytes. These will be discussed in this review. Recent studies have also highlighted that a reduction in podocyte number directly causes proteinuria and glomerulosclerosis. This is owing to several factors, including the relative inability for these cells to proliferate, detachment, and apoptosis. The mechanisms of these events are being elucidated, and are discussed in this review. It is the hope that by delineating the events following injury to podocytes, therapies might be developed to reduce the burden of proteinuric renal diseases.
Collapse
Affiliation(s)
- S J Shankland
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, Washington 98195, USA.
| |
Collapse
|
40
|
Morigi M, Buelli S, Angioletti S, Zanchi C, Longaretti L, Zoja C, Galbusera M, Gastoldi S, Mundel P, Remuzzi G, Benigni A. In response to protein load podocytes reorganize cytoskeleton and modulate endothelin-1 gene: implication for permselective dysfunction of chronic nephropathies. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1309-20. [PMID: 15855633 PMCID: PMC1606387 DOI: 10.1016/s0002-9440(10)62350-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Effacement of podocyte foot processes occurs in many proteinuric nephropathies and is accompanied by rearrangement of the actin cytoskeleton. Here, we studied whether protein overload affects intracellular pathways, leading to cytoskeletal architecture changes and ultimately to podocyte dysfunction. Mouse podocytes bound and endocytosed both albumin and IgG via receptor-specific mechanisms. Protein overload caused redistribution of F-actin fibers instrumental to up-regulation of the prepro-endothelin (ET)-1 gene and production of the corresponding peptide. Increased DNA-binding activity for nuclear factor (NF)-kappaB and Ap-1 nuclear proteins was measured in nuclear extracts of podocytes exposed to excess proteins. Both Y27632, which inhibits Rho kinase-dependent stress fiber formation, and jasplakinolide, an F-actin stabilizer, decreased NF-kappaB and Ap-1 activity and reduced ET-1 expression. This suggested a role for the cytoskeleton, through activated Rho, in the regulation of the ET-1 peptide. Focal adhesion kinase (FAK), an integrin-associated nonreceptor tyrosine kinase, was phosphorylated by albumin treatment via Rho kinase-triggered actin reorganization. FAK activation led to NF-kappaB- and Ap-1-dependent ET-1 expression. These data suggest that reorganization of the actin cytoskeletal network in response to protein load is implicated in modulation of the ET-1 gene via Rho kinase-dependent FAK activation of NF-kappaB and Ap-1 in differentiated podocytes. Increased ET-1 generation might alter glomerular permselectivity and amplify the noxious effect of protein overload on dysfunctional podocytes.
Collapse
Affiliation(s)
- Marina Morigi
- Mario Negri Institute for Pharmacological Research, Via Gavazzeni 11, 24125 Bergamo, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rico M, Mukherjee A, Konieczkowski M, Bruggeman LA, Miller RT, Khan S, Schelling JR, Sedor JR. WT1-interacting protein and ZO-1 translocate into podocyte nuclei after puromycin aminonucleoside treatment. Am J Physiol Renal Physiol 2005; 289:F431-41. [PMID: 15798086 DOI: 10.1152/ajprenal.00389.2004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Podocyte differentiation is required for normal glomerular filtration barrier function and is regulated by the transcription factor WT1. We identified WT1-interacting protein (WTIP) and hypothesized that it functions as both a scaffold for slit diaphragm proteins and a corepressor of WT1 transcriptional activity by shuttling from cell-cell junctions to the nucleus after injury. Endogenous WTIP colocalizes with zonula occludens-1 (ZO-1) in cultured mouse podocyte adherens junctions. To model podocyte injury in vitro, we incubated differentiated podocytes with puromycin aminonucleoside (PAN; 100 microg/ml) for 24 h, which disassembled cell-cell contacts, rearranged actin cytoskeleton, and caused process retraction. Podocyte synaptopodin expression diminished after PAN treatment, consistent with podocyte dedifferentiation in some human glomerular diseases. To assess podocyte function, we measured albumin flux across differentiated podocytes cultured on collagen-coated Transwell filters. Albumin transit across PAN-treated cells increased to levels observed with undifferentiated podocytes. Consistent with our hypothesis, WTIP, as well as ZO-1, translocated from podocyte adherens junctions to nuclei in PAN-treated cells. Because WTIP is a transcriptional corepressor for WT1, we examined the effect of PAN on expression of retinoblastoma binding protein Rbbp7 (also known as RbAp46), a WT1 target gene expressed in S-shaped bodies during nephrogenesis. Rbbp7 expression in PAN-treated podocytes was reduced compared with untreated cells. In conclusion, WTIP translocates from cell-cell junctions to the nucleus in PAN-treated podocytes. We suggest that WTIP monitors slit diaphragm protein assembly and shuttles into the nucleus after podocyte injury, translating changes in slit diaphragm structure into altered gene expression and a less differentiated phenotype.
