1
|
Ali A, Liu Z, Ye K, Guan Y, Chen S, Liu T, Guo Z, Wong MK, Vasquez P, Poudel C, Mustonen BC, Eng DG, Pippin JW, Shankland SJ, Wang S, Vaughan JC. Nanoscale Optical Imaging, Reconstruction, and Spatial Analysis of Whole Mouse Glomeruli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.620364. [PMID: 39554089 PMCID: PMC11565967 DOI: 10.1101/2024.10.31.620364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Renal glomeruli have traditionally been studied by micrometer-scale optical microscopy to interrogate overall physiology or molecular distributions and by nanoscale electron microscopy to interrogate the ultrastructure of thin sections. While these approaches are powerful, they have been limited in their ability to obtain detailed views of the glomeruli as holistic 3D functional units. To fill this knowledge gap, we have developed a novel pipeline for imaging, reconstructing, and analyzing whole mouse glomeruli at 100 nm resolution using super-resolution fluorescence microscopy. This pipeline integrates both manual and machine learning approaches to annotate and analyze glomerular structures. Using this method, we created 18 detailed glomerulus models, from a range of healthy, aged, and model diseased mice, that outline all major structures and cell types. These models have been made publicly accessible in an online repository, providing a valuable resource for further studies. Our results also uncovered a diverse set of novel phenotypes including nuclear enlargement in all glomerular cell types in aging and disease, as well as an aging-related pattern of regional thickening of the Bowman's capsule basement membrane near the tubular-glomerular junction.
Collapse
Affiliation(s)
- Adilijiang Ali
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Zixuan Liu
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, Washington, USA
| | - Kenan Ye
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, Washington, USA
| | - Yun Guan
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Siying Chen
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Tingxuan Liu
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Ziyu Guo
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Madeline K Wong
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Pedro Vasquez
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | | | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Sheng Wang
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, Washington, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, Washington, USA
- Department of Neurobiology and Biophysics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Wu C, Tang H, Cui X, Li N, Fei J, Ge H, Wu L, Wu J, Gu HF. A single-cell profile reveals the transcriptional regulation responded for Abelmoschus manihot (L.) treatment in diabetic kidney disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155642. [PMID: 38759315 DOI: 10.1016/j.phymed.2024.155642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Huangkui capsule (HKC), as an ethanol extract of Abelmoschus manihot (L.), has a significant efficacy in treatment of the patients with diabetic kidney disease (DKD). The bioactive ingredients of HKC mainly include the flavonoids such as rutin, hyperoside, hibifolin, isoquercetin, myricetin, quercetin and quercetin-3-O-robinobioside. PURPOSE To explore the molecular mechanisms of A. manihot in treatment of DKD. STUDY DESIGN A single-cell RNA sequencing analysis of kidneys in db/db mice with and without HKC administration. METHODS Urinary biochemical and histopathological examination in C57BL/6 and db/db mice of DKD and HKC groups was done. Single-cell RNA sequencing pipeline was then performed. The regulatory mechanisms of seven flavonoids in HKC were revealed by cell communication, prediction of transcription factor regulatory network, and molecular docking. RESULTS By constructing ligand-receptor regulatory network and performing molecular docking between 75 receptors with different activities and seven flavonoids. 11 key receptors in 4 cell types (segment 3 proximal convoluted tubular cell, ascending limbs of the loop of Henle, distal convoluted tubule, and T cell) in kidneys were found to be directly interacted with HKC. The interactions regulated 8 downstream regulons. The docking receptors in T cell led to transcriptional event differences in the regulons such as Cebpb, Rel, Tbx21 and Klf2 and consequently affected the activation, differentiation, and infiltration of T cell, while the receptors Tgfbr1 and Ldlr in stromal cells of kidneys were closely associated with the downstream transcriptional events of renal injury and proteinuria in DKD. CONCLUSION The current study provides novel information of the key receptors and regulons in renal cells for a better understanding of the cell type specific molecular mechanisms of A. manihot in treatment of DKD.
Collapse
Affiliation(s)
- Chenhua Wu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China; Laboratory of Minigene Pharmacy, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, China
| | - Haitao Tang
- Suzhong Pharmaceutical Research Institute, Nanjing, Jiangsu Province, 210018, China
| | - Xu Cui
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Nan Li
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210029, China
| | - Jingjin Fei
- Laboratory of Minigene Pharmacy, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, China
| | - Haitao Ge
- Suzhong Pharmaceutical Research Institute, Nanjing, Jiangsu Province, 210018, China
| | - Liang Wu
- Jiangsu Key Laboratory of Drug Screening, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jie Wu
- Laboratory of Minigene Pharmacy, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, China.
| | - Harvest F Gu
- Laboratory of Molecular Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China.
| |
Collapse
|
3
|
Cruzado JM, Manonelles A, Rayego-Mateos S, Doladé N, Amaya-Garrido A, Varela C, Guiteras R, Mosquera JL, Jung M, Codina S, Martínez-Valenzuela L, Draibe J, Couceiro C, Vigués F, Madrid Á, Florian MC, Ruíz-Ortega M, Sola A. Colony stimulating factor-1 receptor drives glomerular parietal epithelial cell activation in focal segmental glomerulosclerosis. Kidney Int 2024; 106:67-84. [PMID: 38428734 DOI: 10.1016/j.kint.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
Parietal epithelial cells (PECs) are kidney progenitor cells with similarities to a bone marrow stem cell niche. In focal segmental glomerulosclerosis (FSGS) PECs become activated and contribute to extracellular matrix deposition. Colony stimulating factor-1 (CSF-1), a hematopoietic growth factor, acts via its specific receptor, CSF-1R, and has been implicated in several glomerular diseases, although its role on PEC activation is unknown. Here, we found that CSF-1R was upregulated in PECs and podocytes in biopsies from patients with FSGS. Through in vitro studies, PECs were found to constitutively express CSF-1R. Incubation with CSF-1 induced CSF-1R upregulation and significant transcriptional regulation of genes involved in pathways associated with PEC activation. Specifically, CSF-1/CSF-1R activated the ERK1/2 signaling pathway and upregulated CD44 in PECs, while both ERK and CSF-1R inhibitors reduced CD44 expression. Functional studies showed that CSF-1 induced PEC proliferation and migration, while reducing the differentiation of PECs into podocytes. These results were validated in the Adriamycin-induced FSGS experimental mouse model. Importantly, treatment with either the CSF-1R-specific inhibitor GW2580 or Ki20227 provided a robust therapeutic effect. Thus, we provide evidence of the role of the CSF-1/CSF-1R pathway in PEC activation in FSGS, paving the way for future clinical studies investigating the therapeutic effect of CSF-1R inhibitors on patients with FSGS.
Collapse
Affiliation(s)
- Josep M Cruzado
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Department of Nephrology, Hospital Universitari Bellvitge, Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Anna Manonelles
- Department of Nephrology, Hospital Universitari Bellvitge, Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
| | - Núria Doladé
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Ana Amaya-Garrido
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Cristian Varela
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Roser Guiteras
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Jose Luis Mosquera
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Sergi Codina
- Department of Nephrology, Hospital Universitari Bellvitge, Barcelona, Spain
| | | | - Juliana Draibe
- Department of Nephrology, Hospital Universitari Bellvitge, Barcelona, Spain
| | - Carlos Couceiro
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Department of Nephrology, Hospital Universitari Bellvitge, Barcelona, Spain
| | - Francesc Vigués
- Department of Urology, Hospital Universitari Bellvitge, Barcelona, Spain
| | - Álvaro Madrid
- Pediatric Nephrology Department, Sant Joan de Deu University Hospital, Barcelona, Spain
| | - M Carolina Florian
- Program of Regenerative Medicine, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain; Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; The Catalan Institution for Research and Advanced Studies (ICREA)
| | - Marta Ruíz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
| | - Anna Sola
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
4
|
Veloso Pereira BM, Zeng Y, Maggiore JC, Schweickart RA, Eng DG, Kaverina N, McKinzie SR, Chang A, Loretz CJ, Thieme K, Hukriede NA, Pippin JW, Wessely O, Shankland SJ. Podocyte injury at young age causes premature senescence and worsens glomerular aging. Am J Physiol Renal Physiol 2024; 326:F120-F134. [PMID: 37855038 PMCID: PMC11198990 DOI: 10.1152/ajprenal.00261.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023] Open
Abstract
As life expectancy continues to rise, age-related diseases are becoming more prevalent. For example, proteinuric glomerular diseases typified by podocyte injury have worse outcomes in the elderly compared with young patients. However, the reasons are not well understood. We hypothesized that injury to nonaged podocytes induces senescence, which in turn augments their aging processes. In primary cultured human podocytes, injury induced by a cytopathic antipodocyte antibody, adriamycin, or puromycin aminonucleoside increased the senescence-related genes CDKN2A (p16INK4a/p14ARF), CDKN2D (p19INK4d), and CDKN1A (p21). Podocyte injury in human kidney organoids was accompanied by increased expression of CDKN2A, CDKN2D, and CDKN1A. In young mice, experimental focal segmental glomerulosclerosis (FSGS) induced by adriamycin and antipodocyte antibody increased the glomerular expression of p16, p21, and senescence-associated β-galactosidase (SA-β-gal). To assess the long-term effects of early podocyte injury-induced senescence, we temporally followed young mice with experimental FSGS through adulthood (12 m of age) and middle age (18 m of age). p16 and Sudan black staining were higher at middle age in mice with earlier FSGS compared with age-matched mice that did not get FSGS when young. This was accompanied by lower podocyte density, reduced canonical podocyte protein expression, and increased glomerular scarring. These results are consistent with injury-induced senescence in young podocytes, leading to increased senescence of podocytes by middle age accompanied by lower podocyte lifespan and health span.NEW & NOTEWORTHY Glomerular function is decreased by aging. However, little is known about the molecular mechanisms involved in age-related glomerular changes and which factors could contribute to a worse glomerular aging process. Here, we reported that podocyte injury in young mice and culture podocytes induced senescence, a marker of aging, and accelerates glomerular aging when compared with healthy aging mice.
