1
|
Fogo AB, Harris RC. Crosstalk between glomeruli and tubules. Nat Rev Nephrol 2025; 21:189-199. [PMID: 39643696 DOI: 10.1038/s41581-024-00907-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Models of kidney injury have classically concentrated on glomeruli as the primary site of injury leading to glomerulosclerosis or on tubules as the primary site of injury leading to tubulointerstitial fibrosis. However, current evidence on the mechanisms of progression of chronic kidney disease indicates that a complex interplay between glomeruli and tubules underlies progressive kidney injury. Primary glomerular injury can clearly lead to subsequent tubule injury. For example, damage to the glomerular filtration barrier can expose tubular cells to serum proteins, including complement and cytokines, that would not be present in physiological conditions and can promote the development of tubulointerstitial fibrosis and progressive decline in kidney function. In addition, although less well-studied, increasing evidence suggests that tubule injury, whether primary or secondary, can also promote glomerular damage. This feedback from the tubule to the glomerulus might be mediated by changes in the reabsorptive capacity of the tubule, which can affect the glomerular filtration rate, or by mediators released by injured proximal tubular cells that can induce damage in both podocytes and parietal epithelial cells. Examining the crosstalk between the various compartments of the kidney is important for understanding the mechanisms underlying kidney pathology and identifying potential therapeutic interventions.
Collapse
Affiliation(s)
- Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Tennessee Department of Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
2
|
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
|
3
|
Villani V, Frank CN, Cravedi P, Hou X, Bin S, Kamitakahara A, Barbati C, Buono R, Da Sacco S, Lemley KV, De Filippo RE, Lai S, Laviano A, Longo VD, Perin L. A kidney-specific fasting-mimicking diet induces podocyte reprogramming and restores renal function in glomerulopathy. Sci Transl Med 2024; 16:eadl5514. [PMID: 39475573 DOI: 10.1126/scitranslmed.adl5514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/24/2024] [Accepted: 09/11/2024] [Indexed: 11/04/2024]
Abstract
Cycles of a fasting-mimicking diet (FMD) promote regeneration and reduce damage in the pancreases, blood, guts, and nervous systems of mice, but their effect on kidney disease is unknown. In addition, a FMD has not been tested in rats. Here, we show that cycles of a newly developed low-salt FMD (LS-FMD) restored normal proteinuria and nephron structure and function in rats with puromycin-induced nephrosis compared with that in animals with renal damage that did not receive the dietary intervention. LS-FMD induced modulation of a nephrogenic gene program, resembling renal developmental processes in multiple kidney structures. LS-FMD also activated podocyte-lineage reprogramming pathways and promoted a quiescent state in mature podocytes in the rat kidney damage model. In a pilot clinical study in patients with chronic kidney disease, FMD cycles of 5 days each month for 3 months promoted renoprotection, including reduction of proteinuria and improved endothelial function, compared with that in patients who did not receive the FMD cycles. These results show that FMD cycles, which promote the reprogramming of multiple renal cell types and lead to glomerular damage reversal in rats, should be tested further for the treatment of progressive kidney diseases.
Collapse
Affiliation(s)
- Valentina Villani
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
| | - Camille Nicolas Frank
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Paolo Cravedi
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
| | - Xiaogang Hou
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
| | - Sofia Bin
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna 40126, Italy
| | - Anna Kamitakahara
- Division of Neurology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cristiani Barbati
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Italian National Institute of Health, Rome 00185, Italy
| | - Roberta Buono
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Stefano Da Sacco
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kevin V Lemley
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Roger E De Filippo
- GOFARR Laboratory, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Silvia Lai
- Department of Translational and Precision Medicine, Nephrology Unit, Sapienza University of Rome, Rome 00185, Italy
| | - Alessandro Laviano
- Department of Translational and Precision Medicine, Sapienza University, Rome 00185, Italy
| | - Valter D Longo
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Laura Perin
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
4
|
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
|
5
|
Gujarati NA, Chow AK, Mallipattu SK. Central role of podocytes in mediating cellular cross talk in glomerular health and disease. Am J Physiol Renal Physiol 2024; 326:F313-F325. [PMID: 38205544 PMCID: PMC11207540 DOI: 10.1152/ajprenal.00328.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes are highly specialized epithelial cells that surround the capillaries of the glomeruli in the kidney. Together with the glomerular endothelial cells, these postmitotic cells are responsible for regulating filtrate from the circulating blood with their organized network of interdigitating foot processes that wrap around the glomerular basement membrane. Although podocyte injury and subsequent loss is the hallmark of many glomerular diseases, recent evidence suggests that the cell-cell communication between podocytes and other glomerular and nonglomerular cells is critical for the development and progression of kidney disease. In this review, we highlight these key cellular pathways of communication and how they might be a potential target for therapy in glomerular disease. We also postulate that podocytes might serve as a central hub for communication in the kidney under basal conditions and in response to cellular stress, which may have implications for the development and progression of glomerular diseases.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Andrew K Chow
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Renal Section, Northport Veterans Affairs Medical Center, Northport, New York, United States
| |
Collapse
|
6
|
van der Pluijm LA, Koudijs A, Stam W, Roelofs JJ, Danser AJ, Rotmans JI, Gross KW, Pieper MP, van Zonneveld AJ, Bijkerk R. SGLT2 inhibition promotes glomerular repopulation by cells of renin lineage in experimental kidney disease. Acta Physiol (Oxf) 2024; 240:e14108. [PMID: 38314444 PMCID: PMC10923162 DOI: 10.1111/apha.14108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/06/2024]
Abstract
AIM Sodium glucose co-transporter-2 (SGLT2) inhibitors stimulate renal excretion of sodium and glucose and exert renal protective effects in patients with (non-)diabetic chronic kidney disease (CKD) and may as well protect against acute kidney injury (AKI). The mechanism behind this kidney protective effect remains unclear. Juxtaglomerular cells of renin lineage (CoRL) have been demonstrated to function as progenitors for multiple adult glomerular cell types in kidney disease. This study assesses the impact of SGLT2 inhibition on the repopulation of glomerular cells by CoRL and examines their phenotypic commitment. METHODS Experiments were performed in Ren1cre-tdTomato lineage-trace mice. Either 5/6 nephrectomy (5/6NX) modeling CKD or bilateral ischaemia reperfusion injury (bIRI) mimicking AKI was applied, while the SGLT2 inhibitor empagliflozin (10 mg/kg) was administered daily via oral gavage for 14 days. RESULTS Both 5/6NX and bIRI-induced kidney injury increased the number of glomerular CoRL-derived cells. SGLT2 inhibition improved kidney function after 5/6NX, indicated by decreased blood creatinine and urea levels, but not after bIRI. In line with this, empagliflozin in 5/6NX animals resulted in less glomerulosclerosis, while it did not affect histopathological features in bIRI. Treatment with empagliflozin resulted in an increase in the number of CoRL-derived glomerular cells in both 5/6NX and bIRI conditions. Interestingly, SGLT2 inhibition led to more CoRL-derived podocytes in 5/6NX animals, whereas empagliflozin-treated bIRI mice presented with increased levels of parietal epithelial and mesangial cells derived from CoRL. CONCLUSION We conclude that SGLT2 inhibition by empagliflozin promotes CoRL-mediated glomerular repopulation with selective CoRL-derived cell types depending on the type of experimental kidney injury. These findings suggest a previously unidentified mechanism that could contribute to the renoprotective effect of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Loïs A.K. van der Pluijm
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Angela Koudijs
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Joris J.T.H. Roelofs
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - A.H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joris I. Rotmans
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael P. Pieper
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
7
|
Li T, Bao Y, Xia Y, Meng H, Zhou C, Huang L, Wang X, Lai EY, Jiang P, Mao J. Loss of MTX2 causes mitochondrial dysfunction, podocyte injury, nephrotic proteinuria and glomerulopathy in mice and patients. Int J Biol Sci 2024; 20:937-952. [PMID: 38250156 PMCID: PMC10797693 DOI: 10.7150/ijbs.89916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Proteinuria is a common and important clinical manifestation of chronic kidney disease (CKD) and an independent risk factor for the progression of kidney disease. As a component of the glomerular filtration barrier (GFB), podocyte plays a key role in the pathogenesis of glomerular diseases and proteinuria. However, the pathophysiology of glomerular diseases associated with mitochondrial function is incompletely understood. Here, we identified three novel mutations in MTX2, encoding a membrane protein in mitochondria, associated with multisystem manifestations including nephrotic proteinuria and kidney injury in two Chinese patients. Conditional podocyte-specific Mtx2 knockout (Pod-Mtx2-KO) mice present a series of podocyte and glomerular abnormalities from 8 weeks to old age, including microalbuminuria, glomerular mesangial hyperplasia, fusion and effacement of foot process. MTX2 deficiency impaired podocyte functions in vitro, manifested by reductions of adhesion, migration and endocytosis, which were further restored by overexpression of MTX2. Moreover, MTX2 defects led to abnormal mitochondrial structure and dysfunction, evidenced with defects of complex I and III, increased production of reactive oxygen species (ROS), and decreased protein levels of Sam50-CHCHD3-Mitofilin axis in the mitochondrial intermembrane space bridging (MIB) complex which is responsible for maintaining mitochondrial cristae morphology. Collectively, these findings reveal that the normal expression of MTX2 in glomerulus plays an important role in the adhesion, migration, endocytosis, proliferation and other physiological functions of podocytes, which may be realized by maintaining the morphological structure and function of mitochondria. Abnormal expression of MTX2 can lead to mitochondrial dysfunction and structural abnormalities by Sam50-CHCHD3-Mitofilin axis in podocyte, which further induces podocyte injury, glomerular lesions and proteinuria.
Collapse
Affiliation(s)
- Ting Li
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Ying Bao
- Department of Pediatric Nephrology, Xi'an Children's Hospital, The Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Xia
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Hanyan Meng
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Chao Zhou
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Limin Huang
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Xiaowen Wang
- Department of Pediatric Nephrology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - En Yin Lai
- Kidney Disease Center of the First Affiliated Hospital and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Pingping Jiang
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Institute of Pharmaceutical Biotechnology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| |
Collapse
|
8
|
Liu C, Cheng Q, Ao Q, Yang G, Liu Y, Zhao J. Induced pluripotent stem cells-podocytes promote repair in acute kidney injury is dependent on Mafb/CCR5/Nampt axis-mediated M2 macrophage polarization. Chem Biol Interact 2023; 380:110534. [PMID: 37182688 DOI: 10.1016/j.cbi.2023.110534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have been the focus of cellular therapy studies. The use of iPSCs in regenerative medicine is limited by their tumorigenic potential. This study sought to determine whether iPSCs-derived podocytes attenuate acute kidney injury (AKI) and the molecular mechanism. Inoculation of iPSCs-podocytes significantly promoted the repair of kidney injury in AKI mice, reduced the levels of kidney injury factors Scr, BUN, and urinary NAG, and alleviated the inflammatory response. Histological analysis revealed a significant increase in the number of M2 macrophages and a significant decrease in M1 macrophages in the kidney tissues. Subsequently, the genes and signaling pathways that may be associated with kidney injury repair in mice were analyzed by RNA-seq and bioinformatics prediction. The polarization of M2 macrophages was promoted by MAF bZIP transcription factor B (Mafb)-mediated activation of C-C motif chemokine receptor 5 (Ccr5) and nicotinamide phosphoribosyltransferase (Nampt) signaling pathway. Taken together, these results show that iPSCs-podocytes depend on Mafb to activate the Nampt signaling pathway through transcriptional activation of Ccr5, thereby promoting the repair of AKI caused by ischemia-reperfusion.
Collapse
Affiliation(s)
- Chang Liu
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Qingli Cheng
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Qiangguo Ao
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Guang Yang
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Yang Liu
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Jiahui Zhao
- Department of Nephrology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, PR China.
| |
Collapse
|
9
|
Bharati J, Chander PN, Singhal PC. Parietal Epithelial Cell Behavior and Its Modulation by microRNA-193a. Biomolecules 2023; 13:266. [PMID: 36830635 PMCID: PMC9953542 DOI: 10.3390/biom13020266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Glomerular parietal epithelial cells (PECs) have been increasingly recognized to have crucial functions. Lineage tracking in animal models showed the expression of a podocyte phenotype by PECs during normal glomerular growth and after acute podocyte injury, suggesting a reparative role of PECs. Conversely, activated PECs are speculated to be pathogenic and comprise extracapillary proliferation in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CrescGN). The reparative and pathogenic roles of PECs seem to represent two sides of PEC behavior directed by the local milieu and mediators. Recent studies suggest microRNA-193a (miR193a) is involved in the pathogenesis of FSGS and CrescGN. In a mouse model of primary FSGS, the induction of miR193a caused the downregulation of Wilms' tumor protein, leading to the dedifferentiation of podocytes. On the other hand, the inhibition of miR193a resulted in reduced crescent lesions in a mouse model of CrescGN. Interestingly, in vitro studies report that the downregulation of miR193a induces trans-differentiation of PECs into a podocyte phenotype. This narrative review highlights the critical role of PEC behavior in health and during disease and its modulation by miR193a.
