1
|
Khater Y, Barakat N, Shokeir A, Hamed M, Samy A, Karrouf G. Synergy of zinc oxide nanoparticles to losartan attenuates kidney injury induced by unilateral ureteral obstruction through modulation of the TNF-α/IL6 and BAX/BCL2 signaling pathways. Int Urol Nephrol 2025:10.1007/s11255-024-04331-y. [PMID: 39810058 DOI: 10.1007/s11255-024-04331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
AIM Although the relief of ureteral obstruction seems to be a radical treatment for obstructive uropathy (OU), progressive kidney damage is the result because of the associated increased apoptosis and fibrosis. Therefore, it is urgent to find a complementary renoprotective therapy against partially obstructed uropathy cascades. Thus, this study investigated the renoprotective effects of both losartan (LOS) and zinc oxide nanoparticles (ZnONPs) in partial unilateral ureteral obstruction (PUUO). MAIN METHODS In controlled (n = 16) and shamed (n = 16) study, 64 healthy male Sprague-Dawley rats, both PUUO and right nephrectomy (RNX) were induced. The rats were equally allocated into four groups according to treatment protocol: (1) PUUO group (no treatment), (2) ZnONPs group, (3) LOS group and (4) ZnONPs/LOS group. Antioxidant status and gene expression were assessed in renal tissues. Moreover, histologic and immunohistochemical examinations were performed. KEY FINDINGS LOS and ZnONPs significantly mitigated the PUUO-induced renal injury, by significant (P < 0.0001) suppressing of oxidative stress (MDA and TOS), upregulating of antioxidant gene (SOD) and antiapoptotic gene (BCL2), and downregulating the expression of inflammatory cytokines (TNF-α, and IL6), apoptotic gene (Bax) and fibrotic marker (β-Catenin). The combination of both agents offered a more powerful renoprotective effect with additional significant upregulation of the antioxidant marker (TAC, P < 0.0001). SIGNIFICANCE Both losartan and ZnONPs and specially their combination have synergistic action in protecting the kidney against PUUO-induced chronic renal cascades through improvement the renal function tests, amelioration of oxidative stress, inhibition of induced apoptosis and fibrosis with marked renal regeneration which highlights the possible application of these drugs as a complementary therapies for different chronic renal degenerative diseases.
Collapse
Affiliation(s)
- Yomna Khater
- Medical Experimental Research Centre, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed Shokeir
- Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
- Centre of Excellence of Genome and Cancer Research, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Mohamed Hamed
- Department of Pathology, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, 35516, Egypt
| | - Alaa Samy
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, 35516, Egypt
| | - Gamal Karrouf
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, 35516, Egypt.
| |
Collapse
|
2
|
López-Martínez M, Armengol MP, Pey I, Farré X, Rodríguez-Martínez P, Ferrer M, Porrini E, Luis-Lima S, Díaz-Martín L, Rodríguez-Rodríguez AE, Cruz-Perera C, Alcalde M, Navarro-Díaz M. Integrated miRNA-mRNA Analysis Reveals Critical miRNAs and Targets in Diet-Induced Obesity-Related Glomerulopathy. Int J Mol Sci 2024; 25:6437. [PMID: 38928144 PMCID: PMC11204096 DOI: 10.3390/ijms25126437] [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: 05/01/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
This study aimed to investigate obesity-related glomerulopathy (ORG) at cellular, structural, and transcriptomic levels. Thirty Wistar rats were randomized into two groups: 15 rats were fed with a standard diet (SD-rats), and 15 rats were fed with a high-fat diet (HFD-rats). After 10 weeks, the weight, kidney function, histological features, and transcriptomic changes were assessed. HFD-rats gained significantly more weight (55.8% vs. 29.2%; p < 0.001) and albuminuria (10,384.04 ng/mL vs. 5845.45 ng/mL; p < 0.001) compared to SD-rats. HFD-rats exhibited early stages of ORG, with predominant mesangial matrix increase and podocyte hypertrophy (PH). These lesions correlated with differentially expressed (DE) genes and miRNAs. Functional analysis showed that miR-205, which was DE in both the kidneys and urine of HFD-rats, negatively regulated the PTEN gene, promoting lipid endocytosis in podocytes. The downregulation of PTEN was proved through a higher PTEN/nephrin ratio in the SD-rats and the presence of lipid vacuoles in HFD-podocytes. This study has found a specific targetome of miRNAs and gene expression in early stages of ORG. Also, it emphasizes the potential value of miR-205 as a urinary biomarker for detecting podocyte injury in ORG, offering a tool for early diagnosis, and opening new avenues for future therapeutic research of obesity-related glomerulopathy.
Collapse
Affiliation(s)
- Marina López-Martínez
- CSUR National Unit of Expertise for Complex Glomerular Diseases of Spain, Nephrology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Research, 08035 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08913 Barcelona, Spain
| | - Maria Pilar Armengol
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | - Irina Pey
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | - Xavier Farré
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
| | | | - Mireia Ferrer
- Statistics and Bioinformatics Unit, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Esteban Porrini
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
- Instituto de Tecnologías Biomédicas (ITB), Faculty of Medicine, University of La Laguna, La Laguna, 38320 Tenerife, Spain
| | - Sergio Luis-Lima
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
- Department of Laboratory Medicine, Complejo Hospitalario Universitario de Canarias, La Laguna, 38320 Tenerife, Spain
| | - Laura Díaz-Martín
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
| | - Ana Elena Rodríguez-Rodríguez
- Research Unit, Hospital Universitario de Canarias, La Laguna, 38320 Tenerife, Spain
- Fundación General de la Universidad, University of La Laguna,38320 Tenerife, Spain
| | - Coriolano Cruz-Perera
- Laboratory of Renal Function (LFR), Faculty of Medicine, University of La Laguna, Complejo Hospitalario Universitario de Canarias, 38320 La Laguna, Spain (L.D.-M.)
| | - Marta Alcalde
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Pharmaco and Device Epidemiology Group, CSM, NDORMS, University of Oxford, Oxford OX1 3PT, UK
| | - Maruja Navarro-Díaz
- Genomic Platform, Germans Trias i Pujol’s Research Institute, Badalona, 08916 Barcelona, Spain
- Nephrology Department, Sant Joan Despí Moisès Broggi Hospital, Sant Joan Despí, 08970 Barcelona, Spain
| |
Collapse
|
3
|
Qiu Y, Lei C, Zeng J, Xie Y, Cao Y, Yuan Q, Su H, Zhang Z, Zhang C. Asparagine endopeptidase protects podocytes in adriamycin-induced nephropathy by regulating actin dynamics through cleaving transgelin. Mol Ther 2023; 31:3337-3354. [PMID: 37689970 PMCID: PMC10638058 DOI: 10.1016/j.ymthe.2023.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is the most common glomerular disorder causing end-stage renal diseases worldwide. Central to the pathogenesis of FSGS is podocyte dysfunction, which is induced by diverse insults. However, the mechanism governing podocyte injury and repair remains largely unexplored. Asparagine endopeptidase (AEP), a lysosomal protease, regulates substrates by residue-specific cleavage or degradation. We identified the increased AEP expression in the primary proteinuria model which was induced by adriamycin (ADR) to mimic human FSGS. In vivo, global AEP knockout mice manifested increased injury-susceptibility of podocytes in ADR-induced nephropathy (ADRN). Podocyte-specific AEP knockout mice exhibited much more severe glomerular lesions and podocyte injury after ADR injection. In contrast, podocyte-specific augmentation of AEP in mice protected against ADRN. In vitro, knockdown and overexpression of AEP in human podocytes revealed the cytoprotection of AEP as a cytoskeleton regulator. Furthermore, transgelin, an actin-binding protein regulating actin dynamics, was cleaved by AEP, and, as a result, removed its actin-binding regulatory domain. The truncated transgelin regulated podocyte actin dynamics and repressed podocyte hypermotility, compared to the native full-length transgelin. Together, our data reveal a link between lysosomal protease AEP and podocyte cytoskeletal homeostasis, which suggests a potential therapeutic role for AEP in proteinuria disease.
Collapse
Affiliation(s)
- Yang Qiu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Chuntao Lei
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Jieyu Zeng
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yaru Xie
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yiling Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Qian Yuan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| |
Collapse
|
4
|
Smith KD, Akilesh S. Collapsing glomerulopathy: unraveling varied pathogeneses. Curr Opin Nephrol Hypertens 2023; 32:213-222. [PMID: 36811644 DOI: 10.1097/mnh.0000000000000873] [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] [Indexed: 02/24/2023]
Abstract
PURPOSE OF REVIEW Collapsing glomerulopathy presents clinically with nephrotic syndrome and rapid progressive loss of kidney function. Animal models and patient studies have uncovered numerous clinical and genetic conditions associated with collapsing glomerulopathy, as well as putative mechanisms, which will be reviewed here. RECENT FINDINGS Collapsing glomerulopathy is classified pathologically as a variant of focal and segmental glomerulosclerosis (FSGS). As such, most research efforts have focused on the causative role of podocyte injury in driving the disease. However, studies have also shown that injury to the glomerular endothelium or interruption of the podocyte-glomerular endothelial cell signaling axis can also cause collapsing glomerulopathy. Furthermore, emerging technologies are now enabling exploration of diverse molecular pathways that can precipitate collapsing glomerulopathy using biopsies from patients with the disease. SUMMARY Since its original description in the 1980s, collapsing glomerulopathy has been the subject of intense study, and these efforts have uncovered numerous insights into potential disease mechanisms. Newer technologies will enable profiling of the intra-patient and inter-patient variability in collapsing glomerulopathy mechanisms directly in patient biopsies, which will improve the diagnosis and classification of collapsing glomerulopathy.
Collapse
Affiliation(s)
- Kelly D Smith
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington
- Kidney Research Institute, Seattle, Washington, USA
| |
Collapse
|
5
|
Ai Z, Wang M, Zhou Y, Yuan D, Jian Q, Wu S, Liu B, Yang Y. Deciphering the pharmacological mechanisms of Rostellularia procumbens (L) Nees. Extract alleviates adriamycin-induced nephropathy in vivo and in vitro. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154736. [PMID: 36907143 DOI: 10.1016/j.phymed.2023.154736] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 02/09/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Rostellularia procumbens (L) Nees. is an effective traditional Chinese herbal medicine for the treatment of patients with chronic glomerulonephritis (CGN) in the clinic. However, the underlying molecular mechanisms need further elucidation. PURPOSE This study aims to investigate the renoprotective mechanisms of n-butanol extract from Rostellularia procumbens (L) Nees. (J-NE) in vivo and in vitro. METHODS The components of J-NE were analyzed by UPLC-MS/MS. In vivo, the nephropathy model was induced in mice by tail vein injection with adriamycin (10 mg·kg-1), and mice were treated with vehicle or J-NE or benazepril by daily gavage. In vitro, MPC5 cells exposed to adriamycin (0.3 μg/ml) were treated with J-NE. The effects of J-NE inhibit podocyte apoptosis and protect against adriamycin-induced nephropathy were determined by Network pharmacology, RNA-seq, qPCR, ELISA, immunoblotting, flow cytometry, and TUNEL assay, according to the experimental protocols. RESULT The results showed that treatment significantly improved ADR-induced renal pathological changes, and the therapeutic mechanism of J-NE was related to the inhibition of podocyte apoptosis. Further molecular mechanism studies found that J-NE inhibited inflammation, increase the proteins expression levels of Nephrin and Podocin, reduce TRPC6 and Desmin expression levels and calcium ion levels in podocytes, and decrease the proteins expression levels of PI3K, p-PI3K, Akt and p-Akt to attenuated apoptosis. Furthermore, 38 compounds of J-NE were identified. CONCLUSION J-NE exerted the renoprotective effects by inhibiting podocyte apoptosis, which provides effective evidence for the treatment of J-NE targeting renal injury in CGN.
Collapse
Affiliation(s)
- Zhongzhu Ai
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan 430065, China; Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Wuhan 430065, China
| | - Mengfan Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan 430065, China; Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Wuhan 430065, China
| | - Yi Zhou
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Dongfeng Yuan
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qiuyuan Jian
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Songtao Wu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Bo Liu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan 430065, China; Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Wuhan 430065, China
| | - Yanfang Yang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan 430065, China; Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Wuhan 430065, China.
| |
Collapse
|
6
|
Shen Q, Teng L, Wang Y, Guo L, Xu F, Huang H, Xie W, Zhou Q, Chen Y, Wang J, Mao Y, Chen J, Jiang H. Integrated genomic, transcriptomic and metabolomic analysis reveals MDH2 mutation-induced metabolic disorder in recurrent focal segmental glomerulosclerosis. Front Immunol 2022; 13:962986. [PMID: 36159820 PMCID: PMC9495259 DOI: 10.3389/fimmu.2022.962986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) has an over 30% risk of recurrence after kidney transplantation (Ktx) and is associated with an extremely high risk of graft loss. However, mechanisms remain largely unclear. Thus, this study identifies novel genes related to the recurrence of FSGS (rFSGS). Whole genome-wide sequencing and next-generation RNA sequencing were used to identify the candidate mutant genes associated with rFSGS in peripheral blood mononuclear cells (PBMCs) from patients with biopsy-confirmed rFSGS after KTx. To confirm the functional role of the identified gene with the MDH2 c.26C >T mutation, a homozygous MDH2 c.26C >T mutation in HMy2.CIR cell line was induced by CRISPR/Cas9 and co-cultured with podocytes, mesangial cells, or HK2 cells, respectively, to detect the potential pathogenicity of the c.26C >T variant in MDH2. A total of 32 nonsynonymous single nucleotide polymorphisms (SNPs) and 610 differentially expressed genes (DEGs) related to rFSGS were identified. DEGs are mainly enriched in the immune and metabolomic-related pathways. A variant in MDH2, c.26C >T, was found in all patients with rFSGS, which was also accompanied by lower levels of mRNA expression in PBMCs from relapsed patients compared with patients with remission after KTx. Functionally, co-cultures of HMy2.CIR cells overexpressing the mutant MDH2 significantly inhibited the expression of synaptopodin, podocin, and F-actin by podocytes compared with those co-cultured with WT HMy2.CIR cells or podocytes alone. We identified that MDH2 is a novel rFSGS susceptibility gene in patients with recurrence of FSGS after KTx. Mutation of the MDH2 c.26C >T variant may contribute to progressive podocyte injury in rFSGS patients.
Collapse
Affiliation(s)
- Qixia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Lisha Teng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Yucheng Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Luying Guo
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Feng Xu
- The Centre for Heart and Lung Innovation, The University of British Columbia, Vancouver, BC, Canada
| | - Hongfeng Huang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Wenqing Xie
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Qin Zhou
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Junwen Wang
- Department of Health Sciences Research and Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Youying Mao
- Dapartment of Nephrology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- *Correspondence: Hong Jiang,
| |
Collapse
|
7
|
Rachubik P, Szrejder M, Rogacka D, Typiak M, Audzeyenka I, Kasztan M, Pollock DM, Angielski S, Piwkowska A. Insulin controls cytoskeleton reorganization and filtration barrier permeability via the PKGIα-Rac1-RhoA crosstalk in cultured rat podocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119301. [PMID: 35642843 DOI: 10.1016/j.bbamcr.2022.119301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Podocyte foot processes are an important cellular layer of the glomerular barrier that regulates glomerular permeability. Insulin via the protein kinase G type Iα (PKGIα) signaling pathway regulates the balance between contractility and relaxation (permeability) of the podocyte barrier by regulation of the actin cytoskeleton. This mechanism was shown to be disrupted in diabetes. Rho family guanosine-5'-triphosphates (GTPases) are dynamic modulators of the actin cytoskeleton and expressed in cells that form the glomerular filtration barrier. Thus, changes in Rho GTPase activity may affect glomerular permeability to albumin. The present study showed that Rho family GTPases control podocyte migration and permeability. Moreover these processes are regulated by insulin in PKGIα-dependent manner. Modulation of the PKGI-dependent activity of Rac1 and RhoA GTPases with inhibitors or small-interfering RNA impair glomerular permeability to albumin. We also demonstrated this mechanism in obese, insulin-resistant Zucker rats. We propose that PKGIα-Rac1-RhoA crosstalk is necessary in proper organization of the podocyte cytoskeleton and consequently the stabilization of glomerular architecture and regulation of filtration barrier permeability.
Collapse
Affiliation(s)
- Patrycja Rachubik
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Maria Szrejder
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Marlena Typiak
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Biology, Department of General and Medical Biochemistry, Gdańsk, Poland
| | - Irena Audzeyenka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Małgorzata Kasztan
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefan Angielski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland.
| |
Collapse
|
8
|
Sun K, Xie Q, Hao CM. Mechanisms of Scarring in Focal Segmental Glomerulosclerosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2021; 7:350-358. [PMID: 34604342 PMCID: PMC8443927 DOI: 10.1159/000517108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a histologic pattern characterized by focal glomerular scarring, which often progresses to systemic and diffuse glomerulosclerosis. Previous studies have emphasized that the initiation of classic FSGS occurs in podocytes. The dysfunction and loss of podocytes have been associated with the development of proteinuria and the progression of various diseases. In addition, primary, secondary, and genetic FSGS are caused by different mechanisms of podocyte injury. SUMMARY The potential sources and mechanism of podocyte supplementation are the focus of our current research. Increasing attention has been paid to the role played by parietal epithelial cells (PECs) during the progression of FSGS. PECs are not only the primary influencing factors in glomerulosclerosis lesions but also have repair abilities, which remain a focus of debate. Notably, other resident glomerular cells also play significant roles in the progression of this disease. KEY MESSAGE In this review, we focus on the mechanism of scarring in FSGS and discuss current and potential therapeutic strategies.
Collapse
Affiliation(s)
- Ke Sun
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qionghong Xie
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- Nephrology Division, Vanderbilt University Medical Center School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
9
|
McCarthy GM, Blasio A, Donovan OG, Schaller LB, Bock-Hughes A, Magraner JM, Suh JH, Tattersfield CF, Stillman IE, Shah SS, Zsengeller ZK, Subramanian B, Friedman DJ, Pollak MR. Recessive, gain-of-function toxicity in an APOL1 BAC transgenic mouse model mirrors human APOL1 kidney disease. Dis Model Mech 2021; 14:dmm048952. [PMID: 34350953 PMCID: PMC8353097 DOI: 10.1242/dmm.048952] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022] Open
Abstract
People of recent sub-Saharan African ancestry develop kidney failure much more frequently than other groups. A large fraction of this disparity is due to two coding sequence variants in the APOL1 gene. Inheriting two copies of these APOL1 risk variants, known as G1 and G2, causes high rates of focal segmental glomerulosclerosis (FSGS), HIV-associated nephropathy and hypertension-associated end-stage kidney disease. Disease risk follows a recessive mode of inheritance, which is puzzling given the considerable data that G1 and G2 are toxic gain-of-function variants. We developed coisogenic bacterial artificial chromosome (BAC) transgenic mice harboring either the wild-type (G0), G1 or G2 forms of human APOL1. Expression of interferon gamma (IFN-γ) via plasmid tail vein injection results in upregulation of APOL1 protein levels together with robust induction of heavy proteinuria and glomerulosclerosis in G1/G1 and G2/G2 but not G0/G0 mice. The disease phenotype was greater in G2/G2 mice. Neither heterozygous (G1/G0 or G2/G0) risk variant mice nor hemizygous (G1/-, G2/-) mice had significant kidney injury in response to IFN-γ, although the heterozygous mice had a greater proteinuric response than the hemizygous mice, suggesting that the lack of significant disease in humans heterozygous for G1 or G2 is not due to G0 rescue of G1 or G2 toxicity. Studies using additional mice (multicopy G2 and a non-isogenic G0 mouse) supported the notion that disease is largely a function of the level of risk variant APOL1 expression. Together, these findings shed light on the recessive nature of APOL1-nephropathy and present an important model for future studies.
Collapse
Affiliation(s)
- Gizelle M. McCarthy
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Angelo Blasio
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Olivia G. Donovan
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Lena B. Schaller
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Althea Bock-Hughes
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jose M. Magraner
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jung Hee Suh
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Calum F. Tattersfield
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Isaac E. Stillman
- Dept. of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Shrijal S. Shah
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Zsuzsanna K. Zsengeller
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Balajikarthick Subramanian
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - David J. Friedman
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Martin R. Pollak
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
10
|
Gu X, Zhang S, Zhang T. Abnormal Crosstalk between Endothelial Cells and Podocytes Mediates Tyrosine Kinase Inhibitor (TKI)-Induced Nephrotoxicity. Cells 2021; 10:cells10040869. [PMID: 33921219 PMCID: PMC8070074 DOI: 10.3390/cells10040869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/04/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor A (VEGFA) and its receptor VEGFR2 are the main targets of antiangiogenic therapies, and proteinuria is one of the common adverse events associated with the inhibition of the VEGFA/VEGFR2 pathway. The proteinuric kidney damage induced by VEGFR2 tyrosine kinase inhibitors (TKIs) is characterized by podocyte foot process effacement. TKI therapy promotes the formation of abnormal endothelial‒podocyte crosstalk, which plays a key role in TKI-induced podocyte injury and proteinuric nephropathy. This review article summarizes the underlying mechanism by which the abnormal endothelial‒podocyte crosstalk mediates podocyte injury and discusses the possible molecules and signal pathways involved in abnormal endothelial‒podocyte crosstalk. What is more, we highlight the molecules involved in podocyte injury and determine the essential roles of Rac1 and Cdc42; this provides evidence for exploring the abnormal endothelial‒podocyte crosstalk in TKI-induced nephrotoxicity.
Collapse
Affiliation(s)
| | | | - Ti Zhang
- Correspondence: ; Tel.: +86-21-6417-5590
| |
Collapse
|
11
|
Glomerular filtrate affects the dynamics of podocyte detachment in a model of diffuse toxic podocytopathy. Kidney Int 2021; 99:1149-1161. [PMID: 33582108 DOI: 10.1016/j.kint.2020.12.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 11/22/2022]
Abstract
Podocyte injury and subsequent detachment are hallmarks of progressive glomerulosclerosis. In addition to cell injury, unknown mechanical forces on the injured podocyte may promote detachment. To identify the nature of these mechanical forces, we studied the dynamics of podocyte detachment using sequential ultrastructural geometry analysis by transmission electron microscopy in NEP25, a mouse model of podocytopathy induced by anti-Tac(Fv)-PE38 (LMB2), a fusion protein attached to Pseudomonas exotoxin A, targeting CD25 on podocytes. After LMB2 injection, foot process effacement occurred on day three but detachment commenced on day eight and extended to day ten, reaching toward the urinary pole in clusters. Podocyte detachment was associated with foot process effacement covering over 60% of the glomerular basement membrane length. However, approximately 25% of glomeruli with diffuse (over 80%) foot process effacement showed no detachment. Blocking glomerular filtration via unilateral ureteral obstruction resulted in diffuse foot process effacement but no pseudocysts or detachment, whereas uninephrectomy increased pseudocysts and accelerated detachment, indicating that glomerular filtrate drives podocyte detachment via pseudocyst formation as a forerunner. Additionally, more detachment was observed in juxtamedullary glomeruli than in superficial glomeruli. Thus, glomerular filtrate drives the dynamics of podocyte detachment in this model of podocytopathy. Hence, foot process effacement may be a prerequisite allowing filtrate to generate local mechanical forces that expand the subpodocyte space forming pseudocysts, promote podocyte detachment and subsequent segmental sclerosis.
Collapse
|
12
|
Chen Y, Liu Q, Shan Z, Zhao Y, Li M, Wang B, Zheng X, Feng W. The protective effect and mechanism of catalpol on high glucose-induced podocyte injury. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:244. [PMID: 31488111 PMCID: PMC6727542 DOI: 10.1186/s12906-019-2656-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Catalpol, a natural iridoid glycoside in Rehmannia glutinosa, can alleviate proteinuria associated with diabetic nephropathy (DN), however, whether catalpol has a protective effect against podocyte injury in DN remains unclear. METHODS In this study, we used a high glucose (HG)-induced podocyte injury model to evaluate the protective effect and mechanism of catalpol against HG-induced podocyte injury. Cell viability was determined by the 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. The levels of lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA) were measured by commercial assay kits. Cell apoptosis and reactive oxygen species (ROS) were determined by using flow cytometry. Tumour necrosis factor α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) levels were determined by enzyme-linked immunosorbent assay (ELISA). The protein expression levels of B-cell lymphoma-2 (Bcl-2), Bcl2-associated x (Bax), cleaved caspase-3, nicotinamide adenine dinucleotide phosphate oxidase enzyme 4 (NOX4), toll-like receptor 4 (TLR4), myeloid differentiation primary response gene 88 (MyD88), p38 mitogen-activated protein kinase (p38 MAPK), phosphorylated p38 MAPK (p-p38 MAPK), nuclear factor kappa B inhibitor alpha (IκBα) and phosphorylated IκBα (p-IκBα) were measured by western blotting. In addition, Bcl-2, Bax, caspase-3 and nuclear factor kappa B (NF-κB) levels were determined by immunofluorescence staining. RESULTS Catalpol significantly increased cell viability and decreased LDH release in HG-induced podocyte injury. Catalpol significantly decreased ROS generation, apoptosis, level of MDA, levels of inflammatory cytokine TNF-α, IL-1β, and IL-6 and increased SOD activity in HG-induced podocyte injury. Moreover, catalpol significantly decreased expression of cleaved caspase-3, Bax, NOX4, TLR4, MyD88, p-p38 MAPK, p-IκBα and NF-κB nuclear translocation, as well as increased Bcl-2 expression in HG-induced podocyte injury. CONCLUSION Catalpol can protect against podocyte injury by ameliorating apoptosis and inflammation. These protective effects may be attributed to the inhibition of NOX4, which alleviates ROS generation and suppression of the TLR4/MyD88 and p38 MAPK signaling pathways to prevent NF-κB activation. Therefore, catalpol could be a promising drug for the prevention of DN.
Collapse
Affiliation(s)
- Yan Chen
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Qingpu Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Zengfu Shan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Yingying Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Meng Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Baiyan Wang
- College of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan, 450046, People's Republic of China.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People's Republic of China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, Henan, 450046, People's Republic of China.
| |
Collapse
|
13
|
Sipovsky VG, Nevorotin AI, Avsiewitsch IV, Smirnov AV. [Ultrastructural study of podocyte alterations in non proliferative glomerulopathy]. Arkh Patol 2019; 81:51-58. [PMID: 31317931 DOI: 10.17116/patol20198103151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ultrastructural changes in podocytes are an important diagnostic and prognostic marker for nephropathies. However, the biomedical understanding of detected submicroscopic changes in podocytes remains controversial. OBJECTIVE To investigate the relationship between the ultrastructural changes of podocytes (fusion of cytopodia and denudation of the basement membrane as a result of their desquamation) with a number of clinical and laboratory indicators of kidney dysfunction in case of non-proliferative glomerulopathies (NPGP). Thirty-seven patients (23 men, 14 women) with NPGP, including 8 with focal segmental glomerulosclerosis (FSGS), 17 with membranous nephropathy (MN), and 12 with minimal change disease (MCD), were examined. SUBJECT AND METHODS All the patients underwent standard laboratory and instrumental studies: determinations of the levels of total serum cholesterol (mmol/l), total serum protein (g/l); serum albumin (g/l); CKD-EPI glomerular filtration rate (GFR) (ml/min/1.73 m2), and daily protein loss (g/day). Light optical changes were measured; completely sclerotic and/or focally segmentally sclerotic glomeruli were taken into account. Quantitative ultrastructural stereological analysis was carried out estimating the cytopodium width (CPW) and the degree of glomerular basement membrane denudation (GBMD) (%). RESULTS NPGP cases showed the largest number of sclerotic glomeruli in FSGS, which was accompanied by the lowest level of daily proteinuria and GFR. Quantitative values of CPW were associated with the level of daily protein loss (r=0.47; p < 0.05) and serum albumin (r=-0.57; p <0.05) in patients with nephrotic syndrome. In MN, the absolute value of CPW was larger than that in the other two patient groups. A correlation analysis of CPW and GBMD values among patients with NPGP revealed a statistically insignificant negative relation between these morphometric parameters. However, when a subgroup of patients with podocytopathies (only MCD and FSGS) was identified in the study group, this relationship was found to be significant (r=-0.54; p=0.012). CONCLUSION The patients with NPGP exhibited a relationship between the severity of nephrotic syndrome and proteinuria/hypoalbuminemia, on the one hand, and CPW, on the other. The established negative relationship between CPW and the percentage of GBMD in the subgroup of patients with podocytopathies may be due to the early stages of podocyte injury, which are accompanied by transient GBMD.
Collapse
Affiliation(s)
- V G Sipovsky
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - A I Nevorotin
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - I V Avsiewitsch
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| | - A V Smirnov
- Acad. I.P. Pavlov First Saint Petersburg State Medical University, Ministry of Health of Russia, Saint-Petersburg, Russia
| |
Collapse
|
14
|
Saito H, Tanaka T, Sugahara M, Tanaka S, Fukui K, Wakashima T, Nangaku M. Inhibition of prolyl hydroxylase domain (PHD) by JTZ-951 reduces obesity-related diseases in the liver, white adipose tissue, and kidney in mice with a high-fat diet. J Transl Med 2019; 99:1217-1232. [PMID: 30952940 DOI: 10.1038/s41374-019-0239-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
The epidemic of obesity and its complications is rapidly increasing worldwide. Recent drug discoveries established the utility of prolyl hydroxylase domain (PHD) inhibitors as stabilizers of hypoxia-inducible factors (HIFs) in vivo, which are currently in human clinical studies for the treatment of anemia in chronic kidney disease (CKD). These studies suggest a role for PHD inhibitors in ameliorating obesity and hyperlipidemia. We hypothesized that HIF activation using a PHD inhibitor, JTZ-951, protects from obesity-related diseases in the white adipose tissue (WAT), liver, and kidney in mice fed with high-fat diet (HFD). Eight-week-old, C57BL/6J mice were fed with HFD for 20 weeks with or without JTZ-951(0.005%; mixed in chow). Body weight and plasma non-high-density lipoprotein (HDL) cholesterol levels were significantly lower in the JTZ-951 group as compared with the vehicle group. PHD inhibition improved liver steatosis, macrophage infiltration into WAT and adipocyte fibrosis. In the kidney, PHD inhibition reduced albuminuria. Histologically, the number of F4/80- positive infiltrating macrophages and mesangial expansion were milder in the JTZ-951 group. Relative mRNA expression of adiponectin in WAT was higher in the JTZ-951-treated group and inversely correlated with hepatic steatosis score, adipocyte macrophage aggregation, and albuminuria. Activation of HIF ameliorates multiple obesity-related consequences in mice with HFD. The results of the present study offer the promising view that pharmacological PHD inhibition may be beneficial for the treatment of obesity-related diseases that can be ameliorated by weight loss.
Collapse
Affiliation(s)
- Hisako Saito
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Mai Sugahara
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kenji Fukui
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Takeshi Wakashima
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Biological and Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
15
|
Wang D, Jin M, Zhao X, Zhao T, Lin W, He Z, Fan M, Jin W, Zhou J, Jin L, Zheng C, Jin H, Zhao Y, Li X, Ying L, Wang Y, Zhu G, Huang Z. FGF1 ΔHBS ameliorates chronic kidney disease via PI3K/AKT mediated suppression of oxidative stress and inflammation. Cell Death Dis 2019; 10:464. [PMID: 31189876 PMCID: PMC6561918 DOI: 10.1038/s41419-019-1696-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022]
Abstract
Currently, there is a lack of effective therapeutic approaches to the treatment of chronic kidney disease (CKD) with irreversible deterioration of renal function. This study aimed to investigate the ability of mutant FGF1 (FGF1ΔHBS, which has reduced mitogenic activity) to alleviate CKD and to study its associated mechanisms. We found that FGF1ΔHBS exhibited much weaker mitogenic activity than wild-type FGF1 (FGF1WT) in renal tissues. RNA-seq analysis revealed that FGF1ΔHBS inhibited oxidative stress and inflammatory signals in mouse podocytes challenged with high glucose. These antioxidative stress and anti-inflammatory activities of FGF1ΔHBS prevented CKD in two mouse models: a diabetic nephropathy model and an adriamycin-induced nephropathy model. Further mechanistic analyses suggested that the inhibitory effects of FGF1ΔHBS on oxidative stress and inflammation were mediated by activation of the GSK-3β/Nrf2 pathway and inhibition of the ASK1/JNK signaling pathway, respectively. An in-depth study demonstrated that both pathways are under control of PI3K/AKT signaling activated by FGF1ΔHBS. This finding expands the potential uses of FGF1ΔHBS for the treatment of various kinds of CKD associated with oxidative stress and inflammation.
Collapse
Affiliation(s)
- Dezhong Wang
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, Zhejiang, China
| | - Mengyun Jin
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xinyu Zhao
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Tianyang Zhao
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wei Lin
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhengle He
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Miaojuan Fan
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wei Jin
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jie Zhou
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lingwei Jin
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Chao Zheng
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hui Jin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yushuo Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,School of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, Zhejiang, China
| | - Lei Ying
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yang Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Guanghui Zhu
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Zhifeng Huang
- School of Pharmaceutical Sciences & Center for Structural Biology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
16
|
Dai R, Liu H, Han X, Liu J, Zhai Y, Rao J, Shen Q, Xu H. Angiopoietin-like-3 knockout protects against glomerulosclerosis in murine adriamycin-induced nephropathy by attenuating podocyte loss. BMC Nephrol 2019; 20:185. [PMID: 31126248 PMCID: PMC6533758 DOI: 10.1186/s12882-019-1383-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Background Angiopoietin-like-3 (Angptl3) knockout is known for its protective effects on podocyte injury and proteinuria in the early stage of adriamycin (ADR) nephropathy. The current study re-evaluated the renoprotective effect of Angptl3 knockout in chronic ADR nephropathy and attempted to explore the mechanism underlying the effect associated with Angptl3 knockout in glomerulosclerosis. Methods B6; 129S5 mice were injected with ADR to induce nephropathy. Kidney structure and serum and urine parameters were observed during long-term follow-up. Cultured primary mouse podocytes were exposed to ADR and analyzed for the expression of some relative proteins. Podocyte loss was analyzed in both in vivo and in vitro experiments. Results Angptl3 knockout attenuated proteinuria and hypoproteinemia, protected renal structure and function, and improved the survival of mice over the whole process of ADR nephropathy. Furthermore, Angptl3 knockout reduced the numbers of the detached and apoptotic cells in the renal tissue and alleviated podocyte loss in mice with ADR chronic nephropathy, thereby, delaying the glomerulosclerosis formation. Additional results in vitro showed that Angptl3 knockout attenuated ADR-induced primary podocyte loss, including podocyte detachment and apoptosis. Conclusion In addition to serving a renoprotective role in the early stage of ADR nephropathy, Angptl3 knockout contributed to disease amelioration throughout the ADR nephropathy process. Angptl3 knockout effectively delayed glomerulosclerosis formation by attenuating podocyte loss through rescuing podocytes from detachment and apoptosis. Angptl3 antagonists or inhibitors might have therapeutic potential in the occurrence and progression of nephropathy. Electronic supplementary material The online version of this article (10.1186/s12882-019-1383-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rufeng Dai
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| | - Haimei Liu
- Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China.,Department of Rheumatism, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xinli Han
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| | - Junchao Liu
- Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China.,Department of Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yihui Zhai
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| | - Jia Rao
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| | - Qian Shen
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| | - Hong Xu
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Shanghai Kidney Development & Pediatric Kidney Disease Research Center, Shanghai, 201102, China.
| |
Collapse
|
17
|
Hamie L, Daoud G, Nemer G, Nammour T, El Chediak A, Uthman IW, Kibbi AG, Eid A, Kurban M. SuPAR, an emerging biomarker in kidney and inflammatory diseases. Postgrad Med J 2018; 94:517-524. [PMID: 30177549 DOI: 10.1136/postgradmedj-2018-135839] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/10/2018] [Accepted: 07/28/2018] [Indexed: 12/12/2022]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a circulating form of a physiological and pathophysiological important cell surface receptor, implicated in inflammation. Recent studies showed that suPAR is a promising biomarker, useful for diagnosis, assessment and prognosis of several diseases. This review summarises the majority of preliminary studies and analyses the significance and the clinical application of suPAR in various clinical conditions. SuPAR seems to have a significant value in the diagnosis as well as prognosis of many diseases; nonetheless, it merits large-scale studies to set cut-off values that help physicians in following up their patients and accordingly tailor their treatment plans.
Collapse
Affiliation(s)
- Lamiaa Hamie
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Daoud
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Tarek Nammour
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Alissar El Chediak
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Imad W Uthman
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Abdul Ghani Kibbi
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Assaad Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mazen Kurban
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon .,Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
18
|
Wu Z, Blessing NA, Simske JS, Bruggeman LA. Fyn-binding protein ADAP supports actin organization in podocytes. Physiol Rep 2018; 5. [PMID: 29192064 PMCID: PMC5727265 DOI: 10.14814/phy2.13483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 09/29/2017] [Indexed: 01/19/2023] Open
Abstract
The renal podocyte is central to the filtration function of the kidney that is dependent on maintaining both highly organized, branched cell structures forming foot processes, and a unique cell-cell junction, the slit diaphragm. Our recent studies investigating the developmental formation of the slit diaphragm identified a novel claudin family tetraspannin, TM4SF10, which is a binding partner for ADAP (also known as Fyn binding protein Fyb). To investigate the role of ADAP in podocyte function in relation to Fyn and TM4SF10, we examined ADAP knockout (KO) mice and podocytes. ADAP KO mice developed glomerular pathology that began as hyalinosis and progressed to glomerulosclerosis, with aged male animals developing low levels of albuminuria. Podocyte cell lines established from the KO mice had slower attachment kinetics compared to wild-type cells, although this did not affect the total number of attached cells nor the ability to form focal contacts. After attachment, the ADAP KO cells did not attain typical podocyte morphology, lacking the elaborate cell protrusions typical of wild-type podocytes, with the actin cytoskeleton forming circumferential stress fibers. The absence of ADAP did not alter Fyn levels nor were there differences between KO and wild-type podocytes in the reduction of Fyn activating phosphorylation events with puromycin aminonucleoside treatment. In the setting of endogenous TM4SF10 overexpression, the absence of ADAP altered the formation of cell-cell contacts containing TM4SF10. These studies suggest ADAP does not alter Fyn activity in podocytes, but appears to mediate downstream effects of Fyn controlled by TM4SF10 involving actin cytoskeleton organization.
Collapse
Affiliation(s)
- Zhenzhen Wu
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Natalya A Blessing
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Jeffrey S Simske
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Leslie A Bruggeman
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
19
|
Yang JW, Dettmar AK, Kronbichler A, Gee HY, Saleem M, Kim SH, Shin JI. Recent advances of animal model of focal segmental glomerulosclerosis. Clin Exp Nephrol 2018; 22:752-763. [PMID: 29556761 DOI: 10.1007/s10157-018-1552-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 02/26/2018] [Indexed: 12/15/2022]
Abstract
In the last decade, great advances have been made in understanding the genetic basis for focal segmental glomerulosclerosis (FSGS). Animal models using specific gene disruption of the slit diaphragm and cytoskeleton of the foot process mirror the etiology of the human disease. Many animal models have been developed to understand the complex pathophysiology of FSGS. Therefore, we need to know the usefulness and exact methodology of creating animal models. Here, we review classic animal models and newly developed genetic animal models. Classic animal models of FSGS involve direct podocyte injury and indirect podocyte injury due to adaptive responses. However, the phenotype depends on the animal background. Renal ablation and direct podocyte toxin (PAN, adriamycin) models are leading animal models for FSGS, which have some limitations depending on mice background. A second group of animal models were developed using combinations of genetic mutation and toxin, such as NEP25, diphtheria toxin, and Thy1.1 models, which specifically injure podocytes. A third group of animal models involves genetic engineering techniques targeting podocyte expression molecules, such as podocin, CD2-associated protein, and TRPC6 channels. More detailed information about podocytopathy and FSGS can be expected in the coming decade. Different animal models should be used to study FSGS depending on the specific aim and sometimes should be used in combination.
Collapse
Affiliation(s)
- Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Gangwon, Republic of Korea
| | - Anne Katrin Dettmar
- Pediatric Nephrology, Department of Pediatrics, Medical University Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Universitätskliniken Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moin Saleem
- Paediatric Renal Medicine, University of Bristol, Bristol, UK.,Children's Renal Unit, Bristol Royal Hospital for Children, Bristol, UK
| | - Seong Heon Kim
- Department of Pediatrics, Pusan National University Children's Hospital, Yangsan, Republic of Korea. .,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, 50 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| |
Collapse
|
20
|
Wang B, Ding W, Zhang M, Li H, Guo H, Lin L, Chen J, Gu Y. Role of FOXO1 in aldosterone-induced autophagy: a compensatory protective mechanism related to podocyte injury. Oncotarget 2018; 7:45331-45351. [PMID: 27244896 PMCID: PMC5216726 DOI: 10.18632/oncotarget.9644] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023] Open
Abstract
This study was undertaken to elucidate whether and how autophagy was regulated in aldosterone (Aldo)-induced podocyte injury and to examine its role in this model both in vitro and in vivo. In cultured podocytes, Aldo increased autophagy flux as indicated by the enhanced expression of LC3-II/LC3-I and the reduction of p62. Autophagy induction with rapamycin (RP) provided a cytoprotective effect, and inhibition of autophagy with Atg7-specific siRNA, chloroquine (CQ) or 3-methyladenine (3-MA) worsened Aldo-induced podocyte injury by attenuating endoplasmic reticulum (ER) stress. Aldo inhibited Akt phosphorylation but increased the mammalian target of rapamycin (mTOR) signaling pathway; however, Aldo up-regulated the expression of FOXO1 and its downstream effector Rab7. Either knockdown of FOXO1 or Rab7 inhibited Aldo-induced autophagy. Additionally, an elevated level of P300-regulated acetylation of FOXO1 and the interaction of acetylated FOXO1 and Atg7 were also confirmed to be involved in regulating autophagy in Aldo-induced podocytes. Similar results were further confirmed in vivo. We propose that autophagy enhancement through enhancing of the FOXO1/Rab7 axis and post-translational modification of FOXO1 may represent a potential therapeutic strategy against podocyte injury by promoting autophagy.
Collapse
Affiliation(s)
- Bin Wang
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China
| | - Wei Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Minmin Zhang
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China
| | - Hongmei Li
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Honglei Guo
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Lilu Lin
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jing Chen
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China
| | - Yong Gu
- Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Yau W, Mausbach L, Littman MP, Cianciolo RE, Brown CA. Focal Segmental Glomerulosclerosis in Related Miniature Schnauzer Dogs. Vet Pathol 2017; 55:277-285. [PMID: 29191133 DOI: 10.1177/0300985817736356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) recently has been recognized as a common cause of proteinuria in dogs in general, and in Miniature Schnauzer dogs in particular. This study describes the morphologic features present in the kidneys of 8 related proteinuric Miniature Schnauzer dogs. The FSGS, characterized by solidification of portions of the capillary tuft, affected 32% to 49% of examined glomeruli in these dogs. Synechiae, often accompanied by hyalinosis, were present in 13% to 54% of glomeruli and were more prevalent in older dogs. Seven of 8 dogs had arteriolar hyalinosis. Ultrastructurally, all dogs had evidence of a podocytopathy in the absence of electron-dense deposits, glomerular basement membrane splitting, or fibrils. All dogs had multifocal to extensive podocyte foot process effacement. Other podocyte changes included microvillous transformation, the presence of vacuoles or protein resorption droplets, cytoplasmic electron-dense aggregates, and occasional binucleation. Variable amounts of intraglomerular lipid were present in all dogs. All dogs were proteinuric, with measured values for the urine protein-to-creatinine ratio ranging from 1.2 to 6.5. Azotemia was mild to absent and dogs were euthanatized at 5.1 to 14 years of age, in all cases due to nonrenal diseases. The underlying cause of FSGS in these Miniature Schnauzer dogs has yet to be determined, but contributors likely include genetic podocytopathy, lipid abnormalities, and glomerular hypertension.
Collapse
Affiliation(s)
- Wilson Yau
- 1 Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | | - Meryl P Littman
- 3 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel E Cianciolo
- 4 Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Cathy A Brown
- 1 Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.,5 Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
22
|
Zhang H, Liang S, Du Y, Li R, He C, Wang W, Liu S, Ye Z, Liang X, Shi W, Zhang B. Inducible ATF3-NFAT axis aggravates podocyte injury. J Mol Med (Berl) 2017; 96:53-64. [PMID: 29038896 PMCID: PMC5760612 DOI: 10.1007/s00109-017-1601-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/12/2017] [Accepted: 10/05/2017] [Indexed: 01/25/2023]
Abstract
Abstract Podocyte injury and loss contribute to proteinuria, glomerulosclerosis, and eventually kidney failure. Activating transcription factor 3 (ATF3) is a stress inducible transcription factor that is transiently expressed following stimulation. However, we show for the first time an induction of ATF3 in podocytes from patients with chronic kidney disease, including minimal change disease, focal segmental glomerulosclerosis, and diabetic nephropathy. The role of ATF3 induction in podocytes under chronic conditions is currently unknown. Compared with the control (C57 or BKS), ATF3 expression was elevated in animal model of proteinuria (LPS-treated C57 mice) and the model of diabetic nephropathy (db/db mice). Similarly, ATF3 was increased in high glucose (HG)-treated, lipopolysaccharide (LPS)-treated, or Ionomycin-treated podocytes in vitro. Overexpression of ATF3 increased podocyte apoptosis and decreased expression of podocin, the cell marker of podocyte; in contrast, ATF3–small interfering RNA knockdown reduced podocyte apoptosis and increased podocin expression. The translocation of ATF3 to the nucleus was increased upon stimulation. ATF3 directly modulates the regulation of NFATc1 gene promoter activity and alters the expression of Wnt6 and Fzd9, direct target genes of NFATc1 signaling. The ATF3 binding site of NFATc1 gene promoter is located at the region 671–775 base pairs upstream of the transcription start site. These results indicate a novel inducible axis of ATF3–NFAT in podocyte injury and loss. Key messages • The stress factor ATF3 is induced in podocytes from proteinuric patients, including diabetes. • ATF3 increased podocyte apoptosis and injury. • ATF3 directly modulates the regulation of NFATc1 gene promoter activity.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China.,Southern Medical University, Guangzhou, 510515, China
| | - Shun Liang
- Department of Nephrology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Yue Du
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China.,School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ruizhao Li
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Chaosheng He
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Wenjian Wang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Shuangxin Liu
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zhiming Ye
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xinling Liang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Wei Shi
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Bin Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106# Zhongshan No. 2 Road, Guangzhou, 510080, China. .,Southern Medical University, Guangzhou, 510515, China. .,School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
23
|
Tsuji K, Suleiman H, Miner JH, Daley JM, Capen DE, Păunescu TG, Lu HAJ. Ultrastructural Characterization of the Glomerulopathy in Alport Mice by Helium Ion Scanning Microscopy (HIM). Sci Rep 2017; 7:11696. [PMID: 28916834 PMCID: PMC5601433 DOI: 10.1038/s41598-017-12064-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/01/2017] [Indexed: 01/19/2023] Open
Abstract
The glomerulus exercises its filtration barrier function by establishing a complex filtration apparatus consisting of podocyte foot processes, glomerular basement membrane and endothelial cells. Disruption of any component of the glomerular filtration barrier leads to glomerular dysfunction, frequently manifested as proteinuria. Ultrastructural studies of the glomerulus by transmission electron microscopy (TEM) and conventional scanning electron microscopy (SEM) have been routinely used to identify and classify various glomerular diseases. Here we report the application of newly developed helium ion scanning microscopy (HIM) to examine the glomerulopathy in a Col4a3 mutant/Alport syndrome mouse model. Our study revealed unprecedented details of glomerular abnormalities in Col4a3 mutants including distorted podocyte cell bodies and disorganized primary processes. Strikingly, we observed abundant filamentous microprojections arising from podocyte cell bodies and processes, and presence of unique bridging processes that connect the primary processes and foot processes in Alport mice. Furthermore, we detected an altered glomerular endothelium with disrupted sub-endothelial integrity. More importantly, we were able to clearly visualize the complex, three-dimensional podocyte and endothelial interface by HIM. Our study demonstrates that HIM provides nanometer resolution to uncover and rediscover critical ultrastructural characteristics of the glomerulopathy in Col4a3 mutant mice.
Collapse
Affiliation(s)
- Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Hani Suleiman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - James M Daley
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Zand L, Glassock RJ, De Vriese AS, Sethi S, Fervenza FC. What are we missing in the clinical trials of focal segmental glomerulosclerosis? Nephrol Dial Transplant 2017; 32:i14-i21. [DOI: 10.1093/ndt/gfw324] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/28/2016] [Indexed: 01/11/2023] Open
|
25
|
Fan X, Yang H, Kumar S, Tumelty KE, Pisarek-Horowitz A, Rasouly HM, Sharma R, Chan S, Tyminski E, Shamashkin M, Belghasem M, Henderson JM, Coyle AJ, Salant DJ, Berasi SP, Lu W. SLIT2/ROBO2 signaling pathway inhibits nonmuscle myosin IIA activity and destabilizes kidney podocyte adhesion. JCI Insight 2016; 1:e86934. [PMID: 27882344 DOI: 10.1172/jci.insight.86934] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The repulsive guidance cue SLIT2 and its receptor ROBO2 are required for kidney development and podocyte foot process structure, but the SLIT2/ROBO2 signaling mechanism regulating podocyte function is not known. Here we report that a potentially novel signaling pathway consisting of SLIT/ROBO Rho GTPase activating protein 1 (SRGAP1) and nonmuscle myosin IIA (NMIIA) regulates podocyte adhesion downstream of ROBO2. We found that the myosin II regulatory light chain (MRLC), a subunit of NMIIA, interacts directly with SRGAP1 and forms a complex with ROBO2/SRGAP1/NMIIA in the presence of SLIT2. Immunostaining demonstrated that SRGAP1 is a podocyte protein and is colocalized with ROBO2 on the basal surface of podocytes. In addition, SLIT2 stimulation inhibits NMIIA activity, decreases focal adhesion formation, and reduces podocyte attachment to collagen. In vivo studies further showed that podocyte-specific knockout of Robo2 protects mice from hypertension-induced podocyte detachment and albuminuria and also partially rescues the podocyte-loss phenotype in Myh9 knockout mice. Thus, we have identified SLIT2/ROBO2/SRGAP1/NMIIA as a potentially novel signaling pathway in kidney podocytes, which may play a role in regulating podocyte adhesion and attachment. Our findings also suggest that SLIT2/ROBO2 signaling might be a therapeutic target for kidney diseases associated with podocyte detachment and loss.
Collapse
Affiliation(s)
- Xueping Fan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Hongying Yang
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Sudhir Kumar
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Kathleen E Tumelty
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Anna Pisarek-Horowitz
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Hila Milo Rasouly
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Richa Sharma
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Stefanie Chan
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Edyta Tyminski
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Michael Shamashkin
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Mostafa Belghasem
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Anthony J Coyle
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - David J Salant
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Stephen P Berasi
- Centers for Therapeutic Innovation, Pfizer Inc., Boston, Massachusetts, USA
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Papeta N, Patel A, D’Agati VD, Gharavi AG. Refinement of the HIVAN1 Susceptibility Locus on Chr. 3A1-A3 via Generation of Sub-Congenic Strains. PLoS One 2016; 11:e0163860. [PMID: 27736906 PMCID: PMC5063463 DOI: 10.1371/journal.pone.0163860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/15/2016] [Indexed: 01/19/2023] Open
Abstract
HIV-1 transgenic mice on the FVB/NJ background (TgFVB) represent a validated model of HIV-associated nephropathy (HIVAN). A major susceptibility locus, HIVAN1, was previously mapped to chromosome 3A1-A3 in a cross between TgFVB and CAST/EiJ (CAST) strains, and introgression of a 51.9 Mb segment encompassing HIVAN1 from CAST into TgFVB resulted in accelerated development of nephropathy. We generated three sub-congenic strains carrying CAST alleles in the proximal or distal regions of the HIVAN1 locus (Sub-II, 3.02–38.93 Mb; Sub-III, 38.45–55.1 Mb and Sub-IV, 47.7–55.1 Mb, build 38). At 5–10 weeks of age, histologic injury and proteinuria did not differ between HIV-1 transgenic Sub-II and TgFVB mice. In contrast, HIV-1 transgenic Sub-III and Sub-IV mice displayed up to 4.4 fold more histopathologic injury and 6-fold more albuminuria compared to TgFVB mice, similar in severity to the full-length congenic mice. The Sub-IV segment defines a maximal 7.4 Mb interval for HIVAN1, and encodes 31 protein coding genes: 15 genes have missense variants differentiating CAST from FVB, and 14 genes show differential renal expression. Of these, Frem1, Foxo1, and Setd7 have been implicated in the pathogenesis of nephropathy. HIVAN1 congenic kidneys are histologically normal without the HIV-1 transgene, yet their global transcriptome is enriched for molecular signatures of apoptosis, adenoviral infection, as well as genes repressed by histone H3 lysine 27 trimethylation, a histone modification associated with HIV-1 life cycle. These data refine HIVAN1to 7.4 Mb and identify latent molecular derangements that may predispose to nephropathy upon exposure to HIV-1.
Collapse
Affiliation(s)
- Natalia Papeta
- Department of Medicine, Columbia University, New York, New York, United States of America
| | - Ami Patel
- Department of Medicine, Columbia University, New York, New York, United States of America
| | - Vivette D. D’Agati
- Department of Pathology, Columbia University, New York, New York, United States of America
| | - Ali G. Gharavi
- Department of Medicine, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
27
|
Xu Y, Lin H, Zheng W, Ye X, Yu L, Zhuang J, Yang Q, Wang D. Matrine ameliorates adriamycin-induced nephropathy in rats by enhancing renal function and modulating Th17/Treg balance. Eur J Pharmacol 2016; 791:491-501. [PMID: 27640745 DOI: 10.1016/j.ejphar.2016.09.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/04/2016] [Accepted: 09/14/2016] [Indexed: 02/07/2023]
Abstract
Matrine (MAT) is an active alkaloid extracted from Radix Sophora flavescens. The present study was to investigate whether MAT could effectively treat Adriamycin-induced nephropathy (AIN). AIN was induced in rats using a single injection of Adriamycin (ADR). Renal interleukin-6 (IL-6), IL-10, IL-17 and transforming growth factor-β (TGF-β) levels, and the expression of forkhead box protein 3 (Foxp3) and retinoid-related orphan nuclear receptor γt (Rorγt) was measured. AIN rats developed severe albuminuria, hypoalbuminaemia, hyperlipidaemia and podocyte injury. Daily administration of MAT (100mg/kg or 200mg/kg) significantly prevented ADR-induced podocyte injury, decreased AIN symptoms and improved renal pathology manifestations. Of note, treatment with MAT (100mg/kg) plus prednisone (Pre, 5mg/kg) had equivalent efficacy to that of Pre alone (10mg/kg). Additional findings showed that ADR triggered a disordered cytokine network and abnormal expression of Foxp3 and Rorγt in rats, as reflected by increased levels of IL-6, IL-10, TGF-β, Rorγt and decreased levels of IL-10 and Foxp3. Interestingly, MAT weakened the disordered cytokine network and normalized the expression of Foxp3 and Rorγt. In addition, a significant negative correlation was observed between the values of Foxp3/Rorγt and renal pathology scores. Finally, MAT normalized regulatory T cells (Treg)/ T-helper17 cells (Th17) ratio in peripheral blood mononuclear cells of AIN rats. These data indicate MAT prevents AIN through the modification of disordered plasma lipids and recovery of renal function, and this bioactivity is at least partly attributed to the suppression of renal inflammation and the regulation of the Treg/Th17 imbalance.
Collapse
Affiliation(s)
- Yixiao Xu
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Pathophysiology, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hongzhou Lin
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wenjie Zheng
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaohua Ye
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lingfang Yu
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jieqiu Zhuang
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qing Yang
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Dexuan Wang
- Department of Pediatrics, the Second Affiliated & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
28
|
Mao N, Tan RZ, Wang SQ, Wei C, Shi XL, Fan JM, Wang L. Ginsenoside Rg1 inhibits angiotensin II-induced podocyte autophagy via AMPK/mTOR/PI3K pathway. Cell Biol Int 2016; 40:917-25. [PMID: 27296076 DOI: 10.1002/cbin.10634] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/09/2016] [Indexed: 01/07/2023]
Abstract
Recent researches have reported the extensive pharmacological activities of Ginsenoside Rg1 including antioxidant, anti-inflammatory, and anticancer properties. Furthermore Rg1 was also shown to protect various kinds of cells from self-digestion by its anti-autophagy activity. In previous studies, angiotensin II (Ang II), a key mediator of renin-angiotensin system, has been demonstrated to contribute to the progression of renal injury including abnormal autophagy. However, whether Rg1 can relieve Ang II-induced autophagy in podocyte as well as the underlying molecular mechanism remains to be elucidated. Here, we employed Ang II-treated podocyte as a model to investigate the effect of Rg1 on autophagy and the involved signal pathways. In the present study, we found that Ang II strongly promoted autophagy in immortalized mouse podocyte cells by observing the formation of autophagosomes and detecting the expression of autophagic marker, for example, LC3-II. Notably, compared to the Ang II-treated cells, treatment with Rg1 significantly inhibited the formation of autophagosomes and expression of autophagy-related proteins in Ang II pre-treated podocyte. Meanwhile, Rg1 downregulated the activity of AMPK and GSK-3β and upregulated the activity of P70S6K in Ang II-treated podocyte. In conclusion, these findings demonstrate that Ang II promotes autophagy in podocyte, and Rg1 effectively attenuates this process through AMPK/mTOR/PI3K pathway, suggesting that Rg1 may be beneficial to alleviate podocyte injury.
Collapse
Affiliation(s)
- Nan Mao
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Rui-Zhi Tan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shao-Qing Wang
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Cong Wei
- Clinical Laboratory, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin-Li Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, 050200, China
| | - Jun-Ming Fan
- Department of Nephrology, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Wang
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
29
|
Elimam H, Papillon J, Kaufman DR, Guillemette J, Aoudjit L, Gross RW, Takano T, Cybulsky AV. Genetic Ablation of Calcium-independent Phospholipase A2γ Induces Glomerular Injury in Mice. J Biol Chem 2016; 291:14468-82. [PMID: 27226532 DOI: 10.1074/jbc.m115.696781] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 12/15/2022] Open
Abstract
Glomerular visceral epithelial cells (podocytes) play a critical role in the maintenance of glomerular permselectivity. Podocyte injury, manifesting as proteinuria, is the cause of many glomerular diseases. We reported previously that calcium-independent phospholipase A2γ (iPLA2γ) is cytoprotective against complement-mediated glomerular epithelial cell injury. Studies in iPLA2γ KO mice have demonstrated an important role for iPLA2γ in mitochondrial lipid turnover, membrane structure, and metabolism. The aim of the present study was to employ iPLA2γ KO mice to better understand the role of iPLA2γ in normal glomerular and podocyte function as well as in glomerular injury. We show that deletion of iPLA2γ did not cause detectable albuminuria; however, it resulted in mitochondrial structural abnormalities and enhanced autophagy in podocytes as well as loss of podocytes in aging KO mice. Moreover, after induction of anti-glomerular basement membrane nephritis in young mice, iPLA2γ KO mice exhibited significantly increased levels of albuminuria, podocyte injury, and loss of podocytes compared with wild type. Thus, iPLA2γ has a protective functional role in the normal glomerulus and in glomerulonephritis. Understanding the role of iPLA2γ in glomerular pathophysiology provides opportunities for the development of novel therapeutic approaches to glomerular injury and proteinuria.
Collapse
Affiliation(s)
- Hanan Elimam
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Joan Papillon
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Daniel R Kaufman
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Julie Guillemette
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Lamine Aoudjit
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Richard W Gross
- the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Tomoko Takano
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| | - Andrey V Cybulsky
- From the Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec H4A 3J1, Canada and
| |
Collapse
|
30
|
Lichtnekert J, Kaverina NV, Eng DG, Gross KW, Kutz JN, Pippin JW, Shankland SJ. Renin-Angiotensin-Aldosterone System Inhibition Increases Podocyte Derivation from Cells of Renin Lineage. J Am Soc Nephrol 2016; 27:3611-3627. [PMID: 27080979 DOI: 10.1681/asn.2015080877] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/20/2016] [Indexed: 12/17/2022] Open
Abstract
Because adult podocytes cannot proliferate and are therefore unable to self-renew, replacement of these cells depends on stem/progenitor cells. Although podocyte number is higher after renin-angiotensin-aldosterone system (RAAS) inhibition in glomerular diseases, the events explaining this increase are unclear. Cells of renin lineage (CoRL) have marked plasticity, including the ability to acquire a podocyte phenotype. To test the hypothesis that RAAS inhibition partially replenishes adult podocytes by increasing CoRL number, migration, and/or transdifferentiation, we administered tamoxifen to Ren1cCreERxRs-tdTomato-R CoRL reporter mice to induce permanent labeling of CoRL with red fluorescent protein variant tdTomato. We then induced experimental FSGS, typified by abrupt podocyte depletion, with a cytopathic antipodocyte antibody. RAAS inhibition by enalapril (angiotensin-converting enzyme inhibitor) or losartan (angiotensin-receptor blocker) in FSGS mice stimulated the proliferation of CoRL, increasing the reservoir of these cells in the juxtaglomerular compartment (JGC). Compared with water or hydralazine, RAAS inhibition significantly increased the migration of CoRL from the JGC to the intraglomerular compartment (IGC), with more glomeruli containing RFP+CoRL and, within these glomeruli, more RFP+CoRL. Moreover, RAAS inhibition in FSGS mice increased RFP+CoRL transdifferentiation in the IGC to phenotypes, consistent with those of podocytes (coexpression of synaptopodin and Wilms tumor protein), parietal epithelial cells (PAX 8), and mesangial cells (α8 integrin). These results show that in the context of podocyte depletion in FSGS, RAAS inhibition augments CoRL proliferation and plasticity toward three different glomerular cell lineages.
Collapse
Affiliation(s)
| | | | | | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - J Nathan Kutz
- Department of Applied Mathematics, University of Washington, Seattle, Washington; and
| | | | | |
Collapse
|
31
|
Yu H, Artomov M, Brähler S, Stander MC, Shamsan G, Sampson MG, White JM, Kretzler M, Miner JH, Jain S, Winkler CA, Mitra RD, Kopp JB, Daly MJ, Shaw AS. A role for genetic susceptibility in sporadic focal segmental glomerulosclerosis. J Clin Invest 2016; 126:1067-78. [PMID: 26901816 DOI: 10.1172/jci82592] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 01/07/2016] [Indexed: 12/20/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a syndrome that involves kidney podocyte dysfunction and causes chronic kidney disease. Multiple factors including chemical toxicity, inflammation, and infection underlie FSGS; however, highly penetrant disease genes have been identified in a small fraction of patients with a family history of FSGS. Variants of apolipoprotein L1 (APOL1) have been linked to FSGS in African Americans with HIV or hypertension, supporting the proposal that genetic factors enhance FSGS susceptibility. Here, we used sequencing to investigate whether genetics plays a role in the majority of FSGS cases that are identified as primary or sporadic FSGS and have no known cause. Given the limited number of biopsy-proven cases with ethnically matched controls, we devised an analytic strategy to identify and rank potential candidate genes and used an animal model for validation. Nine candidate FSGS susceptibility genes were identified in our patient cohort, and three were validated using a high-throughput mouse method that we developed. Specifically, we introduced a podocyte-specific, doxycycline-inducible transactivator into a murine embryonic stem cell line with an FSGS-susceptible genetic background that allows shRNA-mediated targeting of candidate genes in the adult kidney. Our analysis supports a broader role for genetic susceptibility of both sporadic and familial cases of FSGS and provides a tool to rapidly evaluate candidate FSGS-associated genes.
Collapse
|
32
|
Chew-Harris JSC, Florkowski CM, Elmslie JL, Livesey J, Endre ZH, George PM. Lean mass modulates glomerular filtration rate in males of normal and extreme body composition. Intern Med J 2015; 44:749-56. [PMID: 24863461 DOI: 10.1111/imj.12479] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 05/05/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND Understanding determinants of glomerular filtration rate (GFR) is important in aiding prediction and interpretation of kidney function. Body composition is known to affect GFR but is not included in current screening of kidney disease. We investigated the association between GFR and body composition in healthy young men with differing body mass but without known diabetes or kidney injury. METHODS Three groups were recruited: normal BMI (n = 22) with a body mass index (BMI) <25 kg/m(2) , muscular (n = 23) with BMI ≥30 kg/m(2) and bioelectrical impedance body fat ≤20% and obese (n = 22) with BMI ≥30 kg/m(2) and bioelectrical impedance body fat ≥30%. Dietary analyses, GFR clearance by (99m) Tc-DTPA, urine protein and body composition by dual-energy X-ray absorptiometry were measured in all participants. Linear and nonlinear associations of constituents of body composition with GFR were assessed. RESULTS Muscular men had a higher GFR (mean 186.4 mL/min; 95% CI 171.7-201.1) than normal BMI and obese groups (P = 0.0007). Urine protein and albumin excretion were not elevated in any participants. On multiple regression analysis (r(2) = 0.60), the variables with strong associations with GFR were age (P = 0.0009) and lean mass (P = 0.0001). Fat mass, protein intake and smoking status were not associated. Skeletal muscle mass correlated significantly with GFR in all subgroups. CONCLUSION Age and lean mass were strong determinants of GFR. Estimates of GFR should therefore be indexed to an estimate of lean mass.
Collapse
Affiliation(s)
- J S C Chew-Harris
- Clinical Biochemistry Unit, Canterbury Health Laboratories, Christchurch, New Zealand; Christchurch School of Medicine, University of Otago, Christchurch, New Zealand
| | | | | | | | | | | |
Collapse
|
33
|
Li Z, Zhang L, Shi W, Chen Y, Zhang H, Liu S, Liang X, Ling T, Yu C, Huang Z, Tan X, Zhao X, Ye Z, Zhang B, Wang W, Li R, Ma J. Spironolactone inhibits podocyte motility via decreasing integrin β1 and increasing integrin β3 in podocytes under high-glucose conditions. Mol Med Rep 2015; 12:6849-6854. [PMID: 26352002 DOI: 10.3892/mmr.2015.4295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 07/29/2015] [Indexed: 01/19/2023] Open
Abstract
Integrin β1 and β3 expression by podocytes is required to maintain glomerular structural integrity. Previous studies have shown that aldosterone (ALD) is involved in glomerular podocyte injury, and mineralocorticoid receptor (MR) blocker spironolactone effectively reduces proteinuria in patients with diabetic nephropathy. The present study was designed to observe the effects of spironolactone on β1 and β3 integrin expression and podocyte motility under in vitro diabetic conditions. Immortalized mouse podocytes were cultured in media containing normal glucose (NG) levels, high glucose (HG) or HG plus spironolacton. The expression of β1 and β3 integrin in podocytes was detected by reverse transcription quantitative polymerase chain reaction, immunofluorescence and western blot analyses. The effects of spironolacton on podocyte motility was further evaluated using a wound healing assay. HG stimulation markedly decreased mRNA and protein expression of integrin β1, and significantly increased mRNA and protein expression of integrin β3 in cultured podocytes. However, simultaneous treatment with spironolacton (10‑7 mol/l) significantly attenuated HG-mediated increases in integrin β3 and decreases in integrin β1 expression. Furthermore, the migration of podocytes induced by HG was abrogated by concomitant treatment with spironolacton. In conclusion, the present study suggested that HG decreased the expression of integrin β1 in cultured podocytes, accompanied with an increase of integrin β3. Spironolactone inhibited cell motility and stabilized podoctyes treated with HG, probably through partly normalizing the expression of integrin β1 and decreasing the expression of integrin β3.
Collapse
Affiliation(s)
- Zhuo Li
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Li Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wei Shi
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yuanhan Chen
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hong Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shuangxin Liu
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xinling Liang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ting Ling
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Chunping Yu
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhongshun Huang
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaofan Tan
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xinchen Zhao
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhiming Ye
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bin Zhang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wenjian Wang
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ruizhao Li
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Jianchao Ma
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
34
|
De Vriese AS, Fervenza FC. Con: Biomarkers in glomerular diseases: putting the cart before the wheel? Nephrol Dial Transplant 2015; 30:885-90. [DOI: 10.1093/ndt/gfv107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
35
|
Ortiz A, Sanchez-Niño MD, Izquierdo MC, Martin-Cleary C, Garcia-Bermejo L, Moreno JA, Ruiz-Ortega M, Draibe J, Cruzado JM, Garcia-Gonzalez MA, Lopez-Novoa JM, Soler MJ, Sanz AB. Translational value of animal models of kidney failure. Eur J Pharmacol 2015; 759:205-20. [PMID: 25814248 DOI: 10.1016/j.ejphar.2015.03.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/08/2015] [Accepted: 03/12/2015] [Indexed: 11/28/2022]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are associated with decreased renal function and increased mortality risk, while the therapeutic armamentarium is unsatisfactory. The availability of adequate animal models may speed up the discovery of biomarkers for disease staging and therapy individualization as well as design and testing of novel therapeutic strategies. Some longstanding animal models have failed to result in therapeutic advances in the clinical setting, such as kidney ischemia-reperfusion injury and diabetic nephropathy models. In this regard, most models for diabetic nephropathy are unsatisfactory in that they do not evolve to renal failure. Satisfactory models for additional nephropathies are needed. These include anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, IgA nephropathy, anti-phospholipase-A2-receptor (PLA2R) membranous nephropathy and Fabry nephropathy. However, recent novel models hold promise for clinical translation. Thus, the AKI to CKD translation has been modeled, in some cases with toxins of interest for human CKD such as aristolochic acid. Genetically modified mice provide models for Alport syndrome evolving to renal failure that have resulted in clinical recommendations, polycystic kidney disease models that have provided clues for the development of tolvaptan, that was recently approved for the human disease in Japan; and animal models also contributed to target C5 with eculizumab in hemolytic uremic syndrome. Some ongoing trials explore novel concepts derived from models, such TWEAK targeting as tissue protection for lupus nephritis. We now review animal models reproducing diverse, genetic and acquired, causes of AKI and CKD evolving to kidney failure and discuss the contribution to clinical translation and prospects for the future.
Collapse
Affiliation(s)
- Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain; IRSIN, Madrid, Spain
| | | | - Maria C Izquierdo
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain
| | | | - Laura Garcia-Bermejo
- REDinREN, Madrid, Spain; Dpt. of Pathology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain
| | - Juan A Moreno
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Marta Ruiz-Ortega
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain
| | - Juliana Draibe
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Cruzado
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Miguel A Garcia-Gonzalez
- REDinREN, Madrid, Spain; Laboratorio de Nefrología, Complexo Hospitalario de Santiago de Compostela (CHUS), Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Jose M Lopez-Novoa
- REDinREN, Madrid, Spain; Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamnca, Spain
| | - Maria J Soler
- REDinREN, Madrid, Spain; Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Ana B Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain.
| | | |
Collapse
|
36
|
Madan A. Repository corticotropin injection in a patient presenting with focal segmental glomerulosclerosis, rheumatoid arthritis, and optic neuritis: a case report. Int J Gen Med 2015; 8:119-24. [PMID: 25848316 PMCID: PMC4374712 DOI: 10.2147/ijgm.s79815] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Focal segmental glomerulosclerosis (FSGS) causes scarring or sclerosis of glomeruli that act as tiny filters in the kidneys, damage to which results in diminished ability to properly filter blood, resulting in the urinary loss of plasma proteins and subsequent proteinuria. Case presentation A 60-year-old, white female with a history of intermittent proteinuria was referred by her primary care physician for renal dysfunction. Biopsy confirmed FSGS and she was treated with an angiotensin-converting enzyme inhibitor. She also had rheumatoid arthritis (RA) but no active synovitis and was maintained on prednisone 5 mg/d. She also complained of worsening vision in her right eye and was diagnosed with optic neuritis (ON). She remained stable for about 8 months when examination indicated FSGS relapse, and she reported painful RA flares. She was treated with Acthar® Gel (40 mg biweekly) for 6 months, after which proteinuria and urine protein-to-creatinine ratio decreased to about half. Her ON improved, and she reported that she had fewer RA flares and pain improved by 50%. This case of confirmed FSGS showed an improved response to treatment with Acthar Gel for FSGS with concomitant RA and ON. Conclusion This referral case is relevant to primary care practitioners who treat disorders that may be responsive to corticosteroid therapy. The antiproteinuric effects and ancillary improvement in RA and ON symptoms during treatment with Acthar Gel are not entirely explained by its steroidogenic actions. ACTH is a bioactive peptide that, together with α-melanocyte-stimulating hormone, exhibits biologic efficacy by modulating proinflammatory cytokines and subsequent leukocyte extravasation and may have autocrine/paracrine effects in joints. While Acthar Gel was primarily administered in this case to treat proteinuria, it also showed ancillary benefits in patients with concomitant inflammatory disease states.
Collapse
Affiliation(s)
- Arvind Madan
- Nephrology Associates of Central Florida, PA, Orlando, FL, USA
| |
Collapse
|
37
|
Kobayashi N, Ueno T, Ohashi K, Yamashita H, Takahashi Y, Sakamoto K, Manabe S, Hara S, Takashima Y, Dan T, Pastan I, Miyata T, Kurihara H, Matsusaka T, Reiser J, Nagata M. Podocyte injury-driven intracapillary plasminogen activator inhibitor type 1 accelerates podocyte loss via uPAR-mediated β1-integrin endocytosis. Am J Physiol Renal Physiol 2015; 308:F614-26. [PMID: 25587125 PMCID: PMC4360033 DOI: 10.1152/ajprenal.00616.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/08/2015] [Indexed: 11/22/2022] Open
Abstract
Podocyte-endothelial cell cross-talk is paramount for maintaining the filtration barrier. The present study investigated the endothelial response to podocyte injury and its subsequent role in glomerulosclerosis using the podocyte-specific injury model of NEP25/LMB2 mice. NEP25/LMB2 mice showed proteinuria and local podocyte loss accompanied by thrombotic microangiopathy on day 8. Mice showed an increase of glomerular plasminogen activator inhibitor type 1 (PAI-1) mRNA and aberrant endothelial PAI-1 protein already on day 1, before thrombosis and proteinuria. A PAI-1-specific inhibitor reduced proteinuria and thrombosis and preserved podocyte numbers in NEP25/LMB2 mice by stabilization of β1-integrin translocation. Heparin loading significantly reduced thrombotic formation, whereas proteinuria and podocyte numbers were unchanged. Immortalized podocytes treated with PAI-1 and the urokinase plasminogen activator (uPA) complex caused significant cell detachment, whereas podocytes treated with PAI-1 or uPA alone or with the PAI-1/uPA complex pretreated with an anti-uPA receptor (uPAR) antibody failed to cause detachment. Confocal microscopy and cell surface biotinylation experiments showed that internalized β1-integrin was found together with uPAR in endocytotic vesicles. The administration of PAI-1 inhibitor or uPAR-blocking antibody protected cultured podocytes from cell detachment. In conclusion, PAI-1/uPA complex-mediated uPAR-dependent podocyte β1-integrin endocytosis represents a novel mechanism of glomerular injury leading to progressive podocytopenia. This aberrant cross-talk between podocytes and endothelial cells represents a feedforward injury response driving podocyte loss and progressive glomerulosclerosis.
Collapse
Affiliation(s)
- Namiko Kobayashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Toshiharu Ueno
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kumi Ohashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hanako Yamashita
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yukina Takahashi
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuo Sakamoto
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shun Manabe
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Hara
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yasutoshi Takashima
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Takashi Dan
- Center for Translational and Advanced Research, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshio Miyata
- Center for Translational and Advanced Research, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hidetake Kurihara
- Department of Anatomy, Juntendo University School of Medicine, Bunkyo, Tokyo, Japan
| | - Taiji Matsusaka
- Department of Internal Medicine, Institute of Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan; and
| | - Jochen Reiser
- Department of Medicine, Rush University, Chicago, Illinois
| | - Michio Nagata
- Department of Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan;
| |
Collapse
|
38
|
Abstract
Wnt/β-catenin signaling is an evolutionarily conserved, highly complex, key developmental pathway that regulates cell fate, organ development, tissue homeostasis, as well as injury and repair. Although relatively silent in normal adult kidney, Wnt/β-catenin signaling is re-activated after renal injury in a wide variety of animal models and in human kidney disorders. Whereas some data point to a protective role of this signaling in healing and repair after acute kidney injury, increasing evidence suggests that sustained activation of Wnt/β-catenin is associated with the development and progression of renal fibrotic lesions. In kidney cells, Wnt/β-catenin promotes the expression of numerous fibrosis-related genes such as Snail1, plasminogen activator inhibitor-1, and matrix metalloproteinase-7. Recent studies also indicate that multiple components of the renin-angiotensin system are the direct downstream targets of Wnt/β-catenin. Consistently, inhibition of Wnt/β-catenin signaling by an assortment of strategies ameliorates kidney injury and mitigates renal fibrotic lesions in various models of chronic kidney disease, suggesting that targeting this signaling could be a plausible strategy for therapeutic intervention. In this mini review, we will briefly discuss the regulation, downstream targets, and mechanisms of Wnt/β-catenin signaling in the pathogenesis of kidney fibrosis.
Collapse
Affiliation(s)
- Roderick J Tan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dong Zhou
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lili Zhou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
39
|
Olea-Herrero N, Arenas MI, Muñóz-Moreno C, Moreno-Gómez-Toledano R, González-Santander M, Arribas I, Bosch RJ. Bisphenol-A induces podocytopathy with proteinuria in mice. J Cell Physiol 2014; 229:2057-66. [PMID: 24809654 DOI: 10.1002/jcp.24665] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 05/06/2014] [Indexed: 12/19/2022]
Abstract
Bisphenol-A, a chemical used in the production of the plastic lining of food and beverage containers, can be found in significant levels in human fluids. Recently, bisphenol-A has been associated with low-grade albuminuria in adults as well as in children. Since glomerular epithelial cells (podocytes) are commonly affected in proteinuric conditions, herein we explored the effects of bisphenol-A on podocytes in vitro and in vivo. On cultured podocytes we first observed that bisphenol-A-at low or high concentrations-(10 nM and 100 nM, respectively) was able to induce hypertrophy, diminish viability, and promote apoptosis. We also found an increase in the protein expression of TGF-β1 and its receptor, the cyclin-dependent kinase inhibitor p27Kip1, as well as collagen-IV, while observing a diminished expression of the slit diaphragm proteins nephrin and podocin. Furthermore, mice intraperitoneally injected with bisphenol-A (50 mg/Kg for 5 weeks) displayed an increase in urinary albumin excretion and endogenous creatinine clearance. Renal histology showed mesangial expansion. At ultrastructural level, podocytes displayed an enlargement of both cytoplasm and foot processes as well as the presence of condensed chromatin, suggesting apoptosis. Furthermore, immunohistochemistry for WT-1 (specific podocyte marker) and the TUNEL technique showed podocytopenia as well as the presence of apoptosis, respectively. In conclusion, our data demonstrate that Bisphenol-A exposure promotes a podocytopathy with proteinuria, glomerular hyperfiltration and podocytopenia. Further studies are needed to clarify the potential role of bisphenol-A in the pathogenesis as well as in the progression of renal diseases.
Collapse
Affiliation(s)
- Nuria Olea-Herrero
- Laboratory of Renal Physiology and Experimental Nephrology, Department of System Biology/Physiology Unit, University of Alcalá, Alcalá de Henares (28871), Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Riezzo I, Turillazzi E, Bello S, Cantatore S, Cerretani D, Di Paolo M, Fiaschi AI, Frati P, Neri M, Pedretti M, Fineschi V. Chronic nandrolone administration promotes oxidative stress, induction of pro-inflammatory cytokine and TNF-α mediated apoptosis in the kidneys of CD1 treated mice. Toxicol Appl Pharmacol 2014; 280:97-106. [PMID: 25065671 DOI: 10.1016/j.taap.2014.06.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 06/14/2014] [Accepted: 06/15/2014] [Indexed: 12/26/2022]
Abstract
Nandrolone decanoate administration and strenuous exercise increase the extent of renal damage in response to renal toxic injury. We studied the role played by oxidative stress in the apoptotic response caused by nandrolone decanoate in the kidneys of strength-trained male CD1 mice. To measure cytosolic enzyme activity, glutathione peroxidase (GPx), glutathione reductase (GR) and malondialdehyde (MDA) were determined after nandrolone treatment. An immunohistochemical study and Western blot analysis were performed to evaluate cell apoptosis and to measure the effects of renal expression of inflammatory mediators (IL-1β, TNF-α) on the induction of apoptosis (HSP90, TUNEL). Dose-related oxidative damage in the kidneys of treated mice is shown by an increase in MDA levels and by a reduction of antioxidant enzyme GR and GPx activities, resulting in the kidney's reduced radical scavenging ability. Renal specimens of the treated group showed relevant glomeruli alterations and increased immunostaining and protein expressions, which manifested significant focal segmental glomerulosclerosis. The induction of proinflammatory cytokine expression levels was confirmed by Western blot analysis. Long-term administration of nandrolone promotes oxidative injury in the mouse kidneys. TNF-α mediated injury due to nandrolone in renal cells appears to play a role in the activation of both the intrinsic and extrinsic apoptosis pathways.
Collapse
Affiliation(s)
- Irene Riezzo
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | | | - Stefania Bello
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | - Santina Cantatore
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | - Daniela Cerretani
- Pharmacology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Marco Di Paolo
- Department of Forensic Pathology, University of Pisa, Pisa, Italy
| | - Anna Ida Fiaschi
- Pharmacology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, University of Rome Sapienza, Viale Regina Elena 336, 00161 Rome, Italy
| | - Margherita Neri
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | - Monica Pedretti
- Department of Forensic Pathology, University of Pisa, Pisa, Italy
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, University of Rome Sapienza, Viale Regina Elena 336, 00161 Rome, Italy.
| |
Collapse
|
41
|
Sethi S, Zand L, Nasr SH, Glassock RJ, Fervenza FC. Focal and segmental glomerulosclerosis: clinical and kidney biopsy correlations. Clin Kidney J 2014; 7:531-7. [PMID: 25503953 PMCID: PMC4240407 DOI: 10.1093/ckj/sfu100] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/02/2014] [Indexed: 01/26/2023] Open
Abstract
Background Primary focal segmental glomerulosclerosis (FSGS) is a common glomerular disease in adults and ranks among the top causes of a primary glomerular disease causing end-stage renal disease (ESRD). Primary FSGS is, however, a diagnosis of exclusion and distinction between primary versus secondary FSGS is not always obvious, resulting in a number of patients with secondary FSGS undergoing unnecessary immunosuppressive therapy. Methods We reviewed the Mayo Clinic Renal Pathology Database for patients with a diagnosis of FSGS on native renal biopsy and divided the patients into nephrotic syndrome-associated (NS-associated) and non-nephrotic syndrome-associated (NNS-associated) FSGS as a first approximation followed by dividing the lesion according to the degree of foot process effacement (FPE) on electron microscopy (EM) examination. Results A total of 41 patients with FSGS with complete evaluation were identified. Of these, 18 were classified as having NS and 23 were classified as having NNS. Baseline characteristics (age, gender, body mass index, serum creatinine and hematuria) were not different between the groups. All of the patients with NS showed diffuse FPE ranging from 80 to 100% (mean 96%). On the other hand, of the 23 patients in the NNS group, 22 had segmental FPE and showed patchy effacement, with all cases showing 20–60% FPE (mean of 48%). Conclusion Adult patients presenting with NS, an FSGS lesion on LM, extensive FPE (≥80%) on EM examination and no risk factors associated with secondary FSGS are likely to have primary FSGS. Conversely, the absence of NS in a patient with segmental FPE on EM strongly suggests a secondary FSGS. Dividing FSGS into the presence or absence of NS together with the degree of FPE on EM examination is more helpful as it provides a more practical way to separate patients into cases of primary versus secondary FSGS.
Collapse
Affiliation(s)
- Sanjeev Sethi
- Division of Anatomic Pathology , Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Ladan Zand
- Division of Nephrology and Hypertension , Mayo Clinic College of Medicine , Rochester, MN , USA
| | - Samih H Nasr
- Division of Anatomic Pathology , Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Richard J Glassock
- Geffen School of Medicine School at UCLA , Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Fernando C Fervenza
- Division of Nephrology and Hypertension , Mayo Clinic College of Medicine , Rochester, MN , USA
| |
Collapse
|
42
|
Zeng C, Fan Y, Wu J, Shi S, Chen Z, Zhong Y, Zhang C, Zen K, Liu Z. Podocyte autophagic activity plays a protective role in renal injury and delays the progression of podocytopathies. J Pathol 2014; 234:203-13. [PMID: 24870816 DOI: 10.1002/path.4382] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/10/2014] [Accepted: 05/20/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Caihong Zeng
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Yun Fan
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Junnan Wu
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Shaolin Shi
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Zhaohong Chen
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Yongzhong Zhong
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Changming Zhang
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| | - Ke Zen
- JERC-MBB, State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University School of Life Sciences; Nanjing Jiangsu 210093 China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases; Jinling Hospital, Nanjing University School of Medicine; Nanjing Jiangsu 210002 China
| |
Collapse
|
43
|
Yang SM, Chan YL, Hua KF, Chang JM, Chen HL, Tsai YJ, Hsu YJ, Chao LK, Feng-Ling Y, Tsai YL, Wu SH, Wang YF, Tsai CL, Chen A, Ka SM. Osthole improves an accelerated focal segmental glomerulosclerosis model in the early stage by activating the Nrf2 antioxidant pathway and subsequently inhibiting NF-κB-mediated COX-2 expression and apoptosis. Free Radic Biol Med 2014; 73:260-9. [PMID: 24858719 DOI: 10.1016/j.freeradbiomed.2014.05.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 05/03/2014] [Accepted: 05/06/2014] [Indexed: 01/06/2023]
Abstract
Inflammatory reactions and oxidative stress are implicated in the pathogenesis of focal segmental glomerulosclerosis (FSGS), a common chronic kidney disease with relatively poor prognosis and unsatisfactory treatment regimens. Previously, we showed that osthole, a coumarin compound isolated from the seeds of Cnidium monnieri, can inhibit reactive oxygen species generation, NF-κB activation, and cyclooxygenase-2 expression in lipopolysaccharide-activated macrophages. In this study, we further evaluated its renoprotective effect in a mouse model of accelerated FSGS (acFSGS), featuring early development of proteinuria, followed by impaired renal function, glomerular epithelial cell hyperplasia lesions (a sensitive sign that precedes the development of glomerular sclerosis), periglomerular inflammation, and glomerular hyalinosis/sclerosis. The results show that osthole significantly prevented the development of the acFSGS model in the treated group of mice. The mechanisms involved in the renoprotective effects of osthole on the acFSGS model were mainly a result of an activated Nrf2-mediated antioxidant pathway in the early stage (proteinuria and ischemic collapse of the glomeruli) of acFSGS, followed by a decrease in: (1) NF-κB activation and COX-2 expression as well as PGE2 production, (2) podocyte injury, and (3) apoptosis. Our data support that targeting the Nrf2 antioxidant pathway may justify osthole being established as a candidate renoprotective compound for FSGS.
Collapse
Affiliation(s)
- Shun-Min Yang
- Department of Pathology and National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Yi-Lin Chan
- Department of Pathology and National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan, Republic of China
| | - Jia-Ming Chang
- Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan, Republic of China
| | - Hui-Ling Chen
- Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan, Republic of China
| | - Yung-Jen Tsai
- Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan, Republic of China
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital; National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Louis Kuoping Chao
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan, Republic of China
| | - Yang Feng-Ling
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yu-Ling Tsai
- Graduate Institute of Life Sciences; and National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Shih-Hsiung Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yih-Fuh Wang
- Graduate Institute of Electrical Engineering and Computer Science, National Penghu University of Science and Technology, Penghu, Taiwan, Republic of China
| | - Change-Ling Tsai
- Graduate Institute of Electrical Engineering and Computer Science, National Penghu University of Science and Technology, Penghu, Taiwan, Republic of China
| | - Ann Chen
- Department of Pathology and National Defense Medical Center, Taipei 114, Taiwan, Republic of China
| | - Shuk-Man Ka
- Graduate Institute of Aerospace and Undersea Medicine, School of Medicine; National Defense Medical Center, Taipei 114, Taiwan, Republic of China.
| |
Collapse
|
44
|
Penning ME, Bloemenkamp KWM, van der Zon T, Zandbergen M, Schutte JM, Bruijn JA, Bajema IM, Baelde HJ. Association of preeclampsia with podocyte turnover. Clin J Am Soc Nephrol 2014; 9:1377-85. [PMID: 25035270 DOI: 10.2215/cjn.12811213] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND OBJECTIVES Preeclampsia is characterized by hypertension and proteinuria, and increased shedding of podocytes into the urine is a common finding. This finding raises the question of whether preeclamptic nephropathy involves podocyte damage. This study examined podocyte-related changes in a unique sample of renal tissues obtained from women who died of preeclampsia. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS All patients with preeclampsia who died in The Netherlands since 1990 and had available autopsy tissue were identified using a nationwide database of the Dutch Pathology Registry (PALGA). This resulted in a cohort of 11 women who died from preeclampsia. Three control groups were also identified during the same time period, and consisted of normotensive women who died during pregnancy (n=25), and nonpregnant controls either with (n=14) or without (n=13) chronic hypertension. Glomerular lesions, including podocyte numbers, podocyte proliferation, and parietal cell activation, were measured. RESULTS Patients with preeclampsia had prominent characteristic glomerular lesions. The results showed that the number of podocytes per glomerulus did not differ significantly between the patients with preeclampsia and the control groups. However, preeclampsia was associated with a significant increase in intraglomerular cell proliferation (7.3% [SD 9.4] of the glomeruli of patients with preeclampsia had Ki-67-positive cells versus 1.6% [SD 3.3] of the glomeruli of hypertensive controls and 1.1% [SD 1.3] of nonpregnant controls; P=0.004) and activated parietal epithelial cells on a podocyte location (34% [SD 13.1] of the glomeruli of patients with preeclampsia versus 18.0% [SD 15.3] of pregnant controls, 11.9% [SD 13.2] of hypertensive controls, and 10.8% [SD 13.4] of nonpregnant controls; P=0.01). CONCLUSIONS These findings suggest that the recently described mechanisms of podocyte replacement play a role in preeclampsia. These results provide key new insights into the pathogenesis of preeclamptic nephropathy, and they open new possibilities for developing therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | - Joke M Schutte
- Department of Obstetrics and Gynecology, Isala Zwolle, Zwolle, The Netherlands
| | | | | | | |
Collapse
|
45
|
Wei M, Li Z, Yang Z. Crosstalk between protective autophagy and NF-κB signal in high glucose-induced podocytes. Mol Cell Biochem 2014; 394:261-73. [PMID: 24957786 DOI: 10.1007/s11010-014-2102-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/15/2014] [Indexed: 01/07/2023]
Abstract
Despite a great deal of recent studies focused on the pivotal role of autophagy in maintaining podocyte energy homeostasis, the mechanisms of autophagy in regulating transcriptional factors under high glucose (HG) condition are not fully understood. Here, we evaluated the effect of HG on nuclear factor-kappa B (NF-κB) signaling and autophagic process. The results showed that HG promoted autophagy in podocytes. Bafilomycin A1 (Baf A1) further enhanced this effect, but 3-methyadenine (3-MA) inhibited it. The proautophagic effects of HG manifested in the form of enhanced podocyte expression of light chain 3 (LC3)-II. In these cells, blockade of NF-κB signal by ammonium pyrrolidinethiocarbamate constrained in effectively reducing LC3-II up-regulation and increasing podocyte apoptosis. Furthermore, the autophagy inhibitors, such as Baf A1 and 3-MA, significantly enhanced HG-induced NF-κB activation and increased apoptosis. Thus, we conclude that the accumulation of autophagosomes results from enhancement of the autophagic flux, but not the blockage of autophagosome-lysosome fusion by HG. We also prove that HG-induced apoptosis, autophagy, and NF-κB signal are in a close crosstalk through a yet undetermined mechanism in podocytes.
Collapse
Affiliation(s)
- Miaomiao Wei
- College of Medicine, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | | | | |
Collapse
|
46
|
Huang J, Liu G, Zhang YM, Cui Z, Wang F, Liu XJ, Chu R, Zhao MH. Urinary soluble urokinase receptor levels are elevated and pathogenic in patients with primary focal segmental glomerulosclerosis. BMC Med 2014; 12:81. [PMID: 24884842 PMCID: PMC4064821 DOI: 10.1186/1741-7015-12-81] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/31/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a major cause of end-stage renal disease. Recent studies have proposed that plasma soluble urokinase receptor (suPAR) might be a causative circulating factor but this proposal has caused controversy. This study aimed to measure urinary suPAR levels in patients with primary FSGS and its significance in the pathogenesis of FSGS. METHODS Sixty-two patients with primary FSGS, diagnosed between January 2006 and January 2012, with complete clinical and pathologic data were enrolled, together with disease and normal controls. Urinary suPAR levels were measured using commercial ELISA kits and were corrected by urinary creatinine (Cr). The associations between urinary suPAR levels and clinical data at presentation and during follow up were analyzed. Conditionally immortalized human podocytes were used to study the effect of urinary suPAR on activating β3 integrin detected by AP5 staining. RESULTS The urinary suPAR level of patients with primary FSGS (500.56, IQR 262.78 to 1,059.44 pg/μmol Cr) was significantly higher than that of patients with minimal change disease (307.86, IQR 216.54 to 480.18 pg/μmol Cr, P = 0.033), membranous nephropathy (250.23, IQR 170.37 to 357.59 pg/μmol Cr, P <0.001), secondary FSGS (220.45, IQR 149.38 to 335.54 pg/μmol Cr, P <0.001) and normal subjects (183.59, IQR 103.92 to 228.78 pg/μmol Cr, P <0.001). The urinary suPAR level of patients with cellular variant was significantly higher than that of patients with tip variant. The urinary suPAR level in the patients with primary FSGS was positively correlated with 24-hour urine protein (r = 0.287, P = 0.024). During follow up, the urinary suPAR level of patients with complete remission decreased significantly (661.19, IQR 224.32 to 1,115.29 pg/μmol Cr versus 217.68, IQR 121.77 to 415.55 pg/μmol Cr, P = 0.017). The AP5 signal was strongly induced along the cell membrane when human differentiated podocytes were incubated with the urine of patients with FSGS at presentation, and the signal could be reduced by a blocking antibody specific to uPAR. CONCLUSIONS Urinary suPAR was specifically elevated in patients with primary FSGS and was associated with disease severity. The elevated urinary suPAR could activate β3 integrin on human podocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ming-hui Zhao
- Renal Division, Peking University First Hospital, Beijing, PR China.
| |
Collapse
|
47
|
Fervenza FC, Perazella MA, Choi MJ. American Society of Nephrology Quiz and Questionnaire 2013: Glomerulonephritis. Clin J Am Soc Nephrol 2014; 9:987-96. [DOI: 10.2215/cjn.11571113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Na+/H+ exchanger-1 reduces podocyte injury caused by endoplasmic reticulum stress via autophagy activation. J Transl Med 2014; 94:439-54. [PMID: 24566932 DOI: 10.1038/labinvest.2014.4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/14/2013] [Accepted: 12/30/2013] [Indexed: 12/26/2022] Open
Abstract
Podocyte injury has a critical role in the pathogenesis of proteinuria. Induction of endoplasmic reticulum (ER) stress is thought to lead to podocyte injury; however, no effective strategy for reducing ER stress-induced injury has been identified. We investigated specific mechanisms for reducing podocyte injury caused by ER stress. We found that the induction of ER stress in podocytes was related to cytoskeleton injury and increased proteinuria, which was associated with autophagy activation and downregulation of Na(+)/H(+) exchanger-1 (NHE-1) in the rat model of passive Heymann nephritis. Using mouse podocyte cells (MPCs), we showed that ER stress could lead to podocyte injury accompanied by autophagy activation, and the disturbance of autophagy aggravated cytoskeleton loss under conditions of ER stress. The balance between autophagy activation and ER stress was critical to podocyte survival, in which the efficiency of autophagy could have a pivotal role. Strikingly, the overexpression and small interfering RNA knockdown of NHE-1 results suggested that NHE-1 exerts a protective effect by reducing the loss of synaptopodin in MPCs exposed to ER stress. This protective mechanism involves NHE-1 activation of autophagy via the PI3K/Akt pathway to reduce ER stress injury in podocytes. This mechanism may provide a new pathway to prevent podocyte injury.
Collapse
|
49
|
Sethi S, Glassock RJ, Fervenza FC. Focal segmental glomerulosclerosis: towards a better understanding for the practicing nephrologist. Nephrol Dial Transplant 2014; 30:375-84. [PMID: 24589721 DOI: 10.1093/ndt/gfu035] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Focal and segmental glomerulosclerosis (FSGS) is a common histopathological lesion that can represent a primary podocytopathy, or occur as an adaptive phenomenon consequent to nephron mass reduction, a scar from a healing vasculitic lesion, direct drug toxicity or viral infection among other secondary causes. Thus, the presence of an FSGS lesion in a renal biopsy does not confer a disease diagnosis, but rather represents the beginning of an exploratory process, hopefully leading ultimately to identification of a specific etiology and its appropriate treatment. We define primary FSGS as a 'primary' podocytopathy characterized clinically by the presence of nephrotic syndrome in a patient with an FSGS lesion on light microscopy and widespread foot process effacement on electron microscopy (EM). Secondary FSGS is commonly characterized by the absence of nephrotic syndrome and the presence of segmental foot process effacement on EM. Failure to accurately differentiate between the primary and secondary forms of FSGS has resulted in many patients undergoing unnecessary immunosuppressive treatment. Here, we review some key points that may assist the practicing nephrologist to distinguish between primary and secondary FSGS.
Collapse
Affiliation(s)
- Sanjeev Sethi
- Division of Anatomic Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | - Fernando C Fervenza
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
50
|
Yamada S, Nakamura J, Asada M, Takase M, Matsusaka T, Iguchi T, Yamada R, Tanaka M, Higashi AY, Okuda T, Asada N, Fukatsu A, Kawachi H, Graf D, Muso E, Kita T, Kimura T, Pastan I, Economides AN, Yanagita M. Twisted gastrulation, a BMP antagonist, exacerbates podocyte injury. PLoS One 2014; 9:e89135. [PMID: 24586548 PMCID: PMC3934867 DOI: 10.1371/journal.pone.0089135] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/20/2014] [Indexed: 12/12/2022] Open
Abstract
Podocyte injury is the first step in the progression of glomerulosclerosis. Previous studies have demonstrated the beneficial effect of bone morphogenetic protein 7 (Bmp7) in podocyte injury and the existence of native Bmp signaling in podocytes. Local activity of Bmp7 is controlled by cell-type specific Bmp antagonists, which inhibit the binding of Bmp7 to its receptors. Here we show that the product of Twisted gastrulation (Twsg1), a Bmp antagonist, is the central negative regulator of Bmp function in podocytes and that Twsg1 null mice are resistant to podocyte injury. Twsg1 was the most abundant Bmp antagonist in murine cultured podocytes. The administration of Bmp induced podocyte differentiation through Smad signaling, whereas the simultaneous administration of Twsg1 antagonized the effect. The administration of Bmp also inhibited podocyte proliferation, whereas simultaneous administration of Twsg1 antagonized the effect. Twsg1 was expressed in the glomerular parietal cells (PECs) and distal nephron of the healthy kidney, and additionally in damaged glomerular cells in a murine model of podocyte injury. Twsg1 null mice exhibited milder hypoalbuminemia and hyperlipidemia, and milder histological changes while maintaining the expression of podocyte markers during podocyte injury model. Taken together, our results show that Twsg1 plays a critical role in the modulation of protective action of Bmp7 on podocytes, and that inhibition of Twsg1 is a promising means of development of novel treatment for podocyte injury.
Collapse
Affiliation(s)
- Sachiko Yamada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Jin Nakamura
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Misako Asada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Masayuki Takase
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Taiji Matsusaka
- Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Taku Iguchi
- TMK Project, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Ryo Yamada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Mari Tanaka
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Atsuko Y. Higashi
- Deaprtment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Tomohiko Okuda
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Nariaki Asada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | | | - Hiroshi Kawachi
- Department of Cell Biology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Daniel Graf
- Institute of Oral Biology, Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Eri Muso
- Department of Nephrology and Dialysis, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Osaka, Japan
| | - Toru Kita
- Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Takeshi Kimura
- Deaprtment of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aris N. Economides
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- * E-mail:
| |
Collapse
|