Collapse
Affiliation(s)
- Beatriz Maria Veloso Pereira
- Division of Nephrology, University of Washington, Seattle, Washington, United States
- Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | - Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, Washington, United States
| | - Joseph C Maggiore
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | | | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington, United States
| | - Natalya Kaverina
- Division of Nephrology, University of Washington, Seattle, Washington, United States
| | - Sierra R McKinzie
- Division of Nephrology, University of Washington, Seattle, Washington, United States
| | - Anthony Chang
- Department of Pathology, University of Chicago, Chicago, Illinois, United States
| | - Carol J Loretz
- Division of Nephrology, University of Washington, Seattle, Washington, United States
| | - Karina Thieme
- Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | - Neil A Hukriede
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, United States
| | - Oliver Wessely
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
5
|
Bronstein R, Pace J, Gowthaman Y, Salant DJ, Mallipattu SK. Podocyte-Parietal Epithelial Cell Interdependence in Glomerular Development and Disease. J Am Soc Nephrol 2023; 34:737-750. [PMID: 36800545 PMCID: PMC10125654 DOI: 10.1681/asn.0000000000000104] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/04/2023] [Indexed: 02/19/2023] Open
Abstract
Podocytes and parietal epithelial cells (PECs) are among the few principal cell types within the kidney glomerulus, the former serving as a crucial constituent of the kidney filtration barrier and the latter representing a supporting epithelial layer that adorns the inner wall of Bowman's capsule. Podocytes and PECs share a circumscript developmental lineage that only begins to diverge during the S-shaped body stage of nephron formation-occurring immediately before the emergence of the fully mature nephron. These two cell types, therefore, share a highly conserved gene expression program, evidenced by recently discovered intermediate cell types occupying a distinct spatiotemporal gene expression zone between podocytes and PECs. In addition to their homeostatic functions, podocytes and PECs also have roles in kidney pathogenesis. Rapid podocyte loss in diseases, such as rapidly progressive GN and collapsing and cellular subtypes of FSGS, is closely allied with PEC proliferation and migration toward the capillary tuft, resulting in the formation of crescents and pseudocrescents. PECs are thought to contribute to disease progression and severity, and the interdependence between these two cell types during development and in various manifestations of kidney pathology is the primary focus of this review.
Collapse
Affiliation(s)
- Robert Bronstein
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Jesse Pace
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Yogesh Gowthaman
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - David J. Salant
- Division of Nephrology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Sandeep K. Mallipattu
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
- Renal Section, Northport VA Medical Center, Northport, New York
| |
Collapse
|
6
|
Chebotareva N, Vinogradov A, Tsoy L, Varshavskiy V, Stoljarevich E, Bugrova A, Lerner Y, Krasnova T, Biryukova E, Kononikhin AS. CD44 Expression in Renal Tissue Is Associated with an Increase in Urinary Levels of Complement Components in Chronic Glomerulopathies. Int J Mol Sci 2023; 24:ijms24087190. [PMID: 37108355 PMCID: PMC10138917 DOI: 10.3390/ijms24087190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
It is suggested that activated CD44+ cells play a profibrogenic role in the pathogenesis of active glomerulopathies. Complement activation is also involved in renal fibrogenesis. The aim of the study was to evaluate the role of the activation of CD44+ cells in the kidney tissue and complement components' filtration to the urine as factors of renal tissue fibrosis in patients with glomerulopathies. In total, 60 patients with active glomerulopathies were included in our study: 29 patients with focal segmental glomerulosclerosis (FSGS), 10 patients with minimal change disease (MCD), 10 patients with membranous nephropathy (MN), and 11 patients with IgA nephropathy. The immunohistochemical peroxidase method was used to study the expression of CD44+ in kidney biopsies. Components of complement were analyzed in urine by the multiple reaction monitoring (MRM) approach using liquid chromatography. Strong CD44 expression was noted predominantly in PEC and mesangial cells (MC) in patients with FSGS, and to a lesser extent, in patients with MN and IgA nephropathy, and it was absent in patients with MCD. Expression of profibrogenic CD44+ in glomeruli correlated with the levels of proteinuria and complement C2, C3, and C9 components, and CFB and CFI in urine. The CD44+ expression scores in the renal interstitium correlated with the level of C3 and C9 components of complement in the urine and the area of tubulo-interstitial fibrosis. The strongest expression of CD44+ was found in the glomeruli (MC, PEC, and podocytes) of patients with FSGS compared with other glomerulopathies. The CD44 expression score in the glomeruli and interstitium is associated with high levels of complement components in the urine and renal fibrosis.
Collapse
Affiliation(s)
- Natalia Chebotareva
- Department of Nephrology, Sechenov First Moscow State Medical University, Trubezkaya, 8, 119048 Moscow, Russia
| | - Anatoliy Vinogradov
- Institute for Clinical Morphology and Digital Patology, Sechenov First Moscow State Medical University, Trubezkaya, 8, 119048 Moscow, Russia
| | - Larisa Tsoy
- Department of Internal Medicine, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 119991 Moscow, Russia
| | - Vladimir Varshavskiy
- Department of Internal Medicine, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 119991 Moscow, Russia
| | - Ekaterina Stoljarevich
- Morphology Department, Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya Str., 20, 127473 Moscow, Russia
| | - Anna Bugrova
- Emanuel Institute for Biochemical Physics, Russian Academy of Science, Kosygina Str., 4, 119334 Moscow, Russia
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, 121205 Moscow, Russia
| | - Yulia Lerner
- Department of Internal Medicine, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 119991 Moscow, Russia
| | - Tatyana Krasnova
- Institute for Clinical Morphology and Digital Patology, Sechenov First Moscow State Medical University, Trubezkaya, 8, 119048 Moscow, Russia
| | - Evgeniya Biryukova
- Department of Nephrology, Sechenov First Moscow State Medical University, Trubezkaya, 8, 119048 Moscow, Russia
| | - Alexey S Kononikhin
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, 121205 Moscow, Russia
| |
Collapse
|
7
|
Bhargava R, Li H, Tsokos GC. Pathogenesis of lupus nephritis: the contribution of immune and kidney resident cells. Curr Opin Rheumatol 2023; 35:107-116. [PMID: 35797522 DOI: 10.1097/bor.0000000000000887] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Lupus nephritis is associated with significant mortality and morbidity. We lack effective therapeutics and biomarkers mostly because of our limited understanding of its complex pathogenesis. We aim to present an overview of the recent advances in the field to gain a deeper understanding of the underlying cellular and molecular mechanisms involved in lupus nephritis pathogenesis. RECENT FINDINGS Recent studies have identified distinct roles for each resident kidney cell in the pathogenesis of lupus nephritis. Podocytes share many elements of innate and adaptive immune cells and they can present antigens and participate in the formation of crescents in coordination with parietal epithelial cells. Mesangial cells produce pro-inflammatory cytokines and secrete extracellular matrix contributing to glomerular fibrosis. Tubular epithelial cells modulate the milieu of the interstitium to promote T cell infiltration and formation of tertiary lymphoid organs. Modulation of specific genes in kidney resident cells can ward off the effectors of the autoimmune response including autoantibodies, cytokines and immune cells. SUMMARY The development of lupus nephritis is multifactorial involving genetic susceptibility, environmental triggers and systemic inflammation. However, the role of resident kidney cells in the development of lupus nephritis is becoming more defined and distinct. More recent studies point to the restoration of kidney resident cell function using cell targeted approaches to prevent and treat lupus nephritis.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard, Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
8
|
Huang Y, Zhao X, Zhang Q, Yang X, Hou G, Peng C, Jia M, Zhou L, Yamamoto T, Zheng J. Novel therapeutic perspectives for crescentic glomerulonephritis through targeting parietal epithelial cell activation and proliferation. Expert Opin Ther Targets 2023; 27:55-69. [PMID: 36738160 DOI: 10.1080/14728222.2023.2177534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Kidney injury is clinically classified as crescentic glomerulonephritis (CrGN) when ≥50% of the glomeruli in a biopsy sample contain crescentic lesions. However, current strategies, such as systemic immunosuppressive therapy and plasmapheresis for CrGN, are partially effective, and these drugs have considerable systemic side effects. Hence, targeted therapy to prevent glomerular crescent formation and expansion remains an unmet clinical need. AREAS COVERED Hyperproliferative parietal epithelial cells (PECs) are the main constituent cells of the glomerular crescent with cell-tracing evidence. Crescents obstruct the flow of primary urine, pressure the capillaries, and degenerate the affected nephrons. We reviewed the markers of PEC activation and proliferation, potential therapeutic effects of thrombin and thrombin receptor inhibitors, and how podocytes cross-talk with PECs. These experiments may help identify potential early specific targets for the prevention and treatment of glomerular crescentic injury. EXPERT OPINION Inhibiting PEC activation and proliferation in CrGN can alleviate glomerular crescent progression, which has been supported by preclinical studies with evidence of genetic deletion. Clarifying the outcome of PEC transformation to the podocyte phenotype and suppressing thrombin, thrombin receptors, and PEC hyperproliferation in early therapeutic strategies will be the research goals in the next ten years.
Collapse
Affiliation(s)
- Yanjie Huang
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China.,Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xueru Zhao
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qiushuang Zhang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoqing Yang
- Department of Pediatrics, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Gailing Hou
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Chaoqun Peng
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mengzhen Jia
- School of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Li Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tatsuo Yamamoto
- Department of Nephrology, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda, Japan
| | - Jian Zheng
- Institute of Pediatrics of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Melica ME, Antonelli G, Semeraro R, Angelotti ML, Lugli G, Landini S, Ravaglia F, La Regina G, Conte C, De Chiara L, Peired AJ, Mazzinghi B, Donati M, Molli A, Steiger S, Magi A, Bartalucci N, Raglianti V, Guzzi F, Maggi L, Annunziato F, Burger A, Lazzeri E, Anders HJ, Lasagni L, Romagnani P. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice. Sci Transl Med 2022; 14:eabg3277. [PMID: 35947676 PMCID: PMC7614034 DOI: 10.1126/scitranslmed.abg3277] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Crescentic glomerulonephritis is characterized by vascular necrosis and parietal epithelial cell hyperplasia in the space surrounding the glomerulus, resulting in the formation of crescents. Little is known about the molecular mechanisms driving this process. Inducing crescentic glomerulonephritis in two Pax2Cre reporter mouse models revealed that crescents derive from clonal expansion of single immature parietal epithelial cells. Preemptive and delayed histone deacetylase inhibition with panobinostat, a drug used to treat hematopoietic stem cell disorders, attenuated crescentic glomerulonephritis with recovery of kidney function in the two mouse models. Three-dimensional confocal microscopy and stimulated emission depletion superresolution imaging of mouse glomeruli showed that, in addition to exerting an anti-inflammatory and immunosuppressive effect, panobinostat induced differentiation of an immature hyperplastic parietal epithelial cell subset into podocytes, thereby restoring the glomerular filtration barrier. Single-cell RNA sequencing of human renal progenitor cells in vitro identified an immature stratifin-positive cell subset and revealed that expansion of this stratifin-expressing progenitor cell subset was associated with a poor outcome in human crescentic glomerulonephritis. Treatment of human parietal epithelial cells in vitro with panobinostat attenuated stratifin expression in renal progenitor cells, reduced their proliferation, and promoted their differentiation into podocytes. These results offer mechanistic insights into the formation of glomerular crescents and demonstrate that selective targeting of renal progenitor cells can attenuate crescent formation and the deterioration of kidney function in crescentic glomerulonephritis in mice.
Collapse
Affiliation(s)
- Maria Elena Melica
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Giulia Antonelli
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gianmarco Lugli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Samuela Landini
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Fiammetta Ravaglia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Carolina Conte
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Marta Donati
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Alice Molli
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Stefanie Steiger
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence 50139, Italy
| | - Valentina Raglianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Corresponding authors. and
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy,Corresponding authors. and
| |
Collapse
|
10
|
Li ZH, Guo XY, Quan XY, Yang C, Liu ZJ, Su HY, An N, Liu HF. The Role of Parietal Epithelial Cells in the Pathogenesis of Podocytopathy. Front Physiol 2022; 13:832772. [PMID: 35360248 PMCID: PMC8963495 DOI: 10.3389/fphys.2022.832772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 02/05/2023] Open
Abstract
Podocytopathy is the most common feature of glomerular disorder characterized by podocyte injury- or dysfunction-induced excessive proteinuria, which ultimately develops into glomerulosclerosis and results in persistent loss of renal function. Due to the lack of self-renewal ability of podocytes, mild podocyte depletion triggers replacement and repair processes mostly driven by stem cells or resident parietal epithelial cells (PECs). In contrast, when podocyte recovery fails, activated PECs contribute to the establishment of glomerular lesions. Increasing evidence suggests that PECs, more than just bystanders, have a crucial role in various podocytopathies, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, diabetic nephropathy, IgA nephropathy, and lupus podocytopathy. In this review, we attempt to dissect the diverse role of PECs in the pathogenesis of podocytopathy based on currently available information.
Collapse
|
11
|
Ye L, Liu Y, Zhu X, Duan T, Wang C, Fu X, Song P, Yuan S, Liu H, Sun L, Liu F, Lee K, He JC, Chen A. Digital Spatial Profiling of Individual Glomeruli From Patients With Anti-Neutrophil Cytoplasmic Autoantibody-Associated Glomerulonephritis. Front Immunol 2022; 13:831253. [PMID: 35309370 PMCID: PMC8924137 DOI: 10.3389/fimmu.2022.831253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/31/2022] [Indexed: 11/29/2022] Open
Abstract
We previously showed that the rupture of Bowman's capsule (BC) promotes the progression of crescentic glomerulonephritis by enhancing the entry of CD8+ T cells into the glomeruli. In the present study, we utilized digital spatial profiling to simultaneously profile the altered abundances of the messenger RNA (mRNA) transcripts and proteins in the glomerular and periglomerular areas of four biopsy samples of anti-neutrophil cytoplasmic autoantibody-associated glomerulonephritis (ANCA-GN) and two biopsy specimens of minimal change disease (MCD) controls. The paraffin-embedded biopsy samples were stained with collagen IV, CD45, and SYTO 13 to distinguish the glomeruli with periglomerular infiltration but intact BC, with focal BC rupture, and with extensive rupture of BC and glomeruli without crescent formation and leukocytic infiltration in ANCA-GN. By assessing multiple discrete glomerular areas, we found that the transcript expression levels of the secreted phosphoprotein-1 and its receptor CD44 were upregulated significantly in the glomeruli with more severe ruptures of BC, and their expression levels correlated positively with the fibrotic markers. We also found that both alternative and classic complement pathways were activated in the glomeruli from patients with ANCA-GN. Furthermore, M1 macrophages were involved mostly in the early stage of BC rupture, while M2 macrophages were involved in the late stage and may contribute to the fibrosis process of the crescents. Finally, loss of glomerular cells in ANCA-GN was likely mediated by apoptosis. Our results show that digital spatial profiling allows the comparative analysis of the mRNA and protein profiles in individual glomeruli affected differently by the disease process and the identification of potential novel mechanisms in ANCA-GN.
Collapse
Affiliation(s)
- Lin Ye
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Xuejing Zhu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Tongyue Duan
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Chang Wang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Xiao Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Panai Song
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Shuguang Yuan
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Hong Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Lin Sun
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Fuyou Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, New York, NY, United States
| | - Anqun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| |
Collapse
|
12
|
Cai J, Zhang X, Xie W, Li Z, Liu W, Liu A. Identification of a basement membrane-related gene signature for predicting prognosis and estimating the tumor immune microenvironment in breast cancer. Front Endocrinol (Lausanne) 2022; 13:1065530. [PMID: 36531485 PMCID: PMC9751030 DOI: 10.3389/fendo.2022.1065530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION Breast cancer (BC) is the most common malignancy in the world and has a high cancer-related mortality rate. Basement membranes (BMs) guide cell polarity, differentiation, migration and survival, and their functions are closely related to tumor diseases. However, few studies have focused on the association of basement membrane-related genes (BMRGs) with BC. This study aimed to explore the prognostic features of BMRGs in BC and provide new directions for the prevention and treatment of BC. METHODS We collected transcriptomic and clinical data of BC patients from TCGA and GEO datasets and constructed a predictive signature for BMRGs by using univariate, least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analysis. The reliability of the model was further evaluated and validated by Kaplan-Meier survival curves and receiver operating characteristic curves (ROC). Column line plots and corresponding calibration curves were constructed. Possible biological pathways were investigated by enrichment analysis. Afterward, we assessed the mutation status by tumor mutational burden (TMB) analysis and compared different subtypes using cluster analysis. Finally, we examined drug treatment sensitivity and immunological correlation to lay the groundwork for more in-depth studies in this area. RESULTS The prognostic risk model consisted of 7 genes (FBLN5, ITGB2, LAMC3, MMP1, EVA1B, SDC1, UNC5A). After validation, we found that the model was highly reliable and could accurately predict the prognosis of BC patients. Cluster analysis showed that patients with cluster 1 had more sensitive drugs and had better chances of better clinical outcomes. In addition, TMB, immune checkpoint, immune status, and semi-inhibitory concentrations were significantly different between high and low-risk groups, with lower-risk patients having the better anti-cancer ability. DISCUSSION The basement membrane-related gene signature that we established can be applied as an independent prognostic factor for BC and can provide a reference for individualized treatment of BC patients.
Collapse
Affiliation(s)
- Jiehui Cai
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xinkang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wanchun Xie
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhiyang Li
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wei Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang, Hunan, China
| | - An Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang, Hunan, China
- *Correspondence: An Liu,
| |
Collapse
|
13
|
Hudkins KL, Li X, Holland AL, Swaminathan S, Alpers CE. Regression of diabetic nephropathy by treatment with empagliflozin in BTBR ob/ob mice. Nephrol Dial Transplant 2021; 37:847-859. [PMID: 34865099 DOI: 10.1093/ndt/gfab330] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The SGLT2 inhibitor empagliflozin lowers blood glucose via reduced tubular reabsorption of filtered glucose and is an important new therapy for diabetic nephropathy (DN). This study tested whether treatment with empagliflozin would ameliorate proteinuria and the pathologic alterations of DN including podocyte number and integrity in the leptin deficient BTBR ob/ob mouse model of DN. METHODS Study cohorts included wild type BTBR mice, untreated diabetic BTBR ob/ob mice, and mice treated with empagliflozin for six weeks after development of established DN at 18 weeks of age. RESULTS Hyperglycemia, proteinuria, serum creatinine, accumulation of mesangial matrix and the extent of mesangiolysis were reversed with empagliflozin treatment. Treatment with empagliflozin resulted in increased podocyte number and podocyte density, improvement in the degree of podocyte foot process effacement and parietal epithelial cell activation. SGLT2 inhibition reduced renal oxidative stress, measured by urinary excretion of markers of RNA/DNA damage and in situ demonstration of decreased carbonyl oxidation. There was no discernable difference in accumulations of advanced glycation endproducts by immunohistochemistry. CONCLUSION The structural improvements seen in BTBR ob/ob mice treated with empagliflozin provide insight into potential long term benefits for humans with DN, for whom there is no comparable biopsy information to identify structural changes effected by SGLT2 inhibition. The findings suggest SGLT2 inhibition may ameliorate diabetic nephropathy through glucose lowering-dependent and -independent mechanisms that lead to podocyte restoration and delay or reversal of the disease progress.
Collapse
Affiliation(s)
- Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle WA, USA
| | - Xianwu Li
- Department of Pathology, University of Washington, Seattle WA, USA
| | | | | | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle WA, USA
| |
Collapse
|
14
|
Parietal epithelial cell dysfunction in crescentic glomerulonephritis. Cell Tissue Res 2021; 385:345-354. [PMID: 34453566 PMCID: PMC8523405 DOI: 10.1007/s00441-021-03513-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Crescentic glomerulonephritis represents a group of kidney diseases characterized by rapid loss of kidney function and the formation of glomerular crescents. While the role of the immune system has been extensively studied in relation to the development of crescents, recent findings show that parietal epithelial cells play a key role in the pathophysiology of crescent formation, even in the absence of immune modulation. This review highlights our current understanding of parietal epithelial cell biology and the reported physiological and pathological roles that these cells play in glomerular lesion formation, especially in the context of crescentic glomerulonephritis.
Collapse
|
15
|
Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis. Clin Sci (Lond) 2021; 135:1999-2029. [PMID: 34427291 DOI: 10.1042/cs20201016] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD.
Collapse
|
16
|
Barutta F, Kimura S, Hase K, Bellini S, Corbetta B, Corbelli A, Fiordaliso F, Barreca A, Papotti MG, Ghiggeri GM, Salvidio G, Roccatello D, Audrito V, Deaglio S, Gambino R, Bruno S, Camussi G, Martini M, Hirsch E, Durazzo M, Ohno H, Gruden G. Protective Role of the M-Sec-Tunneling Nanotube System in Podocytes. J Am Soc Nephrol 2021; 32:1114-1130. [PMID: 33722931 PMCID: PMC8259684 DOI: 10.1681/asn.2020071076] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 01/21/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocyte dysfunction and loss are major determinants in the development of proteinuria. FSGS is one of the most common causes of proteinuria, but the mechanisms leading to podocyte injury or conferring protection against FSGS remain poorly understood. The cytosolic protein M-Sec has been involved in the formation of tunneling nanotubes (TNTs), membrane channels that transiently connect cells and allow intercellular organelle transfer. Whether podocytes express M-Sec is unknown and the potential relevance of the M-Sec-TNT system in FSGS has not been explored. METHODS We studied the role of the M-Sec-TNT system in cultured podocytes exposed to Adriamycin and in BALB/c M-Sec knockout mice. We also assessed M-Sec expression in both kidney biopsies from patients with FSGS and in experimental FSGS (Adriamycin-induced nephropathy). RESULTS Podocytes can form TNTs in a M-Sec-dependent manner. Consistent with the notion that the M-Sec-TNT system is cytoprotective, podocytes overexpressed M-Sec in both human and experimental FSGS. Moreover, M-Sec deletion resulted in podocyte injury, with mitochondrial abnormalities and development of progressive FSGS. In vitro, M-Sec deletion abolished TNT-mediated mitochondria transfer between podocytes and altered mitochondrial bioenergetics. Re-expression of M-Sec reestablishes TNT formation and mitochondria exchange, rescued mitochondrial function, and partially reverted podocyte injury. CONCLUSIONS These findings indicate that the M-Sec-TNT system plays an important protective role in the glomeruli by rescuing podocytes via mitochondrial horizontal transfer. M-Sec may represent a promising therapeutic target in FSGS, and evidence that podocytes can be rescued via TNT-mediated horizontal transfer may open new avenues of research.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Alessandro Corbelli
- Department of Cardiovascular Medicine, Institute of Pharmacological Research Mario Negri, Scientific Institute for Hospitalization and Care (IRCCS), Milan, Italy
| | - Fabio Fiordaliso
- Department of Cardiovascular Medicine, Institute of Pharmacological Research Mario Negri, Scientific Institute for Hospitalization and Care (IRCCS), Milan, Italy
| | | | | | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis, Transplantation, Gaslini Children’s Hospital, Genoa, Italy
| | - Gennaro Salvidio
- Scientific Institute for Hospitalization and Care (IRCCS), San Martino University Hospital Clinic, Genoa, Italy
| | - Dario Roccatello
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d’Aosta Network for Rare Diseases, S. Giovanni Bosco Hospital, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy,Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | | | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
17
|
Metformin ameliorates the severity of experimental Alport syndrome. Sci Rep 2021; 11:7053. [PMID: 33782421 PMCID: PMC8007696 DOI: 10.1038/s41598-021-86109-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/05/2021] [Indexed: 12/24/2022] Open
Abstract
Metformin is widely used for the treatment of type 2 diabetes, and increasing numbers of studies have shown that metformin also ameliorates tumor progression, inflammatory disease, and fibrosis. However, the ability of metformin to improve non-diabetic glomerular disease and chronic kidney disease (CKD) has not been explored. To investigate the effect of metformin on non-diabetic glomerular disease, we used a mouse model of Alport syndrome (Col4a5 G5X) which were treated with metformin or losartan, used as a control treatment. We also investigated the effect of metformin on adriamycin-induced glomerulosclerosis model. Pathological and biochemical analysis showed that metformin or losartan suppressed proteinuria, renal inflammation, fibrosis, and glomerular injury and extended the lifespan in Alport syndrome mice. Transcriptome analysis showed that metformin and losartan influenced molecular pathways-related to metabolism and inflammation. Metformin altered multiple genes including metabolic genes not affected by losartan. Metformin also suppressed proteinuria and glomerular injury in the adriamycin-induced glomerulosclerosis mouse model. Our results showed that metformin ameliorates the glomerular sclerosis and CKD phenotype in non-diabetic chronic glomerular diseases. Metformin may have therapeutic potential for not only diabetic nephropathy but also non-diabetic glomerular disease including Alport syndrome.
Collapse
|
18
|
Parietal epithelial cells role in repair versus scarring after glomerular injury. Curr Opin Nephrol Hypertens 2021; 29:293-301. [PMID: 32235272 DOI: 10.1097/mnh.0000000000000600] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW The recent years have been marked by the publication of several articles highlighting the pathophysiological role of glomerular parietal epithelial cells (PEC) and refining their phenotypic heterogeneity. RECENT FINDINGS The present review synthetizes recent findings on (i) the potential regenerative role of PEC in glomerular diseases, and (ii) the mechanisms and signaling of leading to PEC pathogenic involvement in crescentic glomerulonephritis (CGN) and focal segmental glomerulosclerosis (FSGS). SUMMARY The debate is still open regarding the podocyte regenerative properties of PEC in glomerular disease, whereas the pathogenic involvement of PEC activation in glomerular disease is increasingly admitted. Recent highlights on the podocyte regenerative role of PEC, on one hand, and on their pathological function, on the other hand, for sure will feed the debate in the kidney community for the next years. Nevertheless, from a therapeutic perspective, the two options, boosting cellular regeneration and blocking PECs pathogenicity, should not be seen as antagonistic but, rather, complementary.
Collapse
|
19
|
Aslam R, Hussain A, Cheng K, Kumar V, Malhotra A, Gupta S, Singhal PC. Transplantation of mesenchymal stem cells preserves podocyte homeostasis through modulation of parietal epithelial cell activation in adriamycin-induced mouse kidney injury model. Histol Histopathol 2020; 35:1483-1492. [PMID: 33124682 DOI: 10.14670/hh-18-276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To determine the role of the transplantation of bone marrow-derived mesenchymal stem cells (MSCs) in podocyte renewal, we studied BALB/C mice with or without adriamycin-induced acute kidney injury. MSCs were transplanted ectopically under the capsule of the left kidney or into the peritoneal cavity after the onset of kidney injury to test testing their local or systemic paracrine effects, respectively. Adriamycin produced increases in urine protein: creatinine ratios, blood urea nitrogen, and blood pressure, which improved after both renal subcapsular and intraperitoneal MSCs transplants. The histological changes of adriamycin kidney changes regressed in both kidneys and in only the ipsilateral kidney after intraperitoneal or renal subcapsular transplants indicating that the benefits of transplanted MSCs were related to the extent of paracrine factor distribution. Analysis of kidney tissues for p57-positive parietal epithelial cells (PECs) showed that MSC transplants restored adriamycin-induced decreases in the abundance of these cells to normal levels, although after renal subcapsular transplants these changes did not extend to contralateral kidneys. Moreover, adriamycin caused inflammatory activation of PECs with coexpression of CD44 and phospho-ERK, which was normalized in both or only ipsilateral kidneys depending on whether MSCs were transplanted in the peritoneal cavity or subcapsular space, respectively.
Collapse
Affiliation(s)
- Rukhsana Aslam
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Ali Hussain
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Kang Cheng
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Vinod Kumar
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Ashwani Malhotra
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Sanjeev Gupta
- Department of Medicine, Department of Pathology, Marion Bessin Liver Research Center, Diabetes Center, The Irwin S. and Sylvia Chanin Institute for Cancer Research, and Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA
| | - Pravin C Singhal
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA.
| |
Collapse
|
20
|
Ye C, Xiong W, Lei CT, Tang H, Su H, Yi F, Zhang C. MAD2B contributes to parietal epithelial cell activation and crescentic glomerulonephritis via Skp2. Am J Physiol Renal Physiol 2020; 319:F636-F646. [PMID: 32830536 DOI: 10.1152/ajprenal.00216.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mitotic spindle assembly checkpoint protein 2 (MAD2B), a well-known anaphase-promoting complex/cyclosome (APC/C) inhibitor and a small subunit of DNA polymerase-ζ, is critical for mitotic control and DNA repair. Previously, we detected a strong increase of MAD2B in the glomeruli from patients with crescentic glomerulonephritis and anti-glomerular basement membrane (anti-GBM) rats, which predominantly originated from activated parietal epithelial cells (PECs). Consistently, in vitro MAD2B was increased in TNF-α-treated PECs, along with cell activation and proliferation, as well as extracellular matrix accumulation, which could be reversed by MAD2B genetic depletion. Furthermore, we found that expression of S phase kinase-associated protein 2 (Skp2), an APC/CCDH1 substrate, was increased in the glomeruli of anti-GBM rats, and TNF-α-stimulated PECs and could be suppressed by MAD2B depletion. Additionally, genetic deletion of Skp2 inhibited TNF-α-induced PEC activation and dysfunction. Finally, TNF-α blockade or glucocorticoid therapy administered to anti-GBM rats could ameliorate MAD2B and Skp2 accumulation as well as weaken PEC activation. Collectively, our data suggest that MAD2B has a pivotal role in the pathogenesis of glomerular PEC activation and crescent formation through induction of Skp2 expression.
Collapse
Affiliation(s)
- Chen Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiong
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Tao Lei
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Tang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Li S, Liu Y, He Y, Rong W, Zhang M, Li L, Liu Z, Zen K. Podocytes present antigen to activate specific T cell immune responses in inflammatory renal disease. J Pathol 2020; 252:165-177. [PMID: 32686090 DOI: 10.1002/path.5508] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
Infiltration of activated T cells into renal tissue plays an essential role in inflammatory nephropathy. However, the mechanism enabling the renal recruitment and activation of T cells remains elusive. Here we report that inflammatory cytokine-promoted antigen presentation by podocytes is a key for recruiting and activating specific T cells. Our results showed that diabetes-associated inflammatory cytokines IFNγ and IL-17 all upregulated expression of MHC-I, MHC-II, CD80 and CD86 on the podocyte surface. Both IFNγ and IL-17 stimulated the uptake and processing of ovalbumin (OVA) by mouse podocytes, resulting in presentation of OVA antigen peptide on the cell surface. OVA antigen presentation by podocytes was also validated using human podocytes. Furthermore, OVA antigen-presenting mouse podocytes were able to activate OT-I mouse T cell proliferation and inflammatory cytokine secretion, which in turn caused podocyte injury and apoptosis. Finally, OT-I mice subjected to direct renal injection of OVA plus IFNγ/IL-17 but not OVA alone exhibited OVA antigen presentation by podocytes and developed nephropathy in 4 weeks. In conclusion, antigen presentation by podocytes under inflammatory conditions plays an important role in activating T cell immune responses and facilitating immune-mediated glomerular disease development. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Shan Li
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| | - Ying Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| | - Yueqin He
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| | - Weiwei Rong
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China
| | - Limin Li
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China
| | - Ke Zen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.,State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, PR China
| |
Collapse
|
22
|
Merchant ML, Barati MT, Caster DJ, Hata JL, Hobeika L, Coventry S, Brier ME, Wilkey DW, Li M, Rood IM, Deegens JK, Wetzels JF, Larsen CP, Troost JP, Hodgin JB, Mariani LH, Kretzler M, Klein JB, McLeish KR. Proteomic Analysis Identifies Distinct Glomerular Extracellular Matrix in Collapsing Focal Segmental Glomerulosclerosis. J Am Soc Nephrol 2020; 31:1883-1904. [PMID: 32561683 DOI: 10.1681/asn.2019070696] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 04/13/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The mechanisms leading to extracellular matrix (ECM) replacement of areas of glomerular capillaries in histologic variants of FSGS are unknown. This study used proteomics to test the hypothesis that glomerular ECM composition in collapsing FSGS (cFSGS) differs from that of other variants. METHODS ECM proteins in glomeruli from biopsy specimens of patients with FSGS not otherwise specified (FSGS-NOS) or cFSGS and from normal controls were distinguished and quantified using mass spectrometry, verified and localized using immunohistochemistry (IHC) and confocal microscopy, and assessed for gene expression. The analysis also quantified urinary excretion of ECM proteins and peptides. RESULTS Of 58 ECM proteins that differed in abundance between cFSGS and FSGS-NOS, 41 were more abundant in cFSGS and 17 in FSGS-NOS. IHC showed that glomerular tuft staining for cathepsin B, cathepsin C, and annexin A3 in cFSGS was significantly greater than in other FSGS variants, in minimal change disease, or in membranous nephropathy. Annexin A3 colocalized with cathepsin B and C, claudin-1, phosphorylated ERK1/2, and CD44, but not with synaptopodin, in parietal epithelial cells (PECs) infiltrating cFSGS glomeruli. Transcripts for cathepsins B and C were increased in FSGS glomeruli compared with normal controls, and urinary excretion of both cathepsins was significantly greater in cFSGS compared with FSGS-NOS. Urinary excretion of ECM-derived peptides was enhanced in cFSGS, although in silico analysis did not identify enhanced excretion of peptides derived from cathepsin B or C. CONCLUSIONS ECM differences suggest that glomerular sclerosis in cFSGS differs from that in other FSGS variants. Infiltration of activated PECs may disrupt ECM remodeling in cFSGS. These cells and their cathepsins may be therapeutic targets.
Collapse
Affiliation(s)
- Michael L Merchant
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Michelle T Barati
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Dawn J Caster
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Jessica L Hata
- Pathology Department, Norton Children's Hospital, Louisville, Kentucky
| | - Liliane Hobeika
- Division of Nephrology, Department of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Susan Coventry
- Pathology Department, Norton Children's Hospital, Louisville, Kentucky
| | - Michael E Brier
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel W Wilkey
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Ming Li
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Ilse M Rood
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen K Deegens
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jack F Wetzels
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Jonathan P Troost
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, Michigan
| | - Jeffrey B Hodgin
- Division of Pathology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Laura H Mariani
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jon B Klein
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Kenneth R McLeish
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
23
|
Hamatani H, Eng DG, Hiromura K, Pippin JW, Shankland SJ. CD44 impacts glomerular parietal epithelial cell changes in the aged mouse kidney. Physiol Rep 2020; 8:e14487. [PMID: 32597007 PMCID: PMC7322268 DOI: 10.14814/phy2.14487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
CD44 contributes to the activation of glomerular parietal epithelial cells (PECs). Although CD44 expression is higher in PECs of healthy aged mice, the biological role of CD44 in PECs in this context remains unclear. Accordingly, young (4 months) and aged (24 months) CD44-/- mice were compared to age-matched CD44+/+ mice, both aged in a nonstressed environment. Parietal epithelial cell densities were similar in both young and aged CD44+/+ and CD44-/- mice. Phosphorylated ERK 1/2 (pERK) was higher in aged CD44+/+ mice. Vimentin and α-SMA, markers of changes to the epithelial cell phenotype, were present in PECs in aged CD44+/+ mice, but absent in aged CD44-/- mice in both outer cortical (OC) and juxtamedullary (JM) glomeruli. Because age-related glomerular hypertrophy was lower in CD44-/- mice, mTOR activation was assessed by phospho-S6 ribosomal protein (pS6RP) staining. Parietal epithelial cells and glomerular tuft staining for pS6RP was lower in aged CD44-/- mice compared to aged CD44+/+ mice. Podocyte density was higher in aged CD44-/- mice in both OC and JM glomeruli. These changes were accompanied by segmental and global glomerulosclerosis in aged CD44+/+ mice, but absent in aged CD44-/- mice. These results show that the increase in CD44 in PECs in aged kidneys contributes to several changes to the glomerulus during healthy aging in mice, and may involve ERK and mTOR activation.
Collapse
Affiliation(s)
- Hiroko Hamatani
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Diana G. Eng
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of MedicineMaebashiJapan
| | - Jeffrey W. Pippin
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| | - Stuart J. Shankland
- Division of NephrologyUniversity of Washington School of MedicineSeattleWAUSA
| |
Collapse
|
24
|
Hudkins KL, Wietecha TA, Steegh F, Alpers CE. Beneficial effect on podocyte number in experimental diabetic nephropathy resulting from combined atrasentan and RAAS inhibition therapy. Am J Physiol Renal Physiol 2020; 318:F1295-F1305. [PMID: 32249614 DOI: 10.1152/ajprenal.00498.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Podocyte loss and proteinuria are both key features of human diabetic nephropathy (DN). The leptin-deficient BTBR mouse strain with the ob/ob mutation develops progressive weight gain, type 2 diabetes, and diabetic nephropathy that has many features of advanced human DN, including increased mesangial matrix, mesangiolysis, podocyte loss, and proteinuria. Selective antagonism of the endothelin-1 type A receptor (ETAR) by atrasentan treatment in combination with renin-angiotensin-aldosterone system inhibition with losartan has been shown to have the therapeutic benefit of lowering proteinuria in patients with DN, but the underlying mechanism for this benefit is not well understood. Using a similar therapeutic approach in diabetic BTBR ob/ob mice, this treatment regimen significantly increased glomerular podocyte number compared with diabetic BTBR ob/ob controls and suggested that parietal epithelial cells were a source for podocyte restoration. Atrasentan treatment alone also increased podocyte number but to a lesser degree. Mice treated with atrasentan demonstrated a reduction in proteinuria, matching the functional improvement reported in humans. This is a first demonstration that treatment with the highly selective ETAR antagonist atrasentan can lead to restoration of the diminished podocyte number characteristic of DN in humans and thereby underlies the reduction in proteinuria in patients with diabetes undergoing similar treatment. The benefit of ETAR antagonism in DN extended to a decrease in mesangial matrix as measured by a reduction in accumulations of collagen type IV in both the atrasentan and atrasentan + losartan-treated groups compared with untreated controls.
Collapse
Affiliation(s)
- Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle, Washington
| | - Tomasz A Wietecha
- Department of Pathology, University of Washington, Seattle, Washington
| | - Floor Steegh
- Department of Pathology, University of Washington, Seattle, Washington
| | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
25
|
Ito N, Sakamoto K, Hikichi C, Matsusaka T, Nagata M. Biphasic MIF and SDF1 expression during podocyte injury promote CD44-mediated glomerular parietal cell migration in focal segmental glomerulosclerosis. Am J Physiol Renal Physiol 2020; 318:F741-F753. [PMID: 32068458 DOI: 10.1152/ajprenal.00414.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glomerular parietal epithelial cell (PEC) activation, as revealed by de novo expression of CD44 and cell migration toward the injured filtration barrier, is a hallmark of podocyte injury-driven focal segmental glomerulosclerosis (FSGS). However, the signaling pathway that mediates activation of PECs in response to podocyte injury is unknown. The present study focused on CD44 signaling, particularly the roles of two CD44-related chemokines, migration inhibitory factor (MIF) and stromal cell-derived factor 1 (SDF1), and their common receptor, chemokine (C-X-C motif) receptor 4 (CXCR4), in the NEP25/LMB2 mouse podocyte-toxin model of FSGS. In the early phase of the disease, CD44-positive PECs were locally evident on the opposite side of the intact glomerular tuft and subsequently increased in the vicinity of synechiae with podocyte loss. Expression of MIF and SDF1 was first increased in injured podocytes and subsequently transferred to activated PECs expressing CD44 and CXCR4. In an immortalized mouse PEC (mPEC) line, recombinant MIF and SDF1 (rMIF and rSDF1, respectively) individually increased CD44 and CXCR4 mRNA and protein levels. rMIF and rSDF1 stimulated endogenous MIF and SDF1 production. rMIF- and rSDF1-induced mPEC migration was suppressed by CD44 siRNA. However, MIF and SDF1 inhibitors failed to show any impact on proteinuria, podocyte number, and CD44 expression in NEP25/LMB2 mice. Our data suggest that injured podocytes upregulate MIF and SDF1 that stimulate CD44 expression and CD44-mediated migration, which is enhanced by endogenous MIF and SDF1 in PECs. This biphasic expression pattern of the chemokine-CD44 axis in podocytes and PECs may be a novel mechanism of "podocyte-PEC cross-talk" signaling underlying podocyte injury-driven FSGS.
Collapse
Affiliation(s)
- Naoko Ito
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazuo Sakamoto
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Chihiro Hikichi
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Taiji Matsusaka
- Department of Basic Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Michio Nagata
- Department of Pathology, Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
26
|
Opriş EC, Suciu H, Jung I, Satală CB, Al Hussein H, Harpa MM, Bănceu CM, Gurzu S. Mesangioproliferative glomerulonephritis with extracapillary crescents - unexpected fatal complication in a 17-year-old patient with implanted left ventricular assist device. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2020; 61:535-544. [PMID: 33544806 PMCID: PMC7864293 DOI: 10.47162/rjme.61.2.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The continuous flow left ventricular assist device (cf-LVAD) is the life-saving solution for patients with end-stage global heart failure. We present the case of a young patient with biventricular dilated cardiomyopathy, who had a cf-LVAD implantation and died as result of progressive renal failure. In the first year after implantation, he suffered repeated strokes and episodes of pneumonia with Klebsiella pneumoniae and Escherichia coli. The patient had hypertension, which was kept under control with angiotensin-converting enzyme (ACE) inhibitors and beta-blockers. After multiple bleeding episodes, the patient died at 21 months after the LVAD implant. At autopsy, parenchymatous brain hemorrhage was found to be associated with pulmonary hemorrhages. The unexpected features related to mesangioproliferative and extracapillary glomerulonephritis, with focal glomerulosclerosis. The proliferated parietal cells of Bowman’s capsule proved to express cluster of differentiation 44 (CD44), whereas remnant podocytes and mesangial cells showed Wilms tumor 1 (WT1) positivity. Since CD44 might be involved in fibrogenesis, but ACE inhibitors can exert a protective role against glomerular deterioration, we performed a synthesis of literature data which enabled us to propose a hypothesis with a potential clinical impact. We conclude that, in patients with LVAD implants, high blood pressure and high serum level of angiotensin II, the association between ACE inhibitors and anti-CD44 agents might exert glomerular protection and increase the survival time. Experimental studies are necessary to support our hypothesis and to explain the mechanism of possible glomerulopathy installed after LVAD implant.
Collapse
Affiliation(s)
- Elena Carmen Opriş
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureş, Romania;
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chan GC, Eng DG, Miner JH, Alpers CE, Hudkins K, Chang A, Pippin JW, Shankland SJ. Differential expression of parietal epithelial cell and podocyte extracellular matrix proteins in focal segmental glomerulosclerosis and diabetic nephropathy. Am J Physiol Renal Physiol 2019; 317:F1680-F1694. [PMID: 31630546 PMCID: PMC6962515 DOI: 10.1152/ajprenal.00266.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 01/03/2023] Open
Abstract
In healthy glomeruli, parietal epithelial cell (PEC)-derived extracellular matrix (ECM) proteins include laminin-β1, perlecan, and collagen type IV-α2 and podocyte-specific ECM proteins include laminin-β2, agrin, and collagen type IV-α4. This study aimed to define individual ECM protein isoform expression by PECs in both experimental and human focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy (DN) and to determine if changes were CD44 dependent. In experimental FSGS induced with a cytotoxic podocyte antibody and in the BTBR ob/ob mouse model of DN, PEC-derived protein staining was significantly increased in PECs. Dual staining also showed de novo expression of the podocyte-specific ECM proteins laminin-β2 and agrin in PECs. Similar findings were observed in biopsies from patients with FSGS and DN. Increases in individual ECM proteins colocalized with CD44 in PECs in disease. To determine the role of CD44, FSGS was induced in CD44-/- and CD44+/+ mice. PEC staining for perlecan, collagen type IV-α2, laminin-β2, and agrin were significantly lower in diseased CD44-/- mice compared with diseased CD44+/+ mice. These results show that in experimental and human FSGS and DN, PECs typically in an activated state, produce both PEC-derived and podocyte-specific ECM protein isoforms, and that the majority of these changes were dependent on CD44.
Collapse
Affiliation(s)
- Gek Cher Chan
- Division of Nephrology, University of Washington, Seattle, Washington
- Division of Nephrology, National University Hospital, Singapore
| | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle, Washington
| | - Kelly Hudkins
- Department of Pathology, University of Washington, Seattle, Washington
| | - Anthony Chang
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington
| | | |
Collapse
|
28
|
Smeets B, Miesen L, Shankland SJ. CD9 Is a Novel Target in Glomerular Diseases Typified by Parietal Epithelial Cell Activation. Am J Kidney Dis 2019; 75:812-814. [PMID: 31668876 DOI: 10.1053/j.ajkd.2019.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 08/29/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Bart Smeets
- Department of Pathology, RIMLS, Radboudumc, Nijmegen, the Netherlands
| | - Laura Miesen
- Department of Pathology, RIMLS, Radboudumc, Nijmegen, the Netherlands
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
29
|
Kaverina NV, Eng DG, Freedman BS, Kutz JN, Chozinski TJ, Vaughan JC, Miner JH, Pippin JW, Shankland SJ. Dual lineage tracing shows that glomerular parietal epithelial cells can transdifferentiate toward the adult podocyte fate. Kidney Int 2019; 96:597-611. [PMID: 31200942 PMCID: PMC7008116 DOI: 10.1016/j.kint.2019.03.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022]
Abstract
Podocytes are differentiated post-mitotic cells that cannot replace themselves after injury. Glomerular parietal epithelial cells are proposed to be podocyte progenitors. To test whether a subset of parietal epithelial cells transdifferentiate to a podocyte fate, dual reporter PEC-rtTA|LC1|tdTomato|Nphs1-FLPo|FRT-EGFP mice, named PEC-PODO, were generated. Doxycycline administration permanently labeled parietal epithelial cells with tdTomato reporter (red), and upon doxycycline removal, the parietal epithelial cells (PECs) cannot label further. Despite the presence or absence of doxycycline, podocytes cannot label with tdTomato, but are constitutively labeled with an enhanced green fluorescent protein (EGFP) reporter (green). Only activation of the Nphs1-FLPo transgene by labeled parietal epithelial cells can generate a yellow color. At day 28 of experimental focal segmental glomerulosclerosis, podocyte density was 20% lower in 20% of glomeruli. At day 56 of experimental focal segmental glomerulosclerosis, podocyte density was 18% lower in 17% of glomeruli. TdTomato+ parietal epithelial cells were restricted to Bowman's capsule in healthy mice. However, by days 28 and 56 of experimental disease, two-thirds of tdTomato+ parietal epithelial cells within glomerular tufts were yellow in color. These cells co-expressed the podocyte markers podocin, nephrin, p57 and VEGF164, but not markers of endothelial (ERG) or mesangial (Perlecan) cells. Expansion microscopy showed primary, secondary and minor processes in tdTomato+EGFP+ cells in glomerular tufts. Thus, our studies provide strong evidence that parietal epithelial cells serve as a source of new podocytes in adult mice.
Collapse
Affiliation(s)
- Natalya V Kaverina
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | | | - J Nathan Kutz
- Department of Applied Mathematics, University of Washington, Seattle, Washington, USA
| | - Tyler J Chozinski
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, Washington, USA; Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
30
|
Zhong J, Whitman JB, Yang HC, Fogo AB. Mechanisms of Scarring in Focal Segmental Glomerulosclerosis. J Histochem Cytochem 2019; 67:623-632. [PMID: 31116068 PMCID: PMC6713971 DOI: 10.1369/0022155419850170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/22/2019] [Indexed: 01/17/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) presents with scar in parts of some glomeruli and often progresses to global and diffuse glomerulosclerosis. Podocyte injury is the initial target in primary FSGS, induced by a circulating factor. Several gene variants, for example, APOL1, are associated with increased susceptibility to FSGS. Primary FSGS may be due to genetic mutation in key podocyte genes. Increased work stress after loss of nephrons, epigenetic mechanisms, and various profibrotic pathways can contribute to progressive sclerosis, regardless of the initial injury. The progression of FSGS lesions also involves crosstalk between podocytes and other kidney cells, such as parietal epithelial cells, glomerular endothelial cells, and even tubular epithelial cells. New insights related to these mechanisms could potentially lead to new therapeutic strategies to prevent progression of FSGS.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jacob B Whitman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hai-Chun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
31
|
Sakhi H, Moktefi A, Bouachi K, Audard V, Hénique C, Remy P, Ollero M, El Karoui K. Podocyte Injury in Lupus Nephritis. J Clin Med 2019; 8:jcm8091340. [PMID: 31470591 PMCID: PMC6780135 DOI: 10.3390/jcm8091340] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by a broad spectrum of renal lesions. In lupus glomerulonephritis, histological classifications are based on immune-complex (IC) deposits and hypercellularity lesions (mesangial and/or endocapillary) in the glomeruli. However, there is compelling evidence to suggest that glomerular epithelial cells, and podocytes in particular, are also involved in glomerular injury in patients with SLE. Podocytes now appear to be not only subject to collateral damage due to glomerular capillary lesions secondary to IC and inflammatory processes, but they are also a potential direct target in lupus nephritis. Improvements in our understanding of podocyte injury could improve the classification of lupus glomerulonephritis. Indeed, podocyte injury may be prominent in two major presentations: lupus podocytopathy and glomerular crescent formation, in which glomerular parietal epithelial cells play also a key role. We review here the contribution of podocyte impairment to different presentations of lupus nephritis, focusing on the podocyte signaling pathways involved in these lesions.
Collapse
Affiliation(s)
- Hamza Sakhi
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Anissa Moktefi
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Pathology, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Khedidja Bouachi
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Vincent Audard
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Carole Hénique
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Philippe Remy
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France
| | - Mario Ollero
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France
| | - Khalil El Karoui
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, 94010 Créteil, France.
- UPEC (Université Paris Est Créteil), UMR-S955, 94010 Créteil, France.
- INSERM (Institut National de la Santé et de la Recherche Médicale) U955, Institut Mondor de Recherche Biomédicale (IMRB), Équipe 21, 94010 Créteil, France.
| |
Collapse
|
32
|
Kidney Injury Molecule-1 Is Upregulated in Renal Lipotoxicity and Mediates Palmitate-Induced Tubular Cell Injury and Inflammatory Response. Int J Mol Sci 2019; 20:ijms20143406. [PMID: 31373312 PMCID: PMC6679556 DOI: 10.3390/ijms20143406] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 12/20/2022] Open
Abstract
Diabetic nephropathy is increasingly recognized as a major contributor to kidney failure in patients with obesity and type 2 diabetes. This study was designed to identify the molecular mediators of kidney injury associated with metabolic syndrome with or without hyperglycemia. We compared renal gene expression profiles in Zucker lean (ZL), Zucker obese (ZO), and Zucker diabetic (ZD) rats using cDNA microarray with quantitative verification of selected transcripts by real-time PCR. Compared to the 20-week-old ZL control (glucose: 110 ± 8 mg/dL), both prediabetic ZO (glucose: 157 ± 11 mg/dL) and diabetic ZD (glucose: 481 ± 37 mg/dL) rats displayed hyperlipidemia and kidney injury with a high degree of proteinuria. cDNA microarray identified 25 inflammation and injury-related transcriptomes whose expression levels were similarly increased in ZO and ZD kidneys. Among them, kidney injury molecule-1 (KIM-1) was found to be the most highly upregulated in both ZO and ZD kidneys. Immunofluorescence staining of kidney sections revealed a strong correlation between lipid overload and KIM-1 upregulation in proximal tubules of ZO and ZD rats. In cultured primary renal tubular epithelial cells (TECs), administration of saturated fatty acid palmitate resulted in an upregulation of KIM-1, osteopontin, and CD44, which was greatly attenuated by U0126, an inhibitor of extracellular signal-regulated kinase (ERK)1/2. Moreover, knockdown of KIM-1 by siRNA interference inhibited palmitate-induced cleaved caspase-3, osteopontin, and CD44 proteins in primary TECs. Our results indicate that KIM-1 expression is upregulated in renal lipotoxicity and may play an important role in fatty acid-induced inflammation and tubular cell damage in obesity and diabetic kidney disease.
Collapse
|
33
|
Zhao X, Chen X, Chima A, Zhang Y, George J, Cobbs A, Emmett N. Albumin induces CD44 expression in glomerular parietal epithelial cells by activating extracellular signal-regulated kinase 1/2 pathway. J Cell Physiol 2019; 234:7224-7235. [PMID: 30362534 PMCID: PMC6344259 DOI: 10.1002/jcp.27477] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/04/2018] [Indexed: 01/21/2023]
Abstract
De novo expression of CD44 in glomerular parietal epithelial cells (PECs) leads to a prosclerotic and migratory PEC phenotype in glomerulosclerosis. However, the regulatory mechanisms underlying CD44 expression by activated PECs remain largely unknown. This study was performed to examine the mediators responsible for CD44 induction in glomerular PECs in association with diabetes. CD44 expression and localization were evaluated in the glomeruli of Zucker diabetic rat kidneys and primary cultured PECs upon albumin stimulation. Real-time polymerase chain reaction confirmed an albuminuria-associated upregulation of the CD44 gene in the glomeruli of diabetic rats. Immunostaining analysis of diabetic kidneys further revealed an increase in CD44 in hypertrophic PECs, which often contain albumin-positive vesicles. Losartan treatment significantly attenuated albuminuria and lowered CD44 protein levels in the diabetic kidneys. In primary cultured rat PECs, rat serum albumin (0.25-1 mg/ml) caused a dose-dependent upregulation of CD44, claudin-1, and megalin protein expression, which was accompanied by an activation of extracellular signal-regulated kinase1/2 (ERK1/2) signaling. Albumin-induced CD44 and claudin-1 expression were greatly suppressed in the presence of the ERK1/2 inhibitor, U0126. In addition, knockdown of megalin by small interfering RNA interference in PECs resulted in a significant reduction of albumin-induced CD44 and claudin-1 proteins. Taken together, our results demonstrate that albumin induces CD44 expression by PECs via the activation of the ERK signaling pathway, which is partially mediated by endocytic receptor megalin.
Collapse
Affiliation(s)
- Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Xiaoming Chen
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Ashmeer Chima
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Yuanyuan Zhang
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Jasmine George
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Alyssa Cobbs
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Nerimiah Emmett
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
34
|
Kuppe C, Leuchtle K, Wagner A, Kabgani N, Saritas T, Puelles VG, Smeets B, Hakroush S, van der Vlag J, Boor P, Schiffer M, Gröne HJ, Fogo A, Floege J, Moeller MJ. Novel parietal epithelial cell subpopulations contribute to focal segmental glomerulosclerosis and glomerular tip lesions. Kidney Int 2019; 96:80-93. [PMID: 31029503 PMCID: PMC7292612 DOI: 10.1016/j.kint.2019.01.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
Abstract
Beside the classical flat parietal epithelial cells (PECs), we investigated proximal tubular epithelial-like cells, a neglected subgroup of PECs. These cells, termed cuboidal PECs, make up the most proximal part of the proximal tubule and may also line parts of Bowman's capsule. Additionally, a third intermediate PEC subgroup was identified at the junction between the flat and cuboidal PEC subgroups at the tubular orifice. The transgenic mouse line PEC-rtTA labeled all three PEC subgroups. Here we show that the inducible Pax8-rtTA mouse line specifically labeled only cuboidal and intermediate PECs, but not flat PECs. In aging Pax8-rtTA mice, cell fate mapping showed no evidence for significant transdifferentiation from flat PECs to cuboidal or intermediate PECs or vice versa. In murine glomerular disease models of crescentic glomerulonephritis, and focal segmental glomerulosclerosis (FSGS), intermediate PECs became more numerous. These intermediate PECs preferentially expressed activation markers CD44 and Ki-67, suggesting that this subgroup of PECs was activated more easily than the classical flat PECs. In mice with FSGS, cuboidal and intermediate PECs formed sclerotic lesions. In patients with FSGS, cells forming the tip lesions expressed markers of intermediate PECs. These novel PEC subgroups form sclerotic lesions and were more prone to cellular activation compared to the classical flat PECs in disease. Thus, colonization of Bowman's capsule by cuboidal PECs may predispose to lesion formation and chronic kidney disease. We propose that tip lesions originate from this novel subgroup of PECs in patients with FSGS.
Collapse
Affiliation(s)
- Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| | - Katja Leuchtle
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Anton Wagner
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nazanin Kabgani
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Turgay Saritas
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Victor G Puelles
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Cardiovascular Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, School of Biomedical Sciences, and Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia; Department of Nephrology, Monash Health, Melbourne Australia
| | - Bart Smeets
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Samy Hakroush
- Institute of Pathology, University Medical Center, Göttingen, Germany
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, University of Erlangen, Erlangen, Germany
| | - Hermann-Josef Gröne
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Agnes Fogo
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Marcus Johannes Moeller
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| |
Collapse
|
35
|
Estrada CC, Maldonado A, Mallipattu SK. Therapeutic Inhibition of VEGF Signaling and Associated Nephrotoxicities. J Am Soc Nephrol 2019; 30:187-200. [PMID: 30642877 DOI: 10.1681/asn.2018080853] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inhibition of vascular endothelial growth factor A (VEGFA)/vascular endothelial growth factor receptor 2 (VEGFR2) signaling is a common therapeutic strategy in oncology, with new drugs continuously in development. In this review, we consider the experimental and clinical evidence behind the diverse nephrotoxicities associated with the inhibition of this pathway. We also review the renal effects of VEGF inhibition's mediation of key downstream signaling pathways, specifically MAPK/ERK1/2, endothelial nitric oxide synthase, and mammalian target of rapamycin (mTOR). Direct VEGFA inhibition via antibody binding or VEGF trap (a soluble decoy receptor) is associated with renal-specific thrombotic microangiopathy (TMA). Reports also indicate that tyrosine kinase inhibition of the VEGF receptors is preferentially associated with glomerulopathies such as minimal change disease and FSGS. Inhibition of the downstream pathway RAF/MAPK/ERK has largely been associated with tubulointerstitial injury. Inhibition of mTOR is most commonly associated with albuminuria and podocyte injury, but has also been linked to renal-specific TMA. In all, we review the experimentally validated mechanisms by which VEGFA-VEGFR2 inhibitors contribute to nephrotoxicity, as well as the wide range of clinical manifestations that have been reported with their use. We also highlight potential avenues for future research to elucidate mechanisms for minimizing nephrotoxicity while maintaining therapeutic efficacy.
Collapse
Affiliation(s)
- Chelsea C Estrada
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York; and
| | - Alejandro Maldonado
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York; and
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York; and .,Renal Section, Northport Veterans Affairs Medical Center, Northport, New York
| |
Collapse
|
36
|
Suzuki T, Eng DG, McClelland AD, Pippin JW, Shankland SJ. Cells of NG2 lineage increase in glomeruli of mice following podocyte depletion. Am J Physiol Renal Physiol 2018; 315:F1449-F1464. [PMID: 30019931 PMCID: PMC6293287 DOI: 10.1152/ajprenal.00118.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Under certain circumstances, podocytes can be partially replaced following their loss in disease. The inability of podocytes to proliferate suggests that replacement derives from other cell types. Because neural/glial antigen 2 (NG2)-expressing cells can serve as progenitors in other organs and because herein we showed increased NG2 staining in podocytes following their loss in experimental focal segmental glomerulosclerosis, we used lineage tracing in NG2-CreER tdTomato mice to test the hypothesis that partial podocyte replacement might derive from this cell population. The percentage of glomeruli with red fluorescence protein (RFP)-labeled NG2 cells increased following podocyte depletion, which was augmented by enalapril. However, BrdU was not detected in RFP-labeled cells, consistent with the migration of these cells to the glomerulus. Within glomeruli, RFP-labeled cells did not coexpress podocyte proteins (p57, synaptopodin, nephrin, or podocin) but did coexpress markers for mesangial (α8 integrin, PDGFβ receptor) and parietal epithelial cells (PAX8, src-suppressed C-kinase substrate). These results suggest that following podocyte depletion, cells of NG2 lineage do not serve as adult podocyte progenitors but have the ability to transdifferentiate to mesangial and parietal epithelial cell fates.
Collapse
Affiliation(s)
- Taihei Suzuki
- Division of Nephrology, University of Washington School of Medicine , Seattle, Washington
| | - Diana G Eng
- Division of Nephrology, University of Washington School of Medicine , Seattle, Washington
| | - Aaron D McClelland
- Division of Nephrology, University of Washington School of Medicine , Seattle, Washington
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington School of Medicine , Seattle, Washington
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine , Seattle, Washington
| |
Collapse
|
37
|
Manonelles A, Guiteras R, Melilli E, Lazzeri E, Goma M, Crespo E, Bestard O, Sola A, Romagnani P, Cruzado JM. The Presence of Urinary Renal Progenitor Cells in Stable Kidney Transplant Recipients Anticipates Allograft Deterioration. Front Physiol 2018; 9:1412. [PMID: 30364198 PMCID: PMC6191504 DOI: 10.3389/fphys.2018.01412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
Long-term kidney transplant outcomes have reached mild improvements recently. Parietal epithelial cells (PECs) are progenitor cells located along the Bowman’s capsule that can be isolated in urine, and display the capability to replace podocytes, but in certain situations cause glomerulosclerosis. In this study, a cohort of stable kidney transplant recipients with 6 months protocol biopsy was divided in two groups depending on the presence (uPEC+; n = 41) or absence (uPEC-; n = 25) of PECs in urine and followed for 2 years. No differences were found between groups at 6 months after transplantation considering clinical variables, alloimmune response, renal function, albuminuria and graft pathology. However, uPEC+ group showed increased podocyturia and a higher rate of proliferating PECs along the Bowman’s capsule, without concomitant enhancement of the CD44 pro-sclerotic activation marker. Accordingly, 2 years follow up evidenced poorer outcomes in the uPEC+ group with worse renal function, increased albuminuria, wider mesangial expansion and more severe IFTA. In summary, chronic allograft damage can progress in certain stable-supposed grafts by podocyte detachment and reactive PECs proliferation, being the uPEC presence a biomarker of this process. This damage-response regenerative process, if sustained in time, might fail in preserve the allograft function and histology. Our study raises new prospects to overcome current limits on long-term allograft results.
Collapse
Affiliation(s)
- Anna Manonelles
- Nephrology Department, L'Hospitalet de Llobregat, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Roser Guiteras
- Experimental Nephrology, Department of Ciències Clíniques, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Edoardo Melilli
- Nephrology Department, L'Hospitalet de Llobregat, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Montse Goma
- Pathology Department, L'Hospitalet de Llobregat, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Elena Crespo
- Experimental Nephrology, Department of Ciències Clíniques, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Oriol Bestard
- Nephrology Department, L'Hospitalet de Llobregat, Hospital Universitari de Bellvitge, Barcelona, Spain.,Experimental Nephrology, Department of Ciències Clíniques, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Anna Sola
- Experimental Nephrology, Department of Ciències Clíniques, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Josep M Cruzado
- Nephrology Department, L'Hospitalet de Llobregat, Hospital Universitari de Bellvitge, Barcelona, Spain.,Experimental Nephrology, Department of Ciències Clíniques, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Liu GW, Prossnitz AN, Eng DG, Cheng Y, Subrahmanyam N, Pippin JW, Lamm RJ, Ngambenjawong C, Ghandehari H, Shankland SJ, Pun SH. Glomerular disease augments kidney accumulation of synthetic anionic polymers. Biomaterials 2018; 178:317-325. [DOI: 10.1016/j.biomaterials.2018.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/31/2018] [Accepted: 06/02/2018] [Indexed: 12/22/2022]
|
39
|
Eymael J, Sharma S, Loeven MA, Wetzels JF, Mooren F, Florquin S, Deegens JK, Willemsen BK, Sharma V, van Kuppevelt TH, Bakker MA, Ostendorf T, Moeller MJ, Dijkman HB, Smeets B, van der Vlag J. CD44 is required for the pathogenesis of experimental crescentic glomerulonephritis and collapsing focal segmental glomerulosclerosis. Kidney Int 2018; 93:626-642. [DOI: 10.1016/j.kint.2017.09.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 09/11/2017] [Accepted: 09/21/2017] [Indexed: 10/18/2022]
|
40
|
Wen Y, Liu L, Xu Q, Zhou P, Li H, Wang Z, Liang J. Knocking down Cabin1 induces glomerular podocyte injury. Int Urol Nephrol 2018; 50:983-991. [PMID: 29368245 DOI: 10.1007/s11255-018-1787-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 01/06/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND Podocyte damage exerts a key role in proteinuria. We have demonstrated that calcineurin-binding protein 1 (Cabin1) upregulated during podocyte injury, yet its function in podocyte is still unclear. METHODS We established 5/6 nephrectomized rats and angiotensin II (AngII)-injured podocyte, as well as knocked down Cabin1 with siRNA in cultured podocytes. Rats were killed at 4 or 8 weeks after 5/6 nephrectomy. The localization of podocyte cytoskeleton was detected after immunofluorescence staining. Podocyte mitochondrial morphology was observed under electron microscopy. Podocyte mitochondrial transmembrane potential (MMP) was measured with MitoCapture kit. Cabin1 and cytochrome c protein expression were detected by western blot. RESULTS Massive proteinuria, as well as obvious segmental glomerular sclerosis, was found in rats at 8 weeks after nephrectomy, accompanied with the disruption of synaptopodin. Moreover, mitochondria changed from large and ellipsoid shape to the small, long, and irregular shape in rats at 4 weeks after operation. At 8 weeks, mitochondria were swollen and cristae were remarkably dissolved. Compared to sham-operated rats, Cabin1 protein expression was obviously upregulated in rats at 8 weeks. AngII induced the decrease in MMP, as well as the overexpression of Cabin1 and cytochrome c protein in podocytes. Knocking down Cabin1 induced the disruption of F-actin and overexpression of cytochrome c (1.81 ± 0.21 in siRNA group vs. 0.86 ± 0.11 in negative control group). CONCLUSIONS Knocking down Cabin1 induces the disruption of cytoskeleton and mitochondrial dysfunction in podocyte. Cabin1 could be a crucial factor in podocyte damage.
Collapse
Affiliation(s)
- Yueqiang Wen
- Department of Nephrology, The Second Affiliated Hospital, GuangZhou Medical University, 250th, Chang Gang East Road, Guangzhou, 510260, China.
| | - Lingling Liu
- Department of General Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingdong Xu
- Department of Nephrology, Jiangmen Central Hospital, Guangdong, China
| | - Peilan Zhou
- Department of Nephrology, The Second Affiliated Hospital, GuangZhou Medical University, 250th, Chang Gang East Road, Guangzhou, 510260, China
| | - Huiyuan Li
- Department of Nephrology, The Second Affiliated Hospital, GuangZhou Medical University, 250th, Chang Gang East Road, Guangzhou, 510260, China
| | - Zebin Wang
- Department of Nephrology, The Second Affiliated Hospital, GuangZhou Medical University, 250th, Chang Gang East Road, Guangzhou, 510260, China
| | - Jianbo Liang
- Department of Nephrology, The Second Affiliated Hospital, GuangZhou Medical University, 250th, Chang Gang East Road, Guangzhou, 510260, China
| |
Collapse
|
41
|
Miolo G, Sturaro G, Cigolini G, Menilli L, Tasso A, Zago I, Conconi MT. 4,6,4'-trimethylangelicin shows high anti-proliferative activity on DU145 cells under both UVA and blue light. Cell Prolif 2018; 51:e12430. [PMID: 29318693 DOI: 10.1111/cpr.12430] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Furocoumarins (psoralens and angelicins) have been already used under ultraviolet A light (UVA) for the treatment of skin diseases and cutaneous T-cell lymphoma. Besides their high anti-proliferative activity, some severe long-term side effects have been observed, for example genotoxicity and mutagenicity, likely strictly related to the formation of crosslinks. It has been demonstrated that blue light (BL) activation of 8-methoxypsoralen, an FDA-approved drug, leads to less mutagenic monoadducts in the DNA. So far, in this work the less toxic and more penetrating BL is proposed to activate 4,6,4'-trimethylangelicin (TMA), an already known UVA photoactivatable compound. MATERIALS AND METHODS Photocleavage, crosslink formation and oxidative damage were detected in pBR322 plasmid DNA treated with 300.0 μmol/L TMA activated with various exposures of BL. Anti-proliferative activity, reactive oxygen species (ROS) formation and activation status of some signalling pathways involved in cell growth and apoptosis were verified on DU145 cells treated with 5.0 μmol/L TMA plus 2.0 J/cm2 of BL. RESULTS Under BL-TMA, no mutagenic crosslinks, no photocleavage and neither photooxidative lesions were detected on isolated plasmid DNA. TMA showed high anti-proliferative activity on DU145 cells through induction of apoptosis. Besides ROS generation, the proapoptotic effect seemed to be related to activation of p38 and inhibition of p44/42 phosphorylation. Interestingly, the decrease in nuclear β-catenin was coupled with a significant dropping of CD44-positive cells. CONCLUSION Overall, our results indicate that TMA can be activated by BL and may be considered for targeted phototherapy of prostate cancer lesions.
Collapse
Affiliation(s)
- G Miolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - G Sturaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - G Cigolini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - L Menilli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - A Tasso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - I Zago
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - M T Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
42
|
McClelland AD, Lichtnekert J, Eng DG, Pippin JW, Gross KW, Gharib SA, Shankland SJ. Charting the transcriptional landscape of cells of renin lineage following podocyte depletion. PLoS One 2017; 12:e0189084. [PMID: 29232382 PMCID: PMC5726629 DOI: 10.1371/journal.pone.0189084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/17/2017] [Indexed: 11/19/2022] Open
Abstract
Renin producing cells of the juxtaglomerulus, herein called cells of renin lineage (CoRL), have garnered recent interest for their propensity to act as a progenitor source for various kidney cell types including podocytes. Despite recent advances, the process of transdifferentiation of CoRL to podocytes is poorly understood. In this study, we employed a transgenic reporter mouse line which permanently labels CoRL with ZsGreen fluorescent protein, allowing for isolation by fluorescence-activated cell sorting. At 5 days following induction of abrupt podocyte ablation via anti-podocyte sheep IgG, mice were sacrificed and CoRL were isolated by FACS. RNA was subsequently analyzed by microarray. Gene set enrichment analysis (GSEA) was performed and revealed that CoRL display a distinct phenotype following podocyte ablation, primarily consisting of downregulation of metabolic processes and upregulation of immuno-modulatory processes. Additionally, RNA-biology and cell cycle-related processes were also upregulated. Changes in gene expression or activity of a core set of transcription factors including HNF1 and E2F were identified through changes in enrichment of their respective target genes. However, integration of results from transcription factor and canonical pathway analysis indicated that ERR1 and PU-box family members may be the major contributors to the post-podocyte ablation phenotype of CoRL. Finally, top ranking genes were selected from the microarray-based analysis and confirmed by qPCR. Collectively, our results provide valuable insights into the transcriptional regulation of CoRL following abrupt podocyte ablation.
Collapse
Affiliation(s)
- Aaron D. McClelland
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Julia Lichtnekert
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Diana G. Eng
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Jeffrey W. Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Sina A. Gharib
- Computational Medicine Core, Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Stuart J. Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
43
|
Compound effects of aging and experimental FSGS on glomerular epithelial cells. Aging (Albany NY) 2017; 9:524-546. [PMID: 28222042 PMCID: PMC5361679 DOI: 10.18632/aging.101176] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/09/2017] [Indexed: 12/27/2022]
Abstract
Advanced age portends a poorer prognosis in FSGS. To understand the impact of age on glomerular podocytes and parietal epithelial cells (PECs), experimental FSGS was induced in 3m-old mice (20-year old human age) and 27m-old mice (78-year old human age) by abruptly depleting podocytes with a cytopathic anti-podocyte antibody. Despite similar binding of the disease-inducing antibody, podocyte density was lower in aged FSGS mice compared to young FSGS mice. Activated PEC density was higher in aged versus young FSGS mice, as was the percentage of total activated PECs. Additionally, the percentage of glomeruli containing PECs with evidence of phosphorylated ERK and EMT was higher in aged FSGS mice. Extracellular matrix, measured by collagen IV and silver staining, was higher in aged FSGS mice along Bowman's capsule. However, collagen IV accumulation in the glomerular tufts alone and in glomeruli with both tuft and Bowman's capsule accumulation were similar in young FSGS and aged FSGS mice. Thus, the major difference in collagen IV staining in FSGS was along Bowman's capsule in aged mice. The significant differences in podocytes, PECs and extracellular matrixaccumulation between young mice and old mice with FSGS might explain the differences in outcomes in FSGS based on age.
Collapse
|
44
|
Nagata M, Kobayashi N, Hara S. Focal segmental glomerulosclerosis; why does it occur segmentally? Pflugers Arch 2017; 469:983-988. [PMID: 28664408 DOI: 10.1007/s00424-017-2023-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
Podocyte loss is the fundamental basis of glomerulosclerosis. Focal segmental glomerulosclerosis (FSGS) is a progressive glomerular disease, and its glomerular features are a prototype of podocyte loss-driven glomerulosclerosis. The glomerular pathology of FSGS is characterized by a focal and segmental location of the sclerotic lesions in human FSGS; segmental sclerosis often shows simultaneous intra- and extra-capillary changes, including parietal cell migration, capillary collapse, hyaline deposition, and intra-capillary thrombi and occasional hypercellularity. This suggests that local cellular events, initiated by podocyte loss, are the basis of the segmental lesions in FSGS. Using podocyte-specific injury by toxin administration, a series of recent works has identified the cellular basis of the glomerular response to podocyte loss. This review discusses the molecular pathway of the local response to podocyte loss and its progression to sclerosis. Recent results suggest that segmental sclerosis is a physiological tissue response aimed at halting protein leakage from a disrupted filtration barrier.
Collapse
Affiliation(s)
- Michio Nagata
- Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Ten-nodai 1-1-1, Tsukuba-City, Ibaraki, 305-8577, Japan.
| | - Namiko Kobayashi
- Nephrology, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-850, Japan
| | - Satoshi Hara
- Rheumatology, Kanazawa University Graduate School of Medicine, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8671, Japan
| |
Collapse
|