Collapse
Affiliation(s)
- Joyita Bharati
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Department of Medicine, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY 11549, USA
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Praveen N. Chander
- New York Medical College, Touro College and University System Valhalla, Valhalla, NY 10595, USA
| | - Pravin C. Singhal
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Department of Medicine, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
10
|
Kumar S, Fan X, Rasouly HM, Sharma R, Salant DJ, Lu W. ZEB2 controls kidney stromal progenitor differentiation and inhibits abnormal myofibroblast expansion and kidney fibrosis. JCI Insight 2023; 8:e158418. [PMID: 36445780 PMCID: PMC9870089 DOI: 10.1172/jci.insight.158418] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
FOXD1+ cell-derived stromal cells give rise to pericytes and fibroblasts that support the kidney vasculature and interstitium but are also major precursors of myofibroblasts. ZEB2 is a SMAD-interacting transcription factor that is expressed in developing kidney stromal progenitors. Here we show that Zeb2 is essential for normal FOXD1+ stromal progenitor development. Specific conditional knockout of mouse Zeb2 in FOXD1+ stromal progenitors (Zeb2 cKO) leads to abnormal interstitial stromal cell development, differentiation, and kidney fibrosis. Immunofluorescent staining analyses revealed abnormal expression of interstitial stromal cell markers MEIS1/2/3, CDKN1C, and CSPG4 (NG2) in newborn and 3-week-old Zeb2-cKO mouse kidneys. Zeb2-deficient FOXD1+ stromal progenitors also took on a myofibroblast fate that led to kidney fibrosis and kidney failure. Cell marker studies further confirmed that these myofibroblasts expressed pericyte and resident fibroblast markers, including PDGFRβ, CSPG4, desmin, GLI1, and NT5E. Notably, increased interstitial collagen deposition associated with loss of Zeb2 in FOXD1+ stromal progenitors was accompanied by increased expression of activated SMAD1/5/8, SMAD2/3, SMAD4, and AXIN2. Thus, our study identifies a key role of ZEB2 in maintaining the cell fate of FOXD1+ stromal progenitors during kidney development, whereas loss of ZEB2 leads to differentiation of FOXD1+ stromal progenitors into myofibroblasts and kidney fibrosis.
Collapse
|
11
|
Narayan P, Bruce AT, Rivera EA, Bertram TA, Jain D. Selected renal cells harbor nephrogenic potential. Front Med (Lausanne) 2022; 9:1062890. [PMID: 36619635 PMCID: PMC9815697 DOI: 10.3389/fmed.2022.1062890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Selected renal cells (SRCs), a renal epithelial cell-enriched platform, are being advanced as an autologous cell-based therapy for the treatment of chronic kidney disease. However, the mechanism underlying its renal reparative and restorative effects remains to be fully elucidated. In this study, we coupled knowledgebase data with empirical findings to demonstrate that genes differentially expressed by SRCs form interactomes within tubules and glomeruli and mediate a suite of renal developmental activities including epithelial cell differentiation, renal vasculature development, and glomerular and nephron development. In culture, SRCs form organoids which self-assemble into tubules in the presence of a scaffold. Implanted into the kidneys of subtotally nephrectomized rats, SRCs are associated with comma- and S-shaped body cell formation and glomerular development, and improvement in renal filtration indices and renal microarchitecture. These data suggest that SRCs harbor nephrogenic potential, which may explain, at least in part, their therapeutic activity.
Collapse
|
12
|
Tøndel C, Thurberg BL, DasMahapatra P, Lyn N, Maski M, Batista JL, George K, Patel H, Hariri A. Clinical relevance of globotriaosylceramide accumulation in Fabry disease and the effect of agalsidase beta in affected tissues. Mol Genet Metab 2022; 137:328-341. [PMID: 36334424 DOI: 10.1016/j.ymgme.2022.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Fabry disease (FD) is a rare lysosomal storage disorder, characterized by a reduction in α-galactosidase A enzyme activity and the progressive accumulation of globotriaosylceramide (GL3) and its metabolites in the cells of various organs. Agalsidase beta, an enzyme replacement therapy (ERT), is approved for use in patients with FD in Europe, Canada, Australia, South America, and Asia, and is the only ERT approved for use in the United States. In this review, we discuss the clinical relevance of GL3 accumulation, the effect of agalsidase beta on GL3 in target tissues, and the association between treatment-related tissue GL3 clearance and long-term structure, function, or clinical outcomes. Accumulation of GL3 in the kidney, heart, vasculature, neurons, skin, gastrointestinal tract and auditory system correlates to cellular damage and irreversible organ damage, as a result of sclerosis, fibrosis, apoptosis, inflammation, and endothelial dysfunction. Damage leads to renal dysfunction and end-stage renal disease; myocardial hypertrophy with heart failure and arrhythmias; ischemic stroke; neuropathic pain; skin lesions; intestinal ischemia and dysmotility; and hearing loss. Treatment with agalsidase beta is effective in substantially clearing GL3 in a range of cells from the tissues affected by FD. Agalsidase beta has also been shown to slow renal decline and lower the overall risk of clinical progression, demonstrating an indirect link between treatment-related GL3 clearance and stabilization of FD.
Collapse
Affiliation(s)
- Camilla Tøndel
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | - Kelly George
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurological Diseases Therapeutic Area, Sanofi, Cambridge, MA, USA
| | | | | |
Collapse
|
13
|
Molecular Mechanisms of Kidney Injury and Repair. Int J Mol Sci 2022; 23:ijms23031542. [PMID: 35163470 PMCID: PMC8835923 DOI: 10.3390/ijms23031542] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) will become the fifth global cause of death by 2040, thus emphasizing the need to better understand the molecular mechanisms of damage and regeneration in the kidney. CKD predisposes to acute kidney injury (AKI) which, in turn, promotes CKD progression. This implies that CKD or the AKI-to-CKD transition are associated with dysfunctional kidney repair mechanisms. Current therapeutic options slow CKD progression but fail to treat or accelerate recovery from AKI and are unable to promote kidney regeneration. Unraveling the cellular and molecular mechanisms involved in kidney injury and repair, including the failure of this process, may provide novel biomarkers and therapeutic tools. We now review the contribution of different molecular and cellular events to the AKI-to-CKD transition, focusing on the role of macrophages in kidney injury, the different forms of regulated cell death and necroinflammation, cellular senescence and the senescence-associated secretory phenotype (SAPS), polyploidization, and podocyte injury and activation of parietal epithelial cells. Next, we discuss key contributors to repair of kidney injury and opportunities for their therapeutic manipulation, with a focus on resident renal progenitor cells, stem cells and their reparative secretome, certain macrophage subphenotypes within the M2 phenotype and senescent cell clearance.
Collapse
|
14
|
Ni L, Yuan C, Wu X. The recruitment mechanisms and potential therapeutic targets of podocytes from parietal epithelial cells. J Transl Med 2021; 19:441. [PMID: 34674704 PMCID: PMC8529729 DOI: 10.1186/s12967-021-03101-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/01/2021] [Indexed: 01/02/2023] Open
Abstract
Podocytes are differentiated postmitotic cells which cannot be replaced after podocyte injury. The mechanism of podocyte repopulation after injury has aroused wide concern. Parietal epithelial cells (PECs) are heterogeneous and only a specific subpopulation of PECs has the capacity to replace podocytes. Major progress has been achieved in recent years regarding the role and function of a subset of PECs which could transdifferentiate toward podocytes. Additionally, several factors, such as Notch, Wnt/ß-catenin, Wilms’ tumor-1, miR-193a and growth arrest-specific protein 1, have been shown to be involved in these processes. Finally, PECs serve as a potential therapeutic target in the conditions of podocyte loss. In this review, we discuss the latest observations and concepts about the recruitment of podocytes from PECs in glomerular diseases as well as newly identified mechanisms and the most recent treatments for this process.
Collapse
Affiliation(s)
- Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Cheng Yuan
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
15
|
Stein MC, Braun F, Krebs CF, Bunders MJ. Kidney organoid systems for studies of immune-mediated kidney diseases: challenges and opportunities. Cell Tissue Res 2021; 385:457-473. [PMID: 34309728 PMCID: PMC8310776 DOI: 10.1007/s00441-021-03499-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022]
Abstract
Acute and chronic kidney diseases are major contributors to morbidity and mortality in the global population. Many nephropathies are considered to be immune-mediated with dysregulated immune responses playing an important role in the pathogenesis. At present, targeted approaches for many kidney diseases are still lacking, as the underlying mechanisms remain insufficiently understood. With the recent development of organoids—a three-dimensional, multicellular culture system, which recapitulates important aspects of human tissues—new opportunities to investigate interactions between renal cells and immune cells in the pathogenesis of kidney diseases arise. To date, kidney organoid systems, which reflect the structure and closer resemble critical aspects of the organ, have been established. Here, we highlight the recent advances in the development of kidney organoid models, including pluripotent stem cell-derived kidney organoids and primary epithelial cell-based tubuloids. The employment and further required advances of current organoid models are discussed to investigate the role of the immune system in renal tissue development, regeneration, and inflammation to identify targets for the development of novel therapeutic approaches of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Melissa C Stein
- Research Department Virus Immunology, Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madeleine J Bunders
- Research Department Virus Immunology, Leibniz-Institute for Experimental Virology, Hamburg, Germany.
| |
Collapse
|
16
|
Sobreiro-Almeida R, Melica ME, Lasagni L, Romagnani P, Neves NM. Retinoic Acid Benefits Glomerular Organotypic Differentiation from Adult Renal Progenitor Cells In Vitro. Stem Cell Rev Rep 2021; 17:1406-1419. [PMID: 33538982 DOI: 10.1007/s12015-021-10128-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
When in certain culture conditions, organotypic cultures are able to mimic developmental stages of an organ, generating higher-order structures containing functional subunits and progenitor niches. Despite the major advances in the area, researchers have not been able to fully recapitulate the complexity of kidney tissue. Pluripotent stem cells are extensively used in the field, but very few studies make use of adult stem cells. Herein, we describe a simple and feasible method for achieving glomerular epithelial differentiation on an organotypic model comprising human renal progenitor cells from adult kidney (hRPCs). Their glomerular differentiative potential was studied using retinoic acid (RA), a fundamental molecule for intermediate mesoderm induction on early embryogenesis. Immunofluorescence, specific cell surface markers expression and gene expression analysis confirm the glomerular differentiative potential of RA in a short-term culture. We also compared the potential of RA with a potent WNT agonist, CHIR99021, on the differentiative capacity of hRPCs. Gene expression and immunofluorescence analysis confirmed that hRPCs are more sensitive to RA stimulation when compared to CHIR9901. Endothelial cells were also included on the spheroids, resulting in a higher organizational level. The assembly potential of these cells and their selective stimulation will give new insights on adult organotypic cell culture studies and will hopefully guide more works in this important area of research.
Collapse
Affiliation(s)
- Rita Sobreiro-Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Elena Melica
- Department of Biomedical, Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Lasagni
- Department of Biomedical, Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
17
|
Kaverina NV, Eng DG, Miner JH, Pippin JW, Shankland SJ. Parietal epithelial cell differentiation to a podocyte fate in the aged mouse kidney. Aging (Albany NY) 2020; 12:17601-17624. [PMID: 32858527 PMCID: PMC7521511 DOI: 10.18632/aging.103788] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Healthy aging is typified by a progressive and absolute loss of podocytes over the lifespan of animals and humans. To test the hypothesis that a subset of glomerular parietal epithelial cell (PEC) progenitors transition to a podocyte fate with aging, dual reporter PEC-rtTA|LC1|tdTomato|Nphs1-FLPo|FRT-EGFP mice were generated. PECs were inducibly labeled with a tdTomato reporter, and podocytes were constitutively labeled with an EGFP reporter. With advancing age (14 and 24 months) glomeruli in the juxta-medullary cortex (JMC) were more severely injured than those in the outer cortex (OC). In aged mice (24m), injured glomeruli with lower podocyte number (41% decrease), showed more PEC migration and differentiation to a podocyte fate than mildly injured or healthy glomeruli. PECs differentiated to a podocyte fate had ultrastructural features of podocytes and co-expressed the podocyte markers podocin, nephrin, p57 and VEGF164, but not markers of mesangial (Perlecan) or endothelial (ERG) cells. PECs differentiated to a podocyte fate did not express CD44, a marker of PEC activation. Taken together, we demonstrate that a subpopulation of PECs differentiate to a podocyte fate predominantly in injured glomeruli in mice of advanced age.
Collapse
Affiliation(s)
| | - Diana G. Eng
- Division of Nephrology, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey H. Miner
- Division of Nephrology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jeffrey W. Pippin
- Division of Nephrology, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
18
|
Understanding Mesangial Pathobiology in AL-Amyloidosis and Monoclonal Ig Light Chain Deposition Disease. Kidney Int Rep 2020; 5:1870-1893. [PMID: 33163710 PMCID: PMC7609979 DOI: 10.1016/j.ekir.2020.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/06/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
Patients with plasma cell dyscrasias produce free abnormal monoclonal Ig light chains that circulate in the blood stream. Some of them, termed glomerulopathic light chains, interact with the mesangial cells and trigger, in a manner dependent of their structural and physicochemical properties, a sequence of pathological events that results in either light chain–derived (AL) amyloidosis (AL-Am) or light chain deposition disease (LCDD). The mesangial cells play a key role in the pathogenesis of both diseases. The interaction with the pathogenic light chain elicits specific cellular processes, which include apoptosis, phenotype transformation, and secretion of extracellular matrix components and metalloproteinases. Monoclonal light chains associated with AL-Am but not those producing LCDD are avidly endocytosed by mesangial cells and delivered to the mature lysosomal compartment where amyloid fibrils are formed. Light chains from patients with LCDD exert their pathogenic signaling effect at the cell surface of mesangial cells. These events are generic mesangial responses to a variety of adverse stimuli, and they are similar to those characterizing other more frequent glomerulopathies responsible for many cases of end-stage renal disease. The pathophysiologic events that have been elucidated allow to propose future therapeutic approaches aimed at preventing, stopping, ameliorating, or reversing the adverse effects resulting from the interactions between glomerulopathic light chains and mesangium.
Collapse
|
19
|
Wu D, Bai J, Cui S, Fu B, Yin Z, Cai G, Chen X. Renal progenitor cells modulated by angiotensin II receptor blocker (ARB) medication and differentiation towards podocytes in anti-thy1.1 nephritis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:355. [PMID: 32355799 PMCID: PMC7186716 DOI: 10.21037/atm.2020.02.58] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Mesangial proliferative glomerulonephritis (MsPGN) is an epidemic disease with increasing occurrence. As important as mesangial cells, podocytes are key innate cells for MsPGN prognosis and recovery. Renal progenitor cells, located at the urinary pole (UP) of Bowman’s capsule (BC), could alleviate kidney injury through their capacity to differentiate into podocytes. Methods Seventy-two male rats were categorized randomly into the sham (n=24), untreated Thy-1 (n=24) and losartan-treated (n=24) groups. We administered vehicle or losartan (50 mg/kg by gavage) daily to treat rats with anti-thy1.1 nephritis, an ideal model to simulate human MsPGN. Two weeks after the intravenous injection of antibody, urinary protein and blood samples were analyzed, pathological changes were examined, the number of podocytes was determined, and renal progenitor cells were studied. Results Anti-thy1.1 nephritis was significantly alleviated after losartan treatment, as reported previously and as expected. Compared with the untreated Thy-1 group, the number of podocytes in the losartan group increased, and the area of renal progenitor cells significantly increased. The protein expression of components of the p-ERK pathway was determined during the development of renal progenitor cells differentiating into podocytes. Conclusions The data in this paper show the direct glomerular cell action of angiotensin II receptor blocker (ARB) treatment in improving outcomes in anti-thy1.1 nephritis. The positive effects of ARB medication on anti-thy1.1 nephritis were due to an increase in the number of renal epithelial progenitor cells (defined as PECs that expressed only stem cell markers without podocyte proteins).
Collapse
Affiliation(s)
- Di Wu
- Medical School of Chinese PLA, Beijing 100853, China.,Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Jiuxu Bai
- Medical School of Chinese PLA, Beijing 100853, China.,Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Zhiwei Yin
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xiangmei Chen
- Medical School of Chinese PLA, Beijing 100853, China.,Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| |
Collapse
|
20
|
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: 47] [Impact Index Per Article: 7.8] [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
|
21
|
Liu GW, Johnson SL, Jain R, Peeler DJ, Shankland SJ, Pun SH. Optimized nonviral gene delivery for primary urinary renal progenitor cells to enhance cell migration. J Biomed Mater Res A 2019; 107:2718-2725. [PMID: 31404486 DOI: 10.1002/jbm.a.36775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 07/31/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Progressive loss of glomerular podocytes during kidney disease leads to irreversible kidney failure, and is exacerbated by the fact that podocytes are terminally differentiated epithelial cells and unable to proliferate. Regeneration of lost podocytes must therefore derive from nonpodocyte sources. Human urine-derived renal progenitor cells (uRPCs) are attractive podocyte progenitors for cell therapy applications due to their availability from patient urine and ability to migrate to injured glomeruli and differentiate into de novo podocytes after intravenous administration. Because gene delivery has emerged as an important strategy to augment the functionality and survival of cell therapies prior to injection, in this work we optimized nonviral gene delivery conditions (cell density, DNA dose, % FBS, and transfection material composition) to primary uRPCs. Using the cationic polymer-peptide conjugate VIPER for gene delivery and the Sleeping Beauty transposon/transposase constructs for gene integration, we optimized transfection parameters to achieve efficient transgene expression (up to 55% transfected cells) and stable transgene expression (>65% integration efficiency) lasting up to 10 days. With these methods, we transfected uRPCs to overexpress CXCR4, an important chemokine receptor that mediates uRPC migration to the kidneys after intravenous injection, and demonstrate that CXCR4-uRPCs exhibit enhanced migration compared to mock-transfected cells.
Collapse
Affiliation(s)
- Gary W Liu
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Soren L Johnson
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Ritika Jain
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - David J Peeler
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| | - Stuart J Shankland
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, Washington
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington
| |
Collapse
|
22
|
Altintas MM, Reiser J. Podocytes: Way to Go. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:226-228. [PMID: 30543788 DOI: 10.1016/j.ajpath.2018.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/28/2018] [Indexed: 12/28/2022]
Abstract
This commentary highlights the article by Hara et al that discusses the clinical implications of mitotic catastrophe in podocyte health during diabetic kidney disease.
Collapse
Affiliation(s)
- Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, Illinois.
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
23
|
Prochnicki A, Amann K, Wegner M, Sock E, Pfister E, Shankland S, Pippin J, Daniel C. Characterization of Glomerular Sox9+ Cells in Anti-Glomerular Basement Membrane Nephritis in the Rat. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2529-2541. [DOI: 10.1016/j.ajpath.2018.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
|
24
|
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
|
25
|
Kaverina NV, Eng DG, Largent AD, Daehn I, Chang A, Gross KW, Pippin JW, Hohenstein P, Shankland SJ. WT1 Is Necessary for the Proliferation and Migration of Cells of Renin Lineage Following Kidney Podocyte Depletion. Stem Cell Reports 2017; 9:1152-1166. [PMID: 28966119 PMCID: PMC5639431 DOI: 10.1016/j.stemcr.2017.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
Wilms' tumor suppressor 1 (WT1) plays an important role in cell proliferation and mesenchymal-epithelial balance in normal development and disease. Here, we show that following podocyte depletion in three experimental models, and in patients with focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, WT1 increased significantly in cells of renin lineage (CoRL). In an animal model of FSGS in RenWt1fl/fl reporter mice with inducible deletion of WT1 in CoRL, CoRL proliferation and migration to the glomerulus was reduced, and glomerular disease was worse compared with wild-type mice. To become podocytes, CoRL undergo mesenchymal-to-epithelial transformation (MET), typified by reduced staining for mesenchymal markers (MYH11, SM22, αSMA) and de novo expression of epithelial markers (E-cadherin and cytokeratin18). Evidence for changes in MET markers was barely detected in RenWt1fl/fl mice. Our results show that following podocyte depletion, WT1 plays essential roles in CoRL proliferation and migration toward an adult podocyte fate.
Collapse
Affiliation(s)
- Natalya V Kaverina
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Andrea D Largent
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Ilse Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA
| | - Anthony Chang
- Department of Pathology, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
26
|
Abstract
Podocytes, the postmitotic and highly branched epithelial cells of the glomerulus, play a pivotal role for the function of the glomerular filtration barrier and the development of chronic kidney disease. It has long been discussed whether podocytes in vivo are motile and can laterally migrate in a coordinated way along the capillaries until they reach the position of naked glomerular basement membrane often found in podocytopathies. Such motility would also be the prerequisite for the replacement of lost podocytes by progenitor cells. Additionally, the change of the podocyte foot processes from a normal to an effaced morphology, like it is found in many kidney diseases, would require a dynamic behavior of podocytes. Since the actin cytoskeleton is expressed in podocytes in vitro and in vivo and the morphology of podocytes is highly dependent on actin, actin-associated, and actin-regulating proteins, it was assumed that podocytes are dynamic and motile. After earlier technical limitations had been overcome and novel microscopic techniques like multiphoton microscopy had been developed, it became possible to continuously study the behavior of podocytes in living rodents and zebrafish larvae under physiological and pathological conditions. Recent in vivo microscopic studies in different model organisms suggest that lateral migration of podocytes in situ is a very unlikely event and only dynamic apical cell protrusions can be observed under pathological conditions. This review discusses recent findings concerning different forms of motility (like lateral translocative (LTM), apical translocative (ATM), and stationary motility (SM)) and their role for podocytopathies.
Collapse
Affiliation(s)
- Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany. .,Institut für Anatomie und Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler-Str. 23c, 17487, Greifswald, Germany.
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
| |
Collapse
|
27
|
Kaverina NV, Kadoya H, Eng DG, Rusiniak ME, Sequeira-Lopez MLS, Gomez RA, Pippin JW, Gross KW, Peti-Peterdi J, Shankland SJ. Tracking the stochastic fate of cells of the renin lineage after podocyte depletion using multicolor reporters and intravital imaging. PLoS One 2017; 12:e0173891. [PMID: 28329012 PMCID: PMC5362207 DOI: 10.1371/journal.pone.0173891] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Podocyte depletion plays a major role in focal segmental glomerular sclerosis (FSGS). Because cells of the renin lineage (CoRL) serve as adult podocyte and parietal epithelial cell (PEC) progenitor candidates, we generated Ren1cCre/R26R-ConfettiTG/WT and Ren1dCre/R26R-ConfettiTG/WT mice to determine CoRL clonality during podocyte replacement. Four CoRL reporters (GFP, YFP, RFP, CFP) were restricted to cells in the juxtaglomerular compartment (JGC) at baseline. Following abrupt podocyte depletion in experimental FSGS, all four CoRL reporters were detected in a subset of glomeruli at day 28, where they co-expressed de novo four podocyte proteins (podocin, nephrin, WT-1 and p57) and two glomerular parietal epithelial cell (PEC) proteins (claudin-1, PAX8). To monitor the precise migration of a subset of CoRL over a 2w period following podocyte depletion, intravital multiphoton microscopy was used. Our findings demonstrate direct visual support for the migration of single CoRL from the JGC to the parietal Bowman's capsule, early proximal tubule, mesangium and glomerular tuft. In summary, these results suggest that following podocyte depletion, multi-clonal CoRL migrate to the glomerulus and replace podocyte and PECs in experimental FSGS.
Collapse
Affiliation(s)
- Natalya V. Kaverina
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, United States of America
| | - Hiroyuki Kadoya
- Department of Physiology & Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Japan
| | - Diana G. Eng
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, United States of America
| | - Michael E. Rusiniak
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Maria Luisa S. Sequeira-Lopez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - R. Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Jeffrey W. Pippin
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, United States of America
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Janos Peti-Peterdi
- Department of Physiology & Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- * E-mail: (SJS); (JPP)
| | - Stuart J. Shankland
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, United States of America
- * E-mail: (SJS); (JPP)
| |
Collapse
|
28
|
XU Z, FAN J. Islet transplantation promotes podocyte regeneration in a model of diabetic nephropathy. Turk J Med Sci 2017; 47:1925-1930. [DOI: 10.3906/sag-1704-102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
29
|
Fall B, Scott CR, Mauer M, Shankland S, Pippin J, Jefferson JA, Wallace E, Warnock D, Najafian B. Urinary Podocyte Loss Is Increased in Patients with Fabry Disease and Correlates with Clinical Severity of Fabry Nephropathy. PLoS One 2016; 11:e0168346. [PMID: 27992580 PMCID: PMC5161377 DOI: 10.1371/journal.pone.0168346] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 11/29/2016] [Indexed: 11/18/2022] Open
Abstract
Chronic kidney disease is a major complication of Fabry disease. Podocytes accumulate globotriaosylceramide inclusions more than other kidney cell types in Fabry patients. Podocyte injury occurs early in age, and is progressive. Since injured podocytes detach into the urine (podocyturia), we hypothesized that podocyturia would increase in Fabry patients and correlate with clinical severity of Fabry nephropathy. Urine specimens from 39 Fabry patients and 24 healthy subjects were evaluated for podocyturia. Most of the Fabry patients and many healthy subjects had podocyturia. The number of podocytes per gram of urine creatinine (UPodo/g Cr) was 3.6 fold greater in Fabry patients (3,741 ± 2796; p = 0.001) than healthy subjects (1,040 ± 972). Fabry patients with normoalbuminuria and normoproteinuria had over 2-fold greater UPodo/g Cr than healthy subjects (p = 0.048). UPodo/gCr was inversely related to eGFR in male patients (r = -0.69, p = 0.003). UPodo/gCr was directly related to urine protein creatinine ratio (r = 0.33; p = 0.04) in all Fabry patients. These studies confirm increased podocyturia in Fabry disease, even when proteinuria and albuminuria are absent. Podocyturia correlates with clinical severity of Fabry nephropathy, and potentially may be of prognostic value.
Collapse
Affiliation(s)
- Brent Fall
- Department of Pathology, University of Washington, Seattle, United States America
| | - C Ronald Scott
- Department of Pediatrics, University of Washington, Seattle, United States America
| | - Michael Mauer
- Departments of Pediatrics, University of Minnesota, Minneapolis, United States America.,Departments of Medicine, University of Minnesota, Minneapolis, United States America
| | - Stuart Shankland
- Department of Medicine, University of Washington, Seattle, United States America
| | - Jeffrey Pippin
- Department of Medicine, University of Washington, Seattle, United States America
| | - Jonathan A Jefferson
- Department of Medicine, University of Washington, Seattle, United States America
| | - Eric Wallace
- Department of Medicine, University of Alabama, Birmingham, United States America
| | - David Warnock
- Department of Medicine, University of Alabama, Birmingham, United States America
| | - Behzad Najafian
- Department of Pathology, University of Washington, Seattle, United States America
| |
Collapse
|
30
|
Embry AE, Mohammadi H, Niu X, Liu L, Moe B, Miller-Little WA, Lu CY, Bruggeman LA, McCulloch CA, Janmey PA, Miller RT. Biochemical and Cellular Determinants of Renal Glomerular Elasticity. PLoS One 2016; 11:e0167924. [PMID: 27942003 PMCID: PMC5152842 DOI: 10.1371/journal.pone.0167924] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/22/2016] [Indexed: 12/22/2022] Open
Abstract
The elastic properties of renal glomeruli and their capillaries permit them to maintain structural integrity in the presence of variable hemodynamic forces. Measured by micro-indentation, glomeruli have an elastic modulus (E, Young's modulus) of 2.1 kPa, and estimates from glomerular perfusion studies suggest that the E of glomeruli is between 2 and 4 kPa. F-actin depolymerization by latrunculin, inhibition of acto-myosin contractility by blebbistatin, reduction in ATP synthesis, and reduction of the affinity of adhesion proteins by EDTA reduced the glomerular E to 1.26, 1.7, 1.5, and 1.43 kPa, respectively. Actin filament stabilization with jasplakinolide and increasing integrin affinity with Mg2+ increased E to 2.65 and 2.87 kPa, respectively. Alterations in glomerular E are reflected in commensurate changes in F/G actin ratios. Disruption of vimentin intermediate filaments by withaferin A reduced E to 0.92 kPa. The E of decellularized glomeruli was 0.74 kPa, indicating that cellular components of glomeruli have dominant effects on their elasticity. The E of glomerular basement membranes measured by magnetic bead displacement was 2.4 kPa. Podocytes and mesangial cells grown on substrates with E values between 3 and 5 kPa had actin fibers and focal adhesions resembling those of podocytes in vivo. Renal ischemia and ischemia-reperfusion reduced the E of glomeruli to 1.58 kPa. These results show that the E of glomeruli is between 2 and 4 kPa. E of the GBM, 2.4 kPa, is consistent with this value, and is supported by the behavior of podocytes and mesangial cells grown on variable stiffness matrices. The podocyte cytoskeleton contributes the major component to the overall E of glomeruli, and a normal E requires ATP synthesis. The reduction in glomerular E following ischemia and in other diseases indicates that reduced glomerular E is a common feature of many forms of glomerular injury and indicative of an abnormal podocyte cytoskeleton.
Collapse
Affiliation(s)
- Addie E. Embry
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - Hamid Mohammadi
- Center for Matrix Biology, University of Toronto, Toronto, Ontario, Canada
| | - Xinying Niu
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - Liping Liu
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - Borren Moe
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - William A. Miller-Little
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - Christopher Y. Lu
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
| | - Leslie A. Bruggeman
- Nephrology, MetroHealth Medical Center, Case-Western Reserve University, Cleveland, Ohio, United States of America
| | | | - Paul A. Janmey
- Physiology and Biophysics, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - R. Tyler Miller
- Nephrology and Medicine, U.T. Southwestern Medical School, Dallas, Texas, United States of America
- Medicine, Dallas VAMC, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
31
|
Eymael J, Smeets B. Origin and fate of the regenerating cells of the kidney. Eur J Pharmacol 2016; 790:62-73. [DOI: 10.1016/j.ejphar.2016.07.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/08/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
|
32
|
IFI27 Is a Useful Genetic Marker for Diagnosis of Immunoglobulin A Nephropathy and Membranous Nephropathy Using Peripheral Blood. PLoS One 2016; 11:e0153252. [PMID: 27100186 PMCID: PMC4839700 DOI: 10.1371/journal.pone.0153252] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/26/2016] [Indexed: 11/19/2022] Open
Abstract
Diagnosis of chronic glomerulonephritis (CGN) depends primarily on renal biopsy, which is expensive and requires hospitalization, creating a demand for noninvasive diagnostic method for this disease. We used DNA microarray analysis to search for genes whose expression levels in peripheral blood mononuclear cells (PBMCs) could distinguish between patients with CGN and healthy volunteers (HVs). We selected immunoglobulin A nephropathy (IgAN) and membranous nephropathy (MN) as typical forms of CGN. The mRNA level of the gene encoding interferon (IFN)-alpha-inducible protein 27, IFI27, which is preferentially expressed in podocytes of glomeruli, was lower in PBMCs of IgAN and MN patients than in those of HVs. This result was confirmed by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Moreover, qRT-PCR analysis revealed that the IFI27 mRNA level was reduced in PBMCs of patients with other types of chronic glomerulonephritis. IFI27 immunohistochemical staining of biopsied specimens also confirmed reduced expression of IFI27 protein in IgAN and MN patients. Based on these results, we propose that IFI27 could serve as a noninvasive diagnostic marker for CGNs using peripheral blood.
Collapse
|
33
|
Links between coagulation, inflammation, regeneration, and fibrosis in kidney pathology. J Transl Med 2016; 96:378-90. [PMID: 26752746 DOI: 10.1038/labinvest.2015.164] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/22/2015] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) involves nephron injury leading to irreversible nephron loss, ie, chronic kidney disease (CKD). Both AKI and CKD are associated with distinct histological patterns of tissue injury, but kidney atrophy in CKD involves tissue remodeling with interstitial inflammation and scarring. No doubt, nephron atrophy, inflammation, fibrosis, and renal dysfunction are associated with each other, but their hierarchical relationships remain speculative. To better understand the pathophysiology, we provide an overview of the fundamental danger response programs that assure host survival upon traumatic injury from as early as the first multicellular organisms, ie, bleeding control by coagulation, infection control by inflammation, epithelial barrier restoration by re-epithelialization, and tissue stabilization by mesenchymal repair. Although these processes assure survival in the majority of the populations, their dysregulation causes kidney disease in a minority. We discuss how, in genetically heterogeneous population, genetic variants shift balances and modulate danger responses toward kidney disease. We further discuss how classic kidney disease entities develop from an insufficient or overshooting activation of these danger response programs. Finally, we discuss molecular pathways linking, for example, inflammation and regeneration or inflammation and fibrosis. Understanding the causative and hierarchical relationships and the molecular links between the danger response programs should help to identify molecular targets to modulate kidney injury and to improve outcomes for kidney disease patients.
Collapse
|
34
|
Nagata M. Podocyte injury and its consequences. Kidney Int 2016; 89:1221-30. [PMID: 27165817 DOI: 10.1016/j.kint.2016.01.012] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/12/2015] [Accepted: 01/27/2016] [Indexed: 01/02/2023]
Abstract
Podocytes maintain the glomerular filtration barrier, and the stability of this barrier depends on their highly differentiated postmitotic phenotype, which also defines the particular vulnerability of the glomerulus. Recent podocyte biology and gene disruption studies in vivo indicate a causal relationship between abnormalities of single podocyte molecules and proteinuria and glomerulosclerosis. Podocytes live under various stresses and pathological stimuli. They adapt to maintain homeostasis, but excessive stress leads to maladaptation with complex biological changes including loss of integrity and dysregulation of cellular metabolism. Podocyte injury causes proteinuria and detachment from the glomerular basement membrane. In addition to "sick" podocytes and their detachment, our understanding of glomerular responses following podocyte loss needs to address the pathways from podocyte injury to sclerosis. Studies have found a variety of glomerular responses to podocyte dysfunction in vivo, such as disruption of podocyte-endothelial cross talk and activation of podocyte-parietal cell interactions, all of which help us to understand the complex scenario of podocyte injury and its consequences. This review focuses on the cellular aspects of podocyte dysfunction and the adaptive or maladaptive glomerular responses to podocyte injury that lead to its major consequence, glomerulosclerosis.
Collapse
Affiliation(s)
- Michio Nagata
- Kidney and Vascular Pathology, University of Tsukuba, Ibaraki, Japan.
| |
Collapse
|
35
|
Andeen NK, Nguyen TQ, Steegh F, Hudkins KL, Najafian B, Alpers CE. The phenotypes of podocytes and parietal epithelial cells may overlap in diabetic nephropathy. Kidney Int 2015; 88:1099-107. [PMID: 26376129 PMCID: PMC4653076 DOI: 10.1038/ki.2015.273] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 07/09/2015] [Accepted: 07/16/2015] [Indexed: 01/15/2023]
Abstract
Reversal of diabetic nephropathy (DN) has been achieved in humans and mice, but only rarely and under special circumstances. Since progression of DN is related to podocyte loss, reversal of DN requires restoration of podocytes. Here we identified and quantified potential glomerular progenitor cells that could be a source for restored podocytes. DN was identified in 31 human renal biopsy cases and separated into morphologically early or advanced lesions. Markers of podocytes (WT-1, p57), parietal epithelial cells (claudin-1) and cell proliferation (Ki-67) were identified by immunohistochemistry. Podocyte density was progressively reduced with DN. Cells marking as podocytes (p57) were present infrequently on Bowman's capsule in controls, but significantly increased in histologically early DN. Ki-67 expressing cells were identified on the glomerular tuft and Bowman's capsule in DN, but rarely in controls. Cells marking as PECs were present on the glomerular tuft, particularly in morphologically advanced DN. These findings show evidence of phenotypic plasticity in podocyte and PEC populations and are consistent with studies in the BTBR ob/ob murine model in which reversibility of DN occurs with podocytes potentially regenerating from PEC precursors. Thus, our findings support, but do not prove, that podocytes may regenerate from PEC progenitors in human DN. If so, progression of DN may represent a modifiable net balance between podocyte loss and regeneration.
Collapse
Affiliation(s)
- Nicole K Andeen
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Tri Q Nguyen
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Floor Steegh
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Behzad Najafian
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
36
|
Pharmacological targeting of actin-dependent dynamin oligomerization ameliorates chronic kidney disease in diverse animal models. Nat Med 2015; 21:601-9. [PMID: 25962121 PMCID: PMC4458177 DOI: 10.1038/nm.3843] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 12/11/2022]
Abstract
Dysregulation of the actin cytoskeleton in podocytes represents a common pathway in the pathogenesis of proteinuria across a spectrum of chronic kidney diseases (CKD). The GTPase dynamin has been implicated in the maintenance of cellular architecture in podocytes through its direct interaction with actin. Furthermore, the propensity of dynamin to oligomerize into higher-order structures in an actin-dependent manner and to crosslink actin microfilaments into higher order structures have been correlated with increased actin polymerization and global organization of the actin cytoskeleton in the cell. We found that use of the small molecule Bis-T-23, which promotes actin-dependent dynamin oligomerization and thus increased actin polymerization in injured podocytes, was sufficient to improve renal health in diverse models of both transient kidney disease and of CKD. In particular, administration of Bis-T-23 in these renal disease models restored the normal ultrastructure of podocyte foot processes, lowered proteinuria, lowered collagen IV deposits in the mesangial matrix, diminished mesangial matrix expansion and extended lifespan. These results further establish that alterations in the actin cytoskeleton of kidney podocytes is a common hallmark of CKD, while also underscoring the significant regenerative potential of injured glomeruli and that targeting the oligomerization cycle of dynamin represents an attractive potential therapeutic target to treat CKD.
Collapse
|
37
|
Suwanpen C, Nouanthong P, Jaruvongvanich V, Pongpirul K, Pongpirul WA, Leelahavanichkul A, Kanjanabuch T. Urinary podocalyxin, the novel biomarker for detecting early renal change in obesity. J Nephrol 2015; 29:37-44. [PMID: 25905599 DOI: 10.1007/s40620-015-0199-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/15/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND The prevalence of obesity is increasing during the past decade along with obesity-related glomerulopathy (ORG), glomeruli injury due to the obesity. The major pathogenesis of ORG is the shedding of podocytes from the glomerular cell barrier into urine. Podocalyxin (PCX), a main surface antigen of podocyte, correlates well with glomerulosclerosis progression and glomerular injury severity, and might be a potential biomarker for early renal alteration in obesity. In addition, vascular endothelial growth factor (VEGF) and alpha-smooth muscle actin (α-SMA) also play a role in promoting glomerulosclerosis. The aim of this study was to explore whether obese subjects without other diseases excrete more PCX-positive (PCX+) cells than non-obese individuals, in comparison with urine protein-creatinine ratio (UPCR) and glomerular filtration rate (GFR) as traditional renal markers. Moreover, the effect of body mass index (BMI) on urinary VEGF, PCX or α-SMA positive cells was also investigated. METHODS Forty-eight obese and 13 non-obese adults were included. Exfoliated cells from fresh first void morning urine were harvested, stained with PCX, VEGF, and α-SMA antibody, and quantified by flow cytometry. Correlation between interested urinary biomarkers (cells positive for PCX, VEGF plus PCX and α-SMA), UPCR and GFR with BMI and metabolic risk factors were analyzed. RESULTS Obese patients had significantly higher PCX+ cells than non-obese [0.62 (0.00-13.13) vs. 0.15 (0.00-0.72) cells/ml × mg cr, p < 0.05]. There was no significant difference in GFR and UPCR between the groups. Of interest, BMI demonstrated a correlation with PCX+ cells (r = 0.343, p = 0.008) and cells positive for PCX plus VEGF (r = 0.374, p = 0.004). CONCLUSION Obese subjects without other diseases and with normal UPCR and GFR showed evidence of renal alteration through the detection of a higher number of PCX+ cells. Increasing BMI also resulted in higher number of PCX+ cells.
Collapse
Affiliation(s)
- Chayanut Suwanpen
- Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok, 10330, Thailand.
| | - Phonethipsavanh Nouanthong
- Pasteur Institute du Laos, Samsenthai Road, Ban Kao-Gnot, Sisattanak district, P.O Box 3560, Vientiane, Lao PDR.
| | - Veeravich Jaruvongvanich
- Department of Internal Medicine, University of Hawaii, 1356 Lusitana Street, Honolulu, HI, 96813, USA.
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Wannarat Amornnimit Pongpirul
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Asada Leelahavanichkul
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Immunology Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Talerngsak Kanjanabuch
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand. .,Kidney and Metabolic Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|