Collapse
Affiliation(s)
- Maribel Rico
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44109-1998, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Griffin SV, Hiromura K, Pippin J, Petermann AT, Blonski MJ, Krofft R, Takahashi S, Kulkarni AB, Shankland SJ. Cyclin-dependent kinase 5 is a regulator of podocyte differentiation, proliferation, and morphology. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1175-85. [PMID: 15466384 PMCID: PMC1618643 DOI: 10.1016/s0002-9440(10)63378-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Podocytes are highly specialized and terminally differentiated glomerular cells that play a vital role in renal physiology, including the prevention of proteinuria. Cyclin-dependent kinase 5 (CDK5) has been shown to influence several cellular processes in other terminally differentiated cells, in particular neurons. In this study, we examined the role of CDK5 in podocyte differentiation, proliferation, and morphology. In conditionally immortalized mouse podocytes in culture, CDK5 increased in association with podocyte differentiation. During mouse glomerulogenesis in vivo, CDK5 expression was predominantly detected in podocytes from the capillary loop stage to maturation and persisted in the podocytes of adult glomeruli. In contrast, CDK5 was markedly decreased in the proliferating and dedifferentiated podocytes of mice with anti-glomerular basement membrane nephritis and in human immunodeficiency virus transgenic mice. p35, the activator of CDK5, was also detected in podocytes and the p35/CDK5 complex was active. Cell fractionation studies showed that active p35/CDK5 was mainly localized to the plasma membrane. Specific inhibition of CDK5 in differentiated cultured podocytes, either pharmacologically or with siRNA, induced shape changes, with cellular elongation and loss of process formation compared to the characteristic arborized phenotype. These data suggest a role for CDK5 as a regulator of podocyte differentiation, proliferation, and morphology.
Collapse
Affiliation(s)
- Sian V Griffin
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Box 356521, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Economou CG, Kitsiou PV, Tzinia AK, Panagopoulou E, Marinos E, Kershaw DB, Kerjaschki D, Tsilibary EC. Enhanced podocalyxin expression alters the structure of podocyte basal surface. J Cell Sci 2004; 117:3281-94. [PMID: 15226400 DOI: 10.1242/jcs.01163] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glomerular basement membrane (GBM) and podocalyxin are essential for podocyte morphology. We provide evidence of functional interconnections between basement membrane components (collagen IV and laminin), the expression of podocalyxin and the morphology of human glomerular epithelial cells (podocytes). We demonstrated that GBM and laminin, but not collagen IV, up-regulated the expression of podocalyxin. Scanning electron microscopy revealed that laminin induced a modified morphology of podocytes with process formation, which was more extensive in the presence of GBM. Under high magnification, podocytes appeared ruffled. Using transmission electron microscopy we observed that raised areas occurred in the basal cell surface. Furthermore, the presence of anti-podocalyxin antibody increased the extent of adhesion and spreading of podocytes to both collagen IV and laminin, thus podocalyxin apparently inhibits cell-matrix interactions. We also performed adhesion and spreading assays on podocytes grown under increased glucose concentration (25 mM). Under these conditions, the expression of podocalyxin was almost totally suppressed. The cells adhered and spread to basement membrane components but there was no increase in the extent of adhesion and spreading in the presence of anti-podocalyxin antibody, or ruffling of the cell edges. Additionally, in podocytes expressing podocalyxin, the presence of anti-podocalyxin antibody partially reversed the inhibition of adhesion to collagen IV provoked by anti-β1 integrin antibody, thus podocalyxin should compete with β1-related cell adhesion. We suggest that the observed podocalyxin-mediated inhibition of binding to the matrix could be in part responsible for the specialized conformation of the basal surface of podocytes.
Collapse
MESH Headings
- Amino Acids, Diamino/metabolism
- Animals
- Antibodies, Monoclonal/chemistry
- Basement Membrane/physiology
- Blotting, Northern
- Blotting, Western
- Cell Adhesion
- Cell Proliferation
- Cell Separation
- Cells, Cultured
- Collagen Type IV/metabolism
- DNA, Complementary/metabolism
- Densitometry
- Diabetes Mellitus, Experimental/metabolism
- Epithelial Cells/metabolism
- Flow Cytometry
- Glucose/metabolism
- Humans
- Immunohistochemistry
- Integrin beta1/metabolism
- Laminin/chemistry
- Laminin/metabolism
- Membrane Proteins/metabolism
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Phosphoproteins/metabolism
- Protein Binding
- Proteins/metabolism
- RNA/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/metabolism
- Up-Regulation
- Zonula Occludens-1 Protein
Collapse
Affiliation(s)
- Constantinos G Economou
- Institute of Biology, National Center for Scientific Research, Demokritos, Agia Paraskevi, 15310 Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Srichai MB, Konieczkowski M, Padiyar A, Konieczkowski DJ, Mukherjee A, Hayden PS, Kamat S, El-Meanawy MA, Khan S, Mundel P, Lee SB, Bruggeman LA, Schelling JR, Sedor JR. A WT1 co-regulator controls podocyte phenotype by shuttling between adhesion structures and nucleus. J Biol Chem 2004; 279:14398-408. [PMID: 14736876 DOI: 10.1074/jbc.m314155200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glomerular podocyte differentiation state is critical for filtration barrier function and is regulated by WT1, a zinc finger transcription factor. A yeast two-hybrid assay identified a novel, WT1-interacting protein (WTIP) that maps to human chromosome 19q13.1, a region with genes linked to familial focal segmental glomerulosclerosis. The domain structure of WTIP is similar to the zyxin subfamily of cytosolic LIM domain-containing proteins, which contain three carboxyl-terminal LIM protein-protein interaction domains and a proline-rich, pre-LIM region with a nuclear export signal. Other LIM domain-containing proteins (zyxin and mouse muscle LIM protein) did not interact with WT1 in two-hybrid assays, and WTIP did not interact with an unrelated transcription factor, LMX1B. WTIP mRNA was detected in cultured podocytes and was developmentally regulated, with expression peaking in mouse kidney at embryonic day 15-16 (E15-E16) in kidney but persisting into adulthood. In situ hybridization demonstrated WTIP expression in developing E15 glomeruli and in cultured podocytes. The partial WTIP clone, which interacted with WTIP in the two-hybrid assay, co-localized with WT1 in nuclei, co-precipitated with WT1, and inhibited WT1-dependent transcriptional activation of the amphiregulin promoter. In contrast, full-length WTIP was excluded from cell nuclei, but after the addition of leptomycin B, an inhibitor of Crm1-mediated nuclear export, it accumulated in the nucleus and co-precipitated with WT1 in whole cell lysates. Epitope-tagged WTIP co-localized with the adaptor protein CD2AP (CMS) in podocyte actin spots and with Mena at cell-cell junctions. We propose that WTIP monitors slit diaphragm protein assembly as part of a multiple protein complex, linking this specialized adhesion junction to the actin cytoskeleton, and shuttles into the nucleus after podocyte injury, providing a mechanism whereby changes in slit diaphragm structure modulate gene expression.
Collapse
Affiliation(s)
- Manakan B Srichai
- Departments of Medicine and Physiology and Biophysics, School of Medicine, Case Western Reserve University and Rammelkamp Center for Research and Education, MetroHealth System Campus, Cleveland, Ohio 44109-1998, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The rapidly developing field of podocyte cell biology is reviewed, focusing on papers published in the last 12 months. RECENT FINDINGS Four areas of particular progress can be discerned. First, podocytes proliferate during early metanephric development, are quiescent after the capillary loop stage, and re-enter the cell cycle only in the disease group termed collapsing glomerulopathy. We have learned that control of the podocyte cell cycle involves both expression of cell-cycle regulating proteins and the process of cytokinesis. Second, the podocyte slit diaphragm is the final component of the filtration barrier. The structure and maintenance of the slit diaphragm has been a major focus of research activity, and a multiplicity of relevant molecular interactions have been defined. Significant advances have been made in understanding the complex and interacting role of nephrin and podocin mutations in the genesis of clinical glomerular disease. Third, several proteins essential to controlling discrete podocyte transcriptional programs have been defined. Finally, conditionally-immortalized podocyte cell lines, derived from mouse and human tissue, have proven their worth as models to advance investigations of podocyte biology. SUMMARY Podocyte injury occurs as a consequence of genetic mutation, immunological injury, viral infection, or abnormal hemodynamic forces within the glomerulus. As we understand more about the podocyte proteome and cell biology, we gain an increasingly detailed molecular understanding of podocyte structure and function. In this drama we have many molecular players and increasing stretches of molecular dialogue, but the script remains largely to be deciphered. Nevertheless, we do understand the consequences that arise when the podocyte cannot put its best foot (processes) forward.
Collapse
|
46
|
Brantley JG, Sharma M, Alcalay NI, Heuvel GBV. Cux-1 transgenic mice develop glomerulosclerosis and interstitial fibrosis. Kidney Int 2003; 63:1240-8. [PMID: 12631340 DOI: 10.1046/j.1523-1755.2003.00889.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cux-1 is a murine homeobox gene that is highly expressed in the nephrogenic zone of the developing kidney. Transgenic mice ectopically expressing Cux-1 develop renal hyperplasia associated with down-regulation of the cyclin kinase inhibitor p27. Because the reduction of p27 has been associated with mesangial cell proliferation and glomerular disease, we evaluated glomerular changes in Cux-1 transgenic mice. METHODS Adult kidneys from Cux-1 transgenic mice were analyzed morphologically for changes in glomerular cell number and for changes in mesangial and interstitial extracellular matrix deposition. Mesangial matrix expansion was identified by light microscopy. Glomerular cell number was performed following immunohistochemistry. Type IV collagen deposition was analyzed by immunofluoresence and Western blotting. Renal function was evaluated by serum protein, blood urea nitrogen (BUN), creatinine, and electrolyte analysis, and by urine protein and creatinine analysis. RESULTS In adult transgenic glomeruli, Cux-1 was ectopically expressed in mesangial cells, and this was associated with an increase in mesangial cell number, resulting from an increase in proliferation. There was a marked increase in mesangial matrix area in transgenic mice compared to non-transgenic littermates, related to an increase in type IV collagen. Podocyte foot process effacement was observed in transgenic mice, and this was related to an increase in urinary albumin. Interstitial fibrosis was also observed in transgenic kidneys. CONCLUSION These observations indicate that increased expression of Cux-1 in mesangial cells results in cell proliferation and mesangial expansion. In addition, these changes are potentially related to disruption of podocyte architecture leading to loss of filtration. These results suggest that expression of Cux-1 is sufficient to induce the early events of mesangioproliferative glomerulonephritis.
Collapse
MESH Headings
- Animals
- Cell Division
- Disease Models, Animal
- Female
- Fibrosis
- Glomerulonephritis, Membranoproliferative/genetics
- Glomerulonephritis, Membranoproliferative/pathology
- Glomerulonephritis, Membranoproliferative/physiopathology
- Glomerulosclerosis, Focal Segmental/genetics
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/physiopathology
- Homeodomain Proteins
- Kidney Glomerulus/pathology
- Kidney Glomerulus/physiopathology
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Nuclear Proteins/genetics
- Proteinuria/genetics
- Proteinuria/pathology
- Proteinuria/physiopathology
- Repressor Proteins/genetics
Collapse
Affiliation(s)
- Jennifer G Brantley
- Department of Anatomy and Cell Biology and University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|