1
|
Liu D, Li J, Xu C, Li Y, Chen X, Zhao F, Tong H, Yang Y, Qiu X, Yu Z. Loss of Nup160 dysregulates Cdc42 in the podocytes of podocyte-specific Nup160 knockout mice. Hum Mol Genet 2025:ddaf064. [PMID: 40298220 DOI: 10.1093/hmg/ddaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/24/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Mutations in four genes encoding the outer ring complex of nuclear pore complexes (NPCs), NUP85, NUP107, NUP133 and NUP160, cause monogenic steroid-resistant nephrotic syndrome (SRNS). Knockout of NUP85, NUP107, or NUP133 in immortalized human podocytes activates CDC42, an important effector of SRNS pathogenesis. However, it is unknown whether or not loss of NUP160 dysregulates CDC42 in the podocytes. Here, we generated a podocyte-specific Nup160 knockout mouse model with double-fluorescent (mT/mG) Cre reporter genes using CRISPR/Cas9 and Cre/loxP technologies. We investigated nephrotic syndrome-associated phenotypes in the Nup160podo-/- mice, and performed single-cell transcriptomic and proteomic analysis of glomerular suspension cells and cultured primary podocytes, respectively. The Nup160podo-/- mice exhibited progressive proteinuria and fusion of podocyte foot processes. We found decreased Cdc42 protein and normal Cdc42 transcriptional level in the podocytes of the Nup160podo-/- mice using analysis of single-cell transcriptomes and proteomes. We subsequently observed that Cdc42 protein decreased in both kidney tissues and cultured primary podocytes of the Nup160podo-/- mice, although Cdc42 mRNA levels were elevated in the cultured primary podocytes of the Nup160podo-/- mice. We also found that Cdc42 activity was significantly reduced in the cultured primary podocytes of the Nup160podo-/- mice. In conclusion, loss of Nup160 dysregulated Cdc42 in the podocytes of the Nup160podo-/- mice with proteinuria and fusion of podocyte foot processes. Our findings suggest that the dysregulation of CDC42 may contribute to the pathogenesis of SRNS in patients with mutations in NUP160.
Collapse
Affiliation(s)
- Deying Liu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| | - Jiaxin Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| | - Chan Xu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| | - Yuanyuan Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| | - Xiaohan Chen
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| | - Feng Zhao
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
| | - Huajuan Tong
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
| | - Yonghui Yang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
| | - Xiaojian Qiu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
| | - Zihua Yu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian 350000, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Department of Nephrology, Rheumatology and Immunology, Fujian Children's Hospital, 966 Heng Yu Road, Fuzhou, Fujian 350014, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, 19 Jin Ji Shan Road, Fuzhou, Fujian 350014, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, 18 Daoshan Road, Fuzhou, Fujian 350000, China
| |
Collapse
|
2
|
Waller AP, Wolfgang KJ, Pruner I, Stevenson ZS, Abdelghani E, Muralidharan K, Wilkie TK, Blissett AR, Calomeni EP, Vetter TA, Brodsky SV, Smoyer WE, Nieman MT, Kerlin BA. Effects of Prothrombin on Podocytopathy and Proteinuria in Glomerular Disease. J Am Soc Nephrol 2025:00001751-990000000-00601. [PMID: 40152945 DOI: 10.1681/asn.0000000676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 03/25/2025] [Indexed: 03/30/2025] Open
Abstract
Key Points
Thrombin injures podocytes through its cognate receptors in vitro, and thrombin generation increases during proteinuric glomerular disease.In this study, plasma prothrombin levels modulated in vivo podocyte health and function in a rat model of glomerular disease.Thrombin antagonism may simultaneously prevent thrombosis and CKD progression due to glomerular disease.
Background
CKD is a leading cause of death; its progression is driven by glomerular podocyte injury and loss, manifesting as proteinuria. Proteinuria includes loss of coagulation zymogens, cofactors, and inhibitors resulting in a hypercoagulable state characterized by enhanced thrombin generation. Both CKD and proteinuria significantly increase the risk of thromboembolic disease. Meanwhile, anticoagulant medications (which antagonize thrombin and thereby prevent thromboembolism) have been shown to reduce proteinuria in rats, and thrombin has been shown to injure cultured human and rat podocytes. We thus aimed to directly determine the influence of circulating prothrombin, the zymogen precursor of thrombin, on glomerular pathobiology. We hypothesized that (pro)thrombin drives podocytopathy, podocytopenia, and proteinuria.
Methods
Glomerular proteinuria was induced with puromycin aminonucleoside in rats. Prothrombin was either knocked down using an antisense oligonucleotide–targeting prothrombin mRNA or elevated by serial intravenous prothrombin protein infusions, previously established methods to model hypoprothrombinemia and hyperprothrombinemia, respectively. After 10 days, plasma prothrombin levels were determined and kidneys were examined for (pro)thrombin colocalization to podocytes, histology, and electron microscopy. Podocytopathy, podocytopenia, proteinuria, and plasma albumin were measured.
Results
Antisense oligonucleotide–mediated prothrombin knockdown significantly reduced prothrombin colocalization to podocytes, tubular injury, podocyte foot process effacement, podocytopathy, and proteinuria, along with improved plasma albumin in the puromycin aminonucleoside glomerular disease model. By contrast, elevated prothrombin levels significantly increased podocytopathy and proteinuria. Podocytopenia was significantly improved in hypoprothrombinemic versus hyperprothrombinemic rats.
Conclusions
Thrombin generation is enhanced by glomerular proteinuria, and thrombin injures conditionally immortalized podocytes in vitro. In this study, prothrombin knockdown ameliorated in vivo podocyte injury and improved podocyte function in the rat puromycin aminonucleoside–induced glomerular disease model, whereas hyperprothrombinemia exacerbated podocyte injury and diminished podocyte function.
Collapse
Affiliation(s)
- Amanda P Waller
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Katelyn J Wolfgang
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Iva Pruner
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Zachary S Stevenson
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Eman Abdelghani
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Kaushik Muralidharan
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Tasha K Wilkie
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Angela R Blissett
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Edward P Calomeni
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Tatyana A Vetter
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio
| | - Sergey V Brodsky
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - William E Smoyer
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Bryce A Kerlin
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
3
|
Waller AP, Wolfgang KJ, Pruner I, Stevenson ZS, Abdelghani E, Muralidharan K, Wilkie TK, Blissett AR, Calomeni EP, Vetter TA, Brodsky SV, Smoyer WE, Nieman MT, Kerlin BA. Prothrombin Knockdown Protects Podocytes and Reduces Proteinuria in Glomerular Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.20.544360. [PMID: 38464017 PMCID: PMC10925217 DOI: 10.1101/2023.06.20.544360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Chronic kidney disease (CKD) is a leading cause of death, and its progression is driven by glomerular podocyte injury and loss, manifesting as proteinuria. Proteinuria includes urinary loss of coagulation zymogens, cofactors, and inhibitors. Importantly, both CKD and proteinuria significantly increase the risk of thromboembolic disease. Prior studies demonstrated that anticoagulants reduced proteinuria in rats and that thrombin injured cultured podocytes. Herein we aimed to directly determine the influence of circulating prothrombin on glomerular pathobiology. We hypothesized that (pro)thrombin drives podocytopathy, podocytopenia, and proteinuria. Glomerular proteinuria was induced with puromycin aminonucleoside (PAN) in Wistar rats. Circulating prothrombin was either knocked down using a rat-specific antisense oligonucleotide or elevated by serial intravenous infusions of prothrombin protein, which are previously established methods to model hypo- (LoPT) and hyper-prothrombinemia (HiPT), respectively. After 10 days (peak proteinuria in this model) plasma prothrombin levels were determined, kidneys were examined for (pro)thrombin co-localization to podocytes, histology, and electron microscopy. Podocytopathy and podocytopenia were determined and proteinuria, and plasma albumin were measured. LoPT significantly reduced prothrombin colocalization to podocytes, podocytopathy, and proteinuria with improved plasma albumin. In contrast, HiPT significantly increased podocytopathy and proteinuria. Podocytopenia was significantly reduced in LoPT vs. HiPT rats. In summary, prothrombin knockdown ameliorated PAN-induced glomerular disease whereas hyper-prothrombinemia exacerbated disease. Thus, (pro)thrombin antagonism may be a viable strategy to simultaneously provide thromboprophylaxis and prevent podocytopathy-mediated CKD progression.
Collapse
|
4
|
Song Z, Jin M, Wang S, Wu Y, Huang Q, Xu W, Fan Y, Tian F. Reciprocal regulation of SIRT1 and AMPK by Ginsenoside compound K impedes the conversion from plasma cells to mitigate for podocyte injury in MRL/ lpr mice in a B cell-specific manner. J Ginseng Res 2024; 48:190-201. [PMID: 38465215 PMCID: PMC10920007 DOI: 10.1016/j.jgr.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 03/12/2024] Open
Abstract
Background Deposition of immune complexes drives podocyte injury acting in the initial phase of lupus nephritis (LN), a process mediated by B cell involvement. Accordingly, targeting B cell subsets represents a potential therapeutic approach for LN. Ginsenoside compound K (CK), a bioavailable component of ginseng, possesses nephritis benefits in lupus-prone mice; however, the underlying mechanisms involving B cell subpopulations remain elusive. Methods Female MRL/lpr mice were administered CK (40 mg/kg) intragastrically for 10 weeks, followed by measurements of anti-dsDNA antibodies, inflammatory chemokines, and metabolite profiles on renal samples. Podocyte function and ultrastructure were detected. Publicly available single-cell RNA sequencing data and flow cytometry analysis were employed to investigate B cell subpopulations. Metabolomics analysis was adopted. SIRT1 and AMPK expression were analyzed by immunoblotting and immunofluorescence assays. Results CK reduced proteinuria and protected podocyte ultrastructure in MRL/lpr mice by suppressing circulating anti-dsDNA antibodies and mitigating systemic inflammation. It activated B cell-specific SIRT1 and AMPK with Rhamnose accumulation, hindering the conversion of renal B cells into plasma cells. This cascade facilitated the resolution of local renal inflammation. CK facilitated the clearance of deposited immune complexes, thus reinstating podocyte morphology and mobility by normalizing the expression of nephrin and SYNPO. Conclusions Our study reveals the synergistic interplay between SIRT1 and AMPK, orchestrating the restoration of renal B cell subsets. This process effectively mitigates immune complex deposition and preserves podocyte function. Accordingly, CK emerges as a promising therapeutic agent, potentially alleviating the hyperactivity of renal B cell subsets during LN.
Collapse
Affiliation(s)
- Ziyu Song
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng Jin
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shenglong Wang
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanzuo Wu
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qi Huang
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wangda Xu
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongsheng Fan
- College of Basic Medical Science, Institute of Basic Research in Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengyuan Tian
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Bhayana S, Dougherty JA, Kamigaki Y, Agrawal S, Wijeratne S, Fitch J, Waller AP, Wolfgang KJ, White P, Kerlin BA, Smoyer WE. Glucocorticoid- and pioglitazone-induced proteinuria reduction in experimental NS both correlate with glomerular ECM modulation. iScience 2024; 27:108631. [PMID: 38188512 PMCID: PMC10770536 DOI: 10.1016/j.isci.2023.108631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/31/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Idiopathic nephrotic syndrome (NS) is a common glomerular disease. Although glucocorticoids (GC) are the primary treatment, the PPARγ agonist pioglitazone (Pio) also reduces proteinuria in patients with NS and directly protects podocytes from injury. Because both drugs reduce proteinuria, we hypothesized these effects result from overlapping transcriptional patterns. Systems biology approaches compared glomerular transcriptomes from rats with PAN-induced NS treated with GC vs. Pio and identified 29 commonly regulated genes-of-interest, primarily involved in extracellular matrix (ECM) remodeling. Correlation with clinical idiopathic NS patient datasets confirmed glomerular ECM dysregulation as a potential mechanism of injury. Cellular deconvolution in silico revealed GC- and Pio-induced amelioration of altered genes primarily within podocytes and mesangial cells. While validation studies are indicated, these analyses identified molecular pathways involved in the early stages of NS (prior to scarring), suggesting that targeting glomerular ECM dysregulation may enable a future non-immunosuppressive approach for proteinuria reduction in idiopathic NS.
Collapse
Affiliation(s)
- Sagar Bhayana
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Julie A. Dougherty
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Yu Kamigaki
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Shipra Agrawal
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Saranga Wijeratne
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - James Fitch
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Amanda P. Waller
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Katelyn J. Wolfgang
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Peter White
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Bryce A. Kerlin
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - William E. Smoyer
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Muso E, Kakita H, Suzuki H, Tsukamoto T. Updated evidence of beneficial effect of LDL apheresis for refractory nephrotic syndrome due to a variety of causative diseases for nationwide and global approval. Ther Apher Dial 2023; 27:987-999. [PMID: 37593995 DOI: 10.1111/1744-9987.14056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/23/2023] [Indexed: 08/19/2023]
Abstract
Low-density lipoprotein apheresis (LDL-A) therapy has shown reasonable efficacy in treating nephrotic syndrome (NS) refractory to initial drug therapy and has been covered by National Health Insurance for the indication of drug-resistant focal segmental glomerulosclerosis (FSGS) since 1992 in Japan and has contributed to liberating substantial number of patients of this disease from entering into end-stage renal disease by easier practical application in actual clinical settings. Subsequently, various beneficial evidence of this treatment has accumulated on those other than FSGS, however, due to the limitation of covered disease insurance only for FSGS, patients with diseases other than FSGS are unlikely to benefit from this treatment in practice. This review summarizes the therapeutic evidence of the beneficial effect of LDL-A accumulated to date and the mechanisms of action analyzed from multifaceted perspectives. examines the applicability of expanding insurance coverage for diseases other than FSGS.
Collapse
Affiliation(s)
- Eri Muso
- Department of Food and Nutrition, Faculty of Contemporary Home Economics, Kyoto Kacho University, Kyoto, Japan
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Hiroko Kakita
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Hiroyuki Suzuki
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Tatsuo Tsukamoto
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| |
Collapse
|
7
|
Haeri H, Eisermann J, Schimm H, Büscher A, Hoyer P, Hinderberger D. Profound Changes in Functional Structure and Dynamics of Serum Albumin in Children with Nephrotic Syndrome: An Exploratory Research Study. J Med Chem 2023; 66:12115-12129. [PMID: 37648246 PMCID: PMC10510392 DOI: 10.1021/acs.jmedchem.3c00680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Indexed: 09/01/2023]
Abstract
Patients with nephrotic syndrome (NS) suffer from urinary loss of albumin. As a cause, previous studies focused on the glomerular filter rather than analyzing the molecular properties of albumin itself. Later one was initiated by clinical observations indicating unexplained molecular alterations of human serum albumin (HSA) in an NS pediatric patient. Therefore, we examined serum from eight pediatric patients with steroid-sensitive and -resistant NS and compared it with serum from healthy subjects as well as commercial HSA. We used dynamic and electrophoretic light scattering to characterize the protein size and effective surface charge and electron paramagnetic resonance spectroscopy to measure the local environment and binding dynamics of up to seven fatty acids associated with HSA. Our findings suggest that pronounced differences in binding behavior and surface charge of HSA could enhance their filtration through the GBM, leading to direct toxicity of HSA to podocytes.
Collapse
Affiliation(s)
- Haleh
H. Haeri
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Jana Eisermann
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Ln, W12 0BZ London, U.K.
| | - Heike Schimm
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Anja Büscher
- Universitätsklinikum
Essen (AöR), Klinik für Kinderheilkunde II, Zentrum
für Kinder- und Jugendmedizin, Hufelandstraße 55, D-45147 Essen, Germany
| | - Peter Hoyer
- Universitätsklinikum
Essen (AöR), Klinik für Kinderheilkunde II, Zentrum
für Kinder- und Jugendmedizin, Hufelandstraße 55, D-45147 Essen, Germany
| | - Dariush Hinderberger
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| |
Collapse
|
8
|
Ma X, Zhang X, Leng T, Ma J, Yuan Z, Gu Y, Hu T, Liu Q, Shen T. Identification of Oxidative Stress-Related Biomarkers in Diabetic Kidney Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:1067504. [PMID: 36624863 PMCID: PMC9825216 DOI: 10.1155/2022/1067504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease throughout the world. In kidney disease, oxidative stress has been linked to both antioxidant depletions and increased reactive oxygen species (ROS) production. Thus, the objective of this study was to identify biomarkers related to oxidative stress in DKD. METHODS The gene expression profile of the DKD was extracted from the Gene Expression Omnibus (GEO) database. The identification of the differentially expressed genes (DEGs) was performed using the "limma" R package, and weighted gene coexpression network analysis (WGCNA) was used to find the gene modules that were most related to DKD. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed using "Org.Hs.eg.db" R package. The protein-protein interaction (PPI) network was constructed using the STRING database. The hub genes were identified by the Molecular Complex Detection (MCODE) plug-in of Cytoscape software. The diagnostic capacity of hub genes was verified using the receiver operating characteristic (ROC) curve. Correlations between diagnostic genes were analyzed using the "corrplot" package. In addition, the miRNA gene transcription factor (TF) network was used to explain the regulatory mechanism of hub genes in DKD. RESULTS DEGs analysis and WGCNA-identified 160 key genes were identified in DKD patients. Among them, nine oxidative stress-related genes were identified as candidate hub genes for DKD. Using the PPI network, five hub genes, NR4A2, DUSP1, FOS, JUN, and PTGS2, were subsequently identified. All the hub genes were downregulated in DKD and had a high diagnostic value of DKD. The regulatory mechanism of hub genes was analyzed from the miRNA gene-TF network. CONCLUSION Our study identified NR4A2, DUSP1, FOS, JUN, and PTGS2 as hub genes of DKD. These genes may serve as potential therapeutic targets for DKD patients.
Collapse
Affiliation(s)
- Xiaoju Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaobo Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tian Leng
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingru Ma
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhongzhu Yuan
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yalin Gu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingting Hu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiuyan Liu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
9
|
Bai M, Zhang J, Su X, Yao X, Li H, Cheng J, Mao J, Li X, Chen J, Lin W. Serum IL-12p40: A novel biomarker for early prediction of minimal change disease relapse following glucocorticoids therapy. Front Med (Lausanne) 2022; 9:922193. [PMID: 36507530 PMCID: PMC9729255 DOI: 10.3389/fmed.2022.922193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
Background Minimal change disease (MCD) has a high recurrence rate, but currently, no biomarker can predict its recurrence. To this end, this study aimed at identifying potential serum cytokines as valuable biomarkers for predicting the risk of MCD recurrence. Materials and methods Raybiotech 440 cytokine antibody microarray was used to detect the serum samples of eight relapsed, eight non-relapsed MCD patients after glucocorticoid treatment, and eight healthy controls. The differentially expressed cytokines were confirmed by enzyme-linked immunosorbent assay (ELISA) with serum samples from 29 non-relapsed and 35 relapsed MCD patients. The study used the receiver operating characteristic (ROC) curve analysis to investigate the sensitivity and specificity of a serum biomarker for predicting the MCD relapse. Results Serum IL-12p40 levels increased significantly in the relapsed group. The Area Under the ROC Curve (AUC) of IL-12p40 was 0.727 (95%CI: 0.597-0.856; P < 0.01). The RNA-sequencing analysis and qPCR assay performed on the IL-12 treated mouse podocytes and the control group showed increased expression of podocyte damage genes, such as connective tissue growth factor (CTGF), matrix metallopeptidase 9 (MMP9), secreted phosphoprotein 1 (SPP1), and cyclooxygenase-2 (COX-2) in the former group. Conclusion IL-12p40 may serve as a new biomarker for predicting the risk of MCD recurrence after glucocorticoid treatment, and it may be involved in the pathogenesis and recurrence of MCD.
Collapse
Affiliation(s)
- Mengqiu Bai
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Department of Nephrology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Jinhua, Zhejiang, China,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Zhang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinwan Su
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Department of Nephrology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Xi Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Li
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Cheng
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiayu Li
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Xiayu Li,
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Jianghua Chen,
| | - Weiqiang Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Department of Nephrology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Jinhua, Zhejiang, China,*Correspondence: Weiqiang Lin,
| |
Collapse
|
10
|
Bryant C, Webb A, Banks AS, Chandler D, Govindarajan R, Agrawal S. Alternatively Spliced Landscape of PPARγ mRNA in Podocytes Is Distinct from Adipose Tissue. Cells 2022; 11:cells11213455. [PMID: 36359851 PMCID: PMC9653906 DOI: 10.3390/cells11213455] [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: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Podocytes are highly differentiated epithelial cells, and their structural and functional integrity is compromised in a majority of glomerular and renal diseases, leading to proteinuria, chronic kidney disease, and kidney failure. Traditional agonists (e.g., pioglitazone) and selective modulators (e.g., GQ-16) of peroxisome-proliferator-activated-receptor-γ (PPARγ) reduce proteinuria in animal models of glomerular disease and protect podocytes from injury via PPARγ activation. This indicates a pivotal role for PPARγ in maintaining glomerular function through preservation of podocytes distinct from its well-understood role in driving insulin sensitivity and adipogenesis. While its transcriptional role in activating adipokines and adipogenic genes is well-established in adipose tissue, liver and muscle, understanding of podocyte PPARγ signaling remains limited. We performed a comprehensive analysis of PPARγ mRNA variants due to alternative splicing, in human podocytes and compared with adipose tissue. We found that podocytes express the ubiquitous PPARγ Var 1 (encoding γ1) and not Var2 (encoding γ2), which is mostly restricted to adipose tissue and liver. Additionally, we detected expression at very low level of Var4, and barely detectable levels of other variants, Var3, Var11, VartORF4 and Var9, in podocytes. Furthermore, a distinct podocyte vs. adipocyte PPAR-promoter-response-element containing gene expression, enrichment and pathway signature was observed, suggesting differential regulation by podocyte specific PPARγ1 variant, distinct from the adipocyte-specific γ2 variant. In summary, podocytes and glomeruli express several PPARγ variants, including Var1 (γ1) and excluding adipocyte-specific Var2 (γ2), which may have implications in podocyte specific signaling and pathophysiology. This suggests that that new selective PPARγ modulators can be potentially developed that will be able to distinguish between the two forms, γ1 and γ2, thus forming a basis of novel targeted therapeutic avenues.
Collapse
Affiliation(s)
- Claire Bryant
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Amy Webb
- Department of Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Alexander S. Banks
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dawn Chandler
- Center for Childhood Cancer and Blood Disease, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Shipra Agrawal
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Nephrology and Hypertension, Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
- Correspondence:
| |
Collapse
|
11
|
Kim EY, Dryer SE. TRPC6 Inactivation Reduces Albuminuria Induced by Protein Overload in Sprague Dawley Rats. Cells 2022; 11:1985. [PMID: 35805070 PMCID: PMC9265922 DOI: 10.3390/cells11131985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 12/02/2022] Open
Abstract
Canonical transient receptor potential-6 (TRPC6) channels have been implicated in familial and acquired forms of focal and segmental glomerulosclerosis (FSGS), and in renal fibrosis following ureteral obstruction in mice. TRPC6 channels also appear to play a role in driving glomerular disease in aging and in autoimmune glomerulonephritis. In the present study, we examine the role of TRPC6 in the proteinuric state caused by prolonged albumin overload (AO) in Sprague Dawley rats induced by daily injections of exogenous albumin. This was assessed in rats with a global and constitutive inactivation of TRPC6 channels (Trpc6del/del rats) and in wild-type littermates (Trpc6wt/wt rats). AO for 14 and 28 days caused increased urine albumin excretion that was significantly attenuated in Trpc6del/del rats compared to Trpc6wt/wt controls. AO overload did not induce significant glomerulosclerosis or azotemia in either genotype. AO induced mild tubulointerstitial disease characterized by fibrosis, hypercellularity and increased expression of markers of fibrosis and inflammation. Those changes were equally severe in Trpc6wt/wt and Trpc6del/del rats. Immunoblot analysis of renal cortex indicated that AO increased the abundances of TRPC3 and TRPC6, and caused a nearly complete loss of TRPC5 in Trpc6wt/wt rats. The increase in TRPC3 and the loss of TRPC5 occurred to the same extent in Trpc6del/del rats. These data also suggest that TRPC6 plays a role in the normal function of the glomerular filtration barrier. However, whether TRPC6 inactivation protects the tubulointerstitial compartments in Sprague Dawley rats depends on the disease model examined.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
| | - Stuart E. Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
12
|
Samynathan R, Subramanian U, Venkidasamy B, Shariati MA, Chung IM, Thiruvengadam M. S-Allylcysteine (SAC) Exerts Renoprotective Effects via Regulation of TGF-
β1/Smad3 Pathway Mediated Matrix Remodeling in Chronic Renal Failure. Curr Pharm Des 2022; 28:661-670. [DOI: 10.2174/1381612828666220401114301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/03/2021] [Accepted: 04/08/2021] [Indexed: 11/22/2022]
Abstract
Background: S-Allylcysteine (SAC), an organosulfur phytochemical sourced from aged garlic extract,
is well known for its varied biomedical applications, such as anti-oxidant, anti-inflammatory, and detoxification
mechanisms. Despite this, the scientific findings on the defensive impact of SAC against kidney failure
(KF) are still unclear. Therefore, in the current investigation, the animal model of KF was induced by adenine
in Wistar rats, and the animals were divided into four groups as control, KF induction using adenine, SAC treated
KF rats for an experimental duration of 8 weeks.
Methods: KF progression was assessed by various serum and tissue markers, and the results demonstrated that
the renal functions’ markers, KIM-1 (kidney injury molecule-1), cystatin, NGAL (neutrophil gelatinase-associated
lipocalin), were found increased in adenine-treated rats compared to control. In addition, the inflammatory
markers, matrix proteins, and fibrosis signatures explicated by RT-PCR, ELISA demonstrated a profound increase.
On the other hand, rats received SAC mitigated KF considerably (p < 0.001) with restored cellular functions.
Besides, SAC pre-treatment abrogated the cytokines and pro-inflammatory signals (COX-2 and PGE2) in
a dose-dependent manner.
Conclusion: Furthermore, the fibrosis signaling markers mediators, such as SMAD-2,-3 were increased with associated
matrix proteins. Thus, the present study substantiated that SAC possesses a significant renoprotective
effect that might have been demonstrated by the inhibition of the TGF-β1/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Ramkumar Samynathan
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India
| | - Umadevi Subramanian
- Translational Research Platform for Veterinary Biologicals, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk
Colony, Chennai 600051, Tamil Nadu, India
| | - Baskar Venkidasamy
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India
| | - Mohammad Ali Shariati
- Department of Technology of Food Products, K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), 73, Zemlyanoy Val St., Moscow, 109004, Russian Federation
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
13
|
Tagaya M, Kume S, Yasuda-Yamahara M, Kuwagata S, Yamahara K, Takeda N, Tanaka Y, Chin-Kanasaki M, Nakae Y, Yokoi H, Mukoyama M, Ishihara N, Nomura M, Araki SI, Maegawa H. Inhibition of mitochondrial fission protects podocytes from albumin-induced cell damage in diabetic kidney disease. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166368. [DOI: 10.1016/j.bbadis.2022.166368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
|
14
|
Mondragón-Huerta CG, Bautista-Pérez R, Baiza-Gutman LA, Escobar-Sánchez ML, Valle-Mondragón LD, Salas-Garrido CG, Castro-Moreno P, Ibarra-Barajas M. Morphology and cyclooxygenase-2 and renin expression in the kidney of young spontaneously hypertensive rats. Vet Pathol 2021; 59:371-384. [PMID: 34841988 DOI: 10.1177/03009858211052663] [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
The kidneys play an important role in blood pressure regulation under normal and pathological conditions. We examined the histological changes and expression patterns of cyclooxygenase-2, renin, and (pro)renin receptor (PRR) in the renal cortex of prehypertensive spontaneously hypertensive rats (SHRs) and Wistar Kyoto rats (WKYs). Moreover, blood pressure and plasma urea, creatinine, angiotensin II, and angiotensin (1-7) levels were measured. The results showed that both strains had similar blood pressure and plasma urea and creatinine levels. The glomerular area, basement membrane thickness, collagen fiber content, and arterial wall thickness were greater in SHRs than in WKYs. By immunohistochemistry, cyclooxygenase-2 was localized in the macula densa and renal tubules of both strains. In SHRs, cyclooxygenase-2 was detected in a larger number of tubules, and the cortical expression of cyclooxygenase-2 was also increased. In both strains, PRR and renin were localized in the tubular epithelium and juxtaglomerular cells, respectively. In SHRs, PRR immunolocalization was increased in the glomerulus. The cortical expression of immature renin was markedly increased in SHRs compared to that in WKYs, while renin was significantly decreased. These changes were associated with higher plasma angiotensin II levels and lower plasma angiotensin (1-7) levels in SHRs. The results indicate that the kidneys of SHRs showed morphological changes and variations in cortical expression patterns of PRR, cyclooxygenase-2, and renin before the development of hypertension.
Collapse
Affiliation(s)
| | - Rocío Bautista-Pérez
- Instituto Nacional de Cardiología, "Dr. Ignacio Chávez," Ciudad de México, México
| | - Luis A Baiza-Gutman
- Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | | | | | | | | | | |
Collapse
|
15
|
Pro-and anti-inflammatory effects of glucocorticoid Fluticasone on ovarian and immune functions in commercial-aged laying hens. Sci Rep 2021; 11:21603. [PMID: 34732778 PMCID: PMC8566555 DOI: 10.1038/s41598-021-01141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/25/2021] [Indexed: 11/09/2022] Open
Abstract
Ovarian chronic inflammation has been created and extended in the laying hen mainly via increasing laying frequency and microbial infection, especially during the late stage of production period. This study was aimed to evaluate glucocorticoid (GC) Fluticasone as an anti-inflammatory agent on the gene expression of the ovarian pro-and anti-inflammatory mediators (follicular cyclooxygenases COX 1, 2, and cytokines), inflammatory responses of the immune system, ovarian functions (ovulation rate and follicular growths), and hormones in the commercial-aged laying hens. White Leghorn hens aged 92-weeks were used for four weeks to be supplemented by 2 ppm Fluticasone as an optimum dose obtained in a pre-trial base on ovulation rate. As compared to control, Fluticasone resulted in a significant decrease in the mRNA expression of COX-1 and pro-and anti-inflammatory cytokines, and increase in COX-2 mRNA expression and heterophil to lymphocyte ratio (P < 0.001). A significant reduction was observed in the ovulation rate, follicular size (P < 0.001), ovarian hormones, immunoglobulins, body weight, and food consummation (P ≤ 0.05) by administering GC Fluticasone. Although a relative anti-inflammatory improvement was created by Fluticasone in the ovarian condition, the administration of this glucocorticoid resulted in a considerable reduction in ovarian hormones and functions of commercial aged laying hens.
Collapse
|
16
|
van den Broek M, Smeets B, Schreuder MF, Jansen J. The podocyte as a direct target of glucocorticoids in nephrotic syndrome. Nephrol Dial Transplant 2021; 37:1808-1815. [PMID: 33515261 DOI: 10.1093/ndt/gfab016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
Nephrotic syndrome (NS) is characterized by massive proteinuria; podocyte loss or altered function is a central event in its pathophysiology. Treatment with glucocorticoids is the mainstay of therapy. However, many patients experience one or multiple relapses and prolonged use may be associated with severe adverse effects. Recently, the beneficial effects of glucocorticoids have been attributed to a direct effect on podocytes in addition to the well-known immunosuppressive effects. The molecular effects of glucocorticoid action have been studied using animal and cell models of NS. This review provides a comprehensive overview of different molecular mediators regulated by glucocorticoids including an overview of the model systems that were used to study them. Glucocorticoids are described to stimulate podocyte recovery by restoring pro-survival signaling of slit diaphragm related proteins and limiting inflammatory responses. Of special interest is the effect of glucocorticoids on stabilizing the cytoskeleton of podocytes, since these effects are also described for other therapeutic agents used in NS, such as cyclosporin. Current models provide much insight, but do not fully recapitulate the human condition since the pathophysiology underlying NS is poorly understood. New and promising models include the glomerulus-on-a-chip and kidney organoids, which have the potential to be further developed into functional NS models in the future.
Collapse
Affiliation(s)
- Martijn van den Broek
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands.,Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Bart Smeets
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Jitske Jansen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands.,Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Amalia Children's Hospital, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Zuo J, Wang SM, Jiang X, Cao M, Zhang Z, Shi T, Qin HL, Tang W. Design, synthesis and biological evaluation of novel arylpropionic esters for the treatment of acute kidney injury. Bioorg Chem 2020; 105:104455. [PMID: 33197847 DOI: 10.1016/j.bioorg.2020.104455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 12/27/2022]
Abstract
Acute kidney injury (AKI) is associated with a strong inflammatory response, and inhibiting the response effectively prevents or ameliorates AKI. A series of novel arylpropionic esters were designed, synthesized and evaluated their biological activity in LPS-stimulated RAW264.7 cells. Novel arylpropionic esters bearing multi-functional groups showed significant anti-inflammatory activity, in which, compound 13b exhibited the most potent activity through dose-dependent inhibiting the production of nitric oxide (NO, IC50 = 3.52 μM), TNF-α and IL-6 (84.1% and 33.6%, respectively), as well as suppressing the expression of iNOS, COX-2 and TLR4 proteins. In C57BL/6 mice with cisplatin-induced AKI, compound 13b improved kidney function, inhibited inflammatory development, and reduced pathological damage of kidney tissues. In brief, this arylpropionic ester scaffold may be developed as anti-inflammatory agents.
Collapse
Affiliation(s)
- Jiawei Zuo
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shi-Meng Wang
- School of Life Science, Wuchang University of Technology, Wuhan 430223, China; School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Xia Jiang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Mengxin Cao
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Ziwen Zhang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Tianlu Shi
- First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Hua-Li Qin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Wenjian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
18
|
Abstract
Physical trauma can affect any individual and is globally accountable for more than one in every ten deaths. Although direct severe kidney trauma is relatively infrequent, extrarenal tissue trauma frequently results in the development of acute kidney injury (AKI). Various causes, including haemorrhagic shock, rhabdomyolysis, use of nephrotoxic drugs and infectious complications, can trigger and exacerbate trauma-related AKI (TRAKI), particularly in the presence of pre-existing or trauma-specific risk factors. Injured, hypoxic and ischaemic tissues expose the organism to damage-associated and pathogen-associated molecular patterns, and oxidative stress, all of which initiate a complex immunopathophysiological response that results in macrocirculatory and microcirculatory disturbances in the kidney, and functional impairment. The simultaneous activation of components of innate immunity, including leukocytes, coagulation factors and complement proteins, drives kidney inflammation, glomerular and tubular damage, and breakdown of the blood-urine barrier. This immune response is also an integral part of the intense post-trauma crosstalk between the kidneys, the nervous system and other organs, which aggravates multi-organ dysfunction. Necessary lifesaving procedures used in trauma management might have ambivalent effects as they stabilize injured tissue and organs while simultaneously exacerbating kidney injury. Consequently, only a small number of pathophysiological and immunomodulatory therapeutic targets for TRAKI prevention have been proposed and evaluated.
Collapse
|
19
|
Abstract
Nuclear receptors have a broad spectrum of biological functions in normal physiology and in the pathology of various diseases, including glomerular disease. The primary therapies for many glomerular diseases are glucocorticoids, which exert their immunosuppressive and direct podocyte protective effects via the glucocorticoid receptor (GR). As glucocorticoids are associated with important adverse effects and a substantial proportion of patients show resistance to these therapies, the beneficial effects of selective GR modulators are now being explored. Peroxisome proliferator-activated receptor-γ (PPARγ) agonism using thiazolidinediones has potent podocyte cytoprotective and nephroprotective effects. Repurposing of thiazolidinediones or identification of novel PPARγ modulators are potential strategies to treat non-diabetic glomerular disease. Retinoic acid receptor-α is the key mediator of the renal protective effects of retinoic acid, and repair of the endogenous retinoic acid pathway offers another potential therapeutic strategy for glomerular disease. Vitamin D receptor, oestrogen receptor and mineralocorticoid receptor modulators regulate podocyte injury in experimental models. Further studies are needed to better understand the mechanisms of these nuclear receptors, evaluate their synergistic pathways and identify their novel modulators. Here, we focus on the role of nuclear receptors in podocyte biology and non-diabetic glomerular disease.
Collapse
|
20
|
Identifying Synergistic Mechanisms of Multiple Ingredients in Shuangbai Tablets against Proteinuria by Virtual Screening and a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1027271. [PMID: 32025234 PMCID: PMC6984745 DOI: 10.1155/2020/1027271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/08/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
Shuangbai Tablets (SBT), a traditional herbal mixture, has shown substantial clinical efficacy. However, a systematic mechanism of its active ingredients and pharmacological mechanisms of action against proteinuria continues being lacking. A network pharmacology approach was effectual in discovering the relationship of multiple ingredients and targets of the herbal mixture. This study aimed to identify key targets, major active ingredients, and pathways of SBT against proteinuria by network pharmacology approach combined with thin layer chromatography (TLC). Human phenotype (HP) disease analysis, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and molecular docking were used in this study. To this end, a total of 48 candidate targets of 118 active ingredients of SBT were identified. Network analysis showed PTGS2, ESR1, and NOS2 to be the three key targets, and beta-sitosterol, quercetin, and berberine were the three major active ingredients; among them one of the major active ingredients, quercetin, was discriminated by TLC. These results of the functional enrichment analysis indicated that the most relevant disease including these 48 candidate proteins is proteinuria, SBT treated proteinuria by sympathetically regulating multiple biological pathways, such as the HIF-1, RAS, AGE-RAGE, and VEGF signaling pathways. Additionally, molecular docking validation suggested that major active ingredients of SBT were capable of binding to HIF-1A and VEGFA of the main pathways. Consequently, key targets, major active ingredients, and pathways based on data analysis of SBT against proteinuria were systematically identified confirming its utility and providing a new drug against proteinuria.
Collapse
|
21
|
Mirabito Colafella KM, Neuman RI, Visser W, Danser AHJ, Versmissen J. Aspirin for the prevention and treatment of pre-eclampsia: A matter of COX-1 and/or COX-2 inhibition? Basic Clin Pharmacol Toxicol 2019; 127:132-141. [PMID: 31420920 PMCID: PMC7496715 DOI: 10.1111/bcpt.13308] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/11/2019] [Indexed: 01/04/2023]
Abstract
Since the 1970s, we have known that aspirin can reduce the risk of pre‐eclampsia. However, the underlying mechanisms explaining this risk reduction are poorly understood. Both cyclooxygenase (COX)‐1‐ and COX‐2‐dependent effects might be involved. As a consequence of this knowledge hiatus, the optimal dose and timing of initiation of aspirin therapy are not clear. Here, we review how (COX‐1 versus COX‐2 inhibition) and when (prevention versus treatment) aspirin therapy may interfere with the mechanisms implicated in the pathogenesis of pre‐eclampsia. The available evidence suggests that both COX‐1‐ and COX‐2‐dependent effects play important roles in the early stage of aberrant placental development and in the next phase leading to the clinical syndrome of pre‐eclampsia. Collectively, these data suggest that high‐dose (dual COX inhibition) aspirin may be superior to standard low‐dose (selective COX‐1 inhibition) aspirin for the prevention and also treatment of pre‐eclampsia. Therefore, we conclude that more functional and biochemical tests are needed to unravel the contribution of prostanoids in the mechanisms implicated in the pathogenesis of pre‐eclampsia and the potential of dual COX and/or selective COX‐2 inhibition for the prevention and treatment of pre‐eclampsia. This information is vital if we are to deduce the suitability, optimal timing and dose of aspirin and/or a specific COX‐2 inhibitor (most likely using modified forms that do not cross the placenta) that can then be tested in a randomized, controlled trial instead of the current practice of empirical dosing regimens.
Collapse
Affiliation(s)
- Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic, Australia.,Department of Physiology, Monash University, Melbourne, Vic, Australia.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rugina I Neuman
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Division of Obstetrics and Perinatal Medicine, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Willy Visser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Division of Obstetrics and Perinatal Medicine, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jorie Versmissen
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Nihalani D, Solanki AK, Arif E, Srivastava P, Rahman B, Zuo X, Dang Y, Fogelgren B, Fermin D, Gillies CE, Sampson MG, Lipschutz JH. Disruption of the exocyst induces podocyte loss and dysfunction. J Biol Chem 2019; 294:10104-10119. [PMID: 31073028 PMCID: PMC6664173 DOI: 10.1074/jbc.ra119.008362] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/06/2019] [Indexed: 11/06/2022] Open
Abstract
Although the slit diaphragm proteins in podocytes are uniquely organized to maintain glomerular filtration assembly and function, little is known about the underlying mechanisms that participate in trafficking these proteins to the correct location for development and homeostasis. Identifying these mechanisms will likely provide novel targets for therapeutic intervention to preserve podocyte function following glomerular injury. Analysis of structural variation in cases of human nephrotic syndrome identified rare heterozygous deletions of EXOC4 in two patients. This suggested that disruption of the highly-conserved eight-protein exocyst trafficking complex could have a role in podocyte dysfunction. Indeed, mRNA profiling of injured podocytes identified significant exocyst down-regulation. To test the hypothesis that the exocyst is centrally involved in podocyte development/function, we generated homozygous podocyte-specific Exoc5 (a central exocyst component that interacts with Exoc4) knockout mice that showed massive proteinuria and died within 4 weeks of birth. Histological and ultrastructural analysis of these mice showed severe glomerular defects with increased fibrosis, proteinaceous casts, effaced podocytes, and loss of the slit diaphragm. Immunofluorescence analysis revealed that Neph1 and Nephrin, major slit diaphragm constituents, were mislocalized and/or lost. mRNA profiling of Exoc5 knockdown podocytes showed that vesicular trafficking was the most affected cellular event. Mapping of signaling pathways and Western blot analysis revealed significant up-regulation of the mitogen-activated protein kinase and transforming growth factor-β pathways in Exoc5 knockdown podocytes and in the glomeruli of podocyte-specific Exoc5 KO mice. Based on these data, we propose that exocyst-based mechanisms regulate Neph1 and Nephrin signaling and trafficking, and thus podocyte development and function.
Collapse
Affiliation(s)
- Deepak Nihalani
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425,
| | - Ashish K Solanki
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Ehtesham Arif
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Pankaj Srivastava
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Bushra Rahman
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Xiaofeng Zuo
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Yujing Dang
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Ben Fogelgren
- the Department of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii 96813
| | | | | | - Matthew G Sampson
- the Department of Pediatrics-Nephrology and.,Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, and
| | - Joshua H Lipschutz
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425.,the Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| |
Collapse
|
23
|
Lipid-lowering agents for the treatment of hyperlipidemia in patients with chronic kidney disease and end-stage renal disease on dialysis: a review. DRUGS & THERAPY PERSPECTIVES 2019. [DOI: 10.1007/s40267-019-00646-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
Ghayur A, Padwal MK, Liu L, Zhang J, Margetts PJ. SMAD3-dependent and -independent pathways in glomerular injury associated with experimental glomerulonephritis. Am J Physiol Renal Physiol 2019; 317:F152-F162. [PMID: 31141397 DOI: 10.1152/ajprenal.00406.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glomerulonephritis (GN) is a common cause of end-stage kidney disease and is characterized by glomerular inflammation, hematuria, proteinuria, and progressive renal dysfunction. Transforming growth factor (TGF)-β is involved in glomerulosclerosis and interstitial fibrosis. TGF-β activates multiple signaling pathways, including the canonical SMAD pathway. We evaluated the role of SMAD signaling in renal injury and proteinuria in a murine model of GN. SMAD3+/+ or SMAD3-/- mice received anti-glomerular basement membrane antibodies to induce GN. We confirmed previous reports that demonstrated that SMAD3 is an important mediator of glomerulosclerosis and renal interstitial fibrosis. Proteinuria was highly SMAD3 dependent. We found differential effects of SMAD3 deletion on podocytes and glomerular endothelial cells. GN led to podocyte injury, including foot process effacement and loss of podocyte-specific markers. Interestingly, these changes were not SMAD3 dependent. Furthermore, there were significant changes to glomerular endothelial cells, including loss of fenestrations, swelling, and basement membrane reduplication, which were SMAD3 dependent. Despite ongoing markers of podocyte injury in SMAD3-/- mice, proteinuria was transient. Renal injury in the setting of GN involves TGF-β and SMAD3 signaling. Cell populations within the glomerulus respond differently to SMAD3 deletion. Proteinuria correlated more with endothelial cell changes as opposed to podocyte injury in this model.
Collapse
Affiliation(s)
- Ayesha Ghayur
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | | | - Limin Liu
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Jing Zhang
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Peter J Margetts
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
25
|
Huang X, Xue H, Ma J, Zhang Y, Zhang J, Liu Y, Qin X, Sun C. Salidroside ameliorates Adriamycin nephropathy in mice by inhibiting β-catenin activity. J Cell Mol Med 2019; 23:4443-4453. [PMID: 30993911 PMCID: PMC6533469 DOI: 10.1111/jcmm.14340] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/21/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Salidroside is a major phenylethanoid glycoside in Rhodiola rosea L., a traditional Chinese medicine, with multiple biological activities. It has been shown that salidroside possesses protective effects for alleviating diabetic renal dysfunction, contrast‐induced‐nephropathy and other kidney diseases. However, the involved molecular mechanism was still not understood well. Herein, we examined the protective effects of salidroside in mice with Adriamycin (ADR)‐induced nephropathy and the underlying molecular mechanism. The results showed that salidroside treatment ameliorates proteinuria; improves expressions of nephrin and podocin; and reduces kidney fibrosis and glomerulosclerosis induced by ADR. Mechanistically, ADR induces a robust accumulation of β‐catenin in the nucleus and stimulates its downstream target gene expression. The application of salidroside largely abolishes the nuclear translocation of β‐catenin and thus inhibits its activity. Furthermore, the activation of β‐catenin almost completely counteracts the protective roles of salidroside in ADR‐injured podocytes. Taken together, our data indicate that salidroside ameliorates proteinuria, renal fibrosis and podocyte injury in ADR nephropathy, which may rely on inhibition of β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Xinzhong Huang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Xue
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| | | | - Jing Zhang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| | - Yue Liu
- Department of Nephrology, Traditional Chinese Medicine Hospital of Tongzhou District, Nantong, China
| | - Xiaogang Qin
- Department of Nephrology, Traditional Chinese Medicine Hospital of Tongzhou District, Nantong, China
| | - Cheng Sun
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| |
Collapse
|
26
|
Korolj A, Laschinger C, James C, Hu E, Velikonja C, Smith N, Gu I, Ahadian S, Willette R, Radisic M, Zhang B. Curvature facilitates podocyte culture in a biomimetic platform. LAB ON A CHIP 2018; 18:3112-3128. [PMID: 30264844 DOI: 10.1039/c8lc00495a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Most kidney diseases begin with abnormalities in glomerular podocytes, motivating the need for podocyte models to study pathophysiological mechanisms and new treatment options. However, podocytes cultured in vitro face a limited ability to maintain appreciable extents of differentiation hallmarks, raising concerns over the relevance of study results. Many key properties such as nephrin expression and morphology reach plateaus that are far from the in vivo levels. Here, we demonstrate that a biomimetic topography, consisting of microhemispheres arrayed over the cell culture substrate, promotes podocyte differentiation in vitro. We define new methods for fabricating microscale curvature on various substrates, including a thin porous membrane. By growing podocytes on our topographic substrates, we found that these biophysical cues augmented nephrin gene expression, supported full-size nephrin protein expression, encouraged structural arrangement of F-actin and nephrin within the cell, and promoted process formation and even interdigitation compared to the flat substrates. Furthermore, the topography facilitated nephrin localization on curved structures while nuclei lay in the valleys between them. The improved differentiation was also evidenced by tracking barrier function to albumin over time using our custom topomembranes. Overall, our work presents accessible methods for incorporating microcurvature on various common substrates, and demonstrates the importance of biophysical stimulation in supporting higher-fidelity podocyte cultivation in vitro.
Collapse
Affiliation(s)
- Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang H, Zheng C, Xu X, Zhao Y, Lu Y, Liu Z. Fibrinogen links podocyte injury with Toll-like receptor 4 and is associated with disease activity in FSGS patients. Nephrology (Carlton) 2018; 23:418-429. [PMID: 28407405 DOI: 10.1111/nep.13046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/24/2017] [Accepted: 03/15/2017] [Indexed: 12/19/2022]
Abstract
AIM Fibrinogen (Fg) is reported to participate in inflammation through Toll-like receptor 4 (TLR4). However, it remains unknown whether Fg might induce podocyte damage through TLR4 and be related to disease activity in patients with focal segmental glomerulosclerosis (FSGS). METHODS We observed Fg-induced alterations in actin and apoptosis in cultured human podocytes transfected with or without TLR4 siRNA. Expression of TLR4, phospho-p38 MAPK and phospho-NF-κB p65 was evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR) or western blotting, and we analysed urinary Fg levels in adriamycin-treated mice and double immunofluorescence staining for TLR4, Fg and podocin. Urinary Fg changes were also analyzed in FSGS patients under prednisone treatment. RESULTS First, Fg dose-dependently induced actin damage and apoptosis in cultured human podocytes, with an Fg-induced increase in TLR4 expression, and TLR4 siRNA transfection prevented these effects. TLR4 knockdown inhibited activation of p38 MAPK and NF-κB p65 in podocytes. Elevated urinary Fg levels were positively correlated with albuminuria in adriamycin-treated mice, in which Fg and TLR4 colocalized and exhibited increased expression in podocytes. Additionally, elevated urinary Fg levels were positively correlated with 24-h proteinuria and foot process width in FSGS patients. Urinary Fg levels were significantly decreased in patients with complete remission but not in those without remission. CONCLUSIONS Fg induced podocytes injury via the TLR4-p38 MAPK-NF-κB p65 pathway. In FSGS patients, urinary Fg levels reflect therapeutic response to prednisone and disease activity.
Collapse
Affiliation(s)
- Hongtian Wang
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Chunxia Zheng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yue Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yinghui Lu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
28
|
Li Y, Xia W, Zhao F, Wen Z, Zhang A, Huang S, Jia Z, Zhang Y. Prostaglandins in the pathogenesis of kidney diseases. Oncotarget 2018; 9:26586-26602. [PMID: 29899878 PMCID: PMC5995175 DOI: 10.18632/oncotarget.25005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
Prostaglandins (PGs) are important lipid mediators produced from arachidonic acid via the sequential catalyzation of cyclooxygenases (COXs) and specific prostaglandin synthases. There are five subtypes of PGs, namely PGE2, PGI2, PGD2, PGF2α, and thromboxane A2 (TXA2). PGs exert distinct roles by combining to a diverse family of membrane-spanning G protein-coupled prostanoid receptors. The distribution of these PGs, their specific synthases and receptors vary a lot in the kidney. This review summarized the recent findings of PGs together with the COXs and their specific synthases and receptors in regulating renal function and highlighted the insights into their roles in the pathogenesis of various kidney diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Weiwei Xia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Fei Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhaoying Wen
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
29
|
Zhao X, Hwang DY, Kao HY. The Role of Glucocorticoid Receptors in Podocytes and Nephrotic Syndrome. NUCLEAR RECEPTOR RESEARCH 2018; 5. [PMID: 30417008 PMCID: PMC6224173 DOI: 10.11131/2018/101323] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Glucocorticoid receptor (GC), a founding member of the nuclear hormone receptor superfamily, is a glucocorticoid-activated transcription factor that regulates gene expression and controls the development and homeostasis of human podocytes. Synthetic glucocorticoids are the standard treatment regimens for proteinuria (protein in the urine) and nephrotic syndrome (NS) caused by kidney diseases. These include minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), membranous nephropathy (MN) and immunoglobulin A nephropathy (IgAN) or subsequent complications due to diabetes mellitus or HIV infection. However, unwanted side effects and steroid-resistance remain major issues for their long-term use. Furthermore, the mechanism by which glucocorticoids elicit their renoprotective activity in podocyte and glomeruli is poorly understood. Podocytes are highly differentiated epithelial cells that contribute to the integrity of kidney glomerular filtration barrier. Injury or loss of podocytes leads to proteinuria and nephrotic syndrome. Recent studies in multiple experimental models have begun to explore the mechanism of GC action in podocytes. This review will discuss progress in our understanding of the role of glucocorticoid receptor and glucocorticoids in podocyte physiology and their renoprotective activity in nephrotic syndrome.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | - Daw-Yang Hwang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| |
Collapse
|
30
|
Xu X, Wang L, Luo Z, Ni Y, Sun H, Gao X, Li Y, Zhang S, Li Y, Wei S. Facile and Versatile Strategy for Construction of Anti-Inflammatory and Antibacterial Surfaces with Polydopamine-Mediated Liposomes Releasing Dexamethasone and Minocycline for Potential Implant Applications. ACS APPLIED MATERIALS & INTERFACES 2017; 9:43300-43314. [PMID: 29140074 DOI: 10.1021/acsami.7b06295] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Reducing early nonbacterial inflammation induced by implanted materials and infection resulting from bacterial contamination around the implant-abutment interface could greatly decrease implant failure rates, which would be of clinical significance. In this work, we presented a facile and versatile strategy for the construction of anti-inflammatory and antibacterial surfaces. Briefly, the surfaces of polystyrene culture plates were first coated with polydopamine and then decorated with dexamethasone plus minocycline-loaded liposomes (Dex/Mino liposomes), which was validated by contact angle goniometry, quartz crystal microbalance, and fluorescence microscopy. Dex/Mino liposomes were dispersed on functional surfaces and the drug release kinetics exhibited the sustained release of dexamethasone and minocycline. Our results demonstrated that the Dex/Mino liposome-modified surfaces had good biocompatibility. Additionally, liposomal dexamethasone reduced proinflammatory mediator expression (particularly IL-6 and TNF-α) in lipopolysaccharide-stimulated human gingival fibroblasts and human mesenchymal stem cells. Moreover, liposomal minocycline prevented the adhesion and proliferation of Porphyromonas gingivalis (Gram-negative bacteria) and Streptococcus mutans (Gram-positive bacteria). These findings demonstrate that an anti-inflammatory and antibacterial surface was developed, using dopamine as a medium and combining a liposomal delivery device, which has potential for use to reduce implant failure rates. Accordingly, the surface modification strategy presented could be useful in biofunctionalization of implant materials.
Collapse
Affiliation(s)
- Xiao Xu
- Central Laboratory/Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology , Beijing 100081, P. R. China
| | - Lixin Wang
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University , Beijing 100038, P. R. China
| | - Zuyuan Luo
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University , Beijing 100871, P. R. China
| | - Yaofeng Ni
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University , Beijing 100038, P. R. China
| | - Haitao Sun
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University , Beijing 100038, P. R. China
| | - Xiang Gao
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University , Chongqing 401147, P. R. China
| | - Yongliang Li
- Central Laboratory/Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology , Beijing 100081, P. R. China
| | - Siqi Zhang
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University , Beijing 100871, P. R. China
| | - Yan Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University , Beijing 100871, P. R. China
| | - Shicheng Wei
- Central Laboratory/Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology , Beijing 100081, P. R. China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University , Beijing 100871, P. R. China
| |
Collapse
|
31
|
Nie X, Chanley MA, Pengal R, Thomas DB, Agrawal S, Smoyer WE. Pharmacological and genetic inhibition of downstream targets of p38 MAPK in experimental nephrotic syndrome. Am J Physiol Renal Physiol 2017; 314:F602-F613. [PMID: 29187369 DOI: 10.1152/ajprenal.00207.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Nie X, Chanley MA, Pengal R, Thomas DB, Agrawal S, Smoyer WE. Pharmacological and genetic inhibition of downstream targets of p38 MAPK in experimental nephrotic syndrome. Am J Physiol Renal Physiol 314: F602-F613, 2018. First published November 29, 2017; doi: 10.1152/ajprenal.00207.2017 .-The p38 MAPK pathway plays a crucial role in various glomerulopathies, with activation being associated with disease and inhibition being associated with disease amelioration. We hypothesized that the downstream targets of p38 MAPK, MAPK-activated protein kinase 2 and/or 3 (MK2 and/or MK3), play an important role in mediating injury in experimental nephrotic syndrome via their actions on their downstream substrates heat shock protein B1 (HSPB1) and cyclooxygenase-2 (COX-2). To test this hypothesis, the effects of both pharmacological and genetic inhibition of MK2 and MK3 were examined in mouse adriamycin (ADR) and rat puromycin aminonucleoside (PAN) nephropathy models. MK2-/-, MK3-/-, and MK2-/-MK3-/- mice were generated in the Sv129 background and subjected to ADR-induced nephropathy. MK2 and MK3 protein expression was completely abrogated in the respective knockout genotypes, and massive proteinuria and renal histopathological changes developed after ADR treatment. Furthermore, renal cortical HSPB1 was induced in all four genotypes by day 21, but HSPB1 was activated only in the wild-type and MK3-/- mice. Expression of the stress proteins HSPB8 and glucose-regulated protein 78 (GRP78) remained unaltered across all genotypes. Finally, while MK2 and/or MK3-knockout downregulated the proinflammatory enzyme COX-2, ADR significantly induced renal cortical COX-2 only in MK2-/- mice. Additionally, pharmacological MK2 inhibition with PF-318 during PAN-induced nephropathy did not result in significant proteinuria reduction in rats. Together, these data suggest that while the inhibition of MK2 and/or MK3 regulates the renal stress response, our currently available approaches are not yet able to safely and effectively reduce proteinuria in experimental nephrotic syndrome and that other p38MAPK downstream targets should also be considered to improve the future treatment of glomerular disease.
Collapse
Affiliation(s)
- Xiaojing Nie
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,Department of Pediatrics, Fuzhou Dongfang Hospital, Xiamen University , Fuzhou , China
| | - Melinda A Chanley
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Ruma Pengal
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - David B Thomas
- University of Miami Miller School of Medicine , Miami, Florida
| | - Shipra Agrawal
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,Department of Pediatrics, College of Medicine, The Ohio State University , Columbus, Ohio
| | - William E Smoyer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,Department of Pediatrics, College of Medicine, The Ohio State University , Columbus, Ohio
| |
Collapse
|
32
|
Abstract
Nephrotic syndrome is a highly prevalent disease that is associated with high morbidity despite notable advances in its treatment. Many of the complications of nephrotic syndrome, including the increased risk of atherosclerosis and thromboembolism, can be linked to dysregulated lipid metabolism and dyslipidaemia. These abnormalities include elevated plasma levels of cholesterol, triglycerides and the apolipoprotein B-containing lipoproteins VLDL and IDL; decreased lipoprotein lipase activity in the endothelium, muscle and adipose tissues; decreased hepatic lipase activity; and increased levels of the enzyme PCSK9. In addition, there is an increase in the plasma levels of immature HDL particles and reduced cholesterol efflux. Studies from the past few years have markedly improved our understanding of the molecular pathogenesis of nephrotic syndrome-associated dyslipidaemia, and also heightened our awareness of the associated exacerbated risks of cardiovascular complications, progressive kidney disease and thromboembolism. Despite the absence of clear guidelines regarding treatment, various strategies are being increasingly utilized, including statins, bile acid sequestrants, fibrates, nicotinic acid and ezetimibe, as well as lipid apheresis, which seem to also induce partial or complete clinical remission of nephrotic syndrome in a substantial percentage of patients. Future potential treatments will likely also include inhibition of PCSK9 using recently-developed anti-PCSK9 monoclonal antibodies and small inhibitory RNAs, as well as targeting newly identified molecular regulators of lipid metabolism that are dysregulated in nephrotic syndrome.
Collapse
|
33
|
Huang W, Liu H, Zhu S, Woodson M, Liu R, Tilton RG, Miller JD, Zhang W. Sirt6 deficiency results in progression of glomerular injury in the kidney. Aging (Albany NY) 2017; 9:1069-1083. [PMID: 28351995 PMCID: PMC5391219 DOI: 10.18632/aging.101214] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 03/18/2017] [Indexed: 02/06/2023]
Abstract
Aging is associated with an increased incidence and prevalence of renal glomerular diseases. Sirtuin (Sirt) 6, a nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase, has been shown to protect against multiple age-associated phenotypes; however it is unknown whether Sirt6 has a direct pathophysiologic role in the kidney. In the present study, we demonstrate that Sirt6 is expressed in the kidney and aging Sirt6-deficient mice exhibit renal hypertrophy with glomerular enlargement. Sirt6 deletion induces podocyte injury, including decreases in slit diaphragm proteins, foot process effacement, and cellular loss, resulting in proteinuria. Knockdown of Sirt6 in cultured primary murine podocytes induces shape changes with loss of process formation and cell apoptosis. Moreover, Sirt6 deficiency results in progressive renal inflammation and fibrosis. Collectively, these data provide compelling evidence that Sirt6 is important for podocyte homeostasis and maintenance of glomerular function, and warrant further investigation into the role of Sirt6 in age-associated kidney dysfunction.
Collapse
Affiliation(s)
- Wen Huang
- Department of Healthcare, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China.,Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hua Liu
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shuang Zhu
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Michael Woodson
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rong Liu
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ronald G Tilton
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jordan D Miller
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Wenbo Zhang
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
34
|
Sharma I, Tupe RS, Wallner AK, Kanwar YS. Contribution of myo-inositol oxygenase in AGE:RAGE-mediated renal tubulointerstitial injury in the context of diabetic nephropathy. Am J Physiol Renal Physiol 2017; 314:F107-F121. [PMID: 28931523 DOI: 10.1152/ajprenal.00434.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advanced glycation end products (AGEs) play a role in pathogenesis of diabetic nephropathy (DN). Myo-inositol oxygenase (MIOX) has been implicated in tubulointerstitial injury in the context of DN. We investigated the effect of AGEs on MIOX expression and delineated mechanisms that lead to tubulointerstitial injury. The status of MIOX, RAGE, and relevant cellular signaling pathways activated following AGE:RAGE interaction was examined in tubular cells and kidneys of AGE-BSA-treated mice. A solid-phase assay revealed an enhanced binding of RAGE with AGE-BSA, AGE-laminin, and AGE-collagen IV. The cells treated with AGE-BSA had increased MIOX activity/expression and promoter activity. This was associated with activation of various signaling kinases of phosphatidylinositol 3-kinase (PI3K)-AKT pathway and increased expression of NF-κB, transforming growth factor (TGF)-β, and fibronectin, which was negated with the treatment of MIOX/RAGE- small interfering (si) RNA. Concomitant with MIOX upregulation, there was an increased generation of reactive oxygen species (ROS), which could be abrogated with MIOX/RAGE- siRNA treatment. The kidneys of mice treated with AGE-BSA had significantly high urinary A/C ratio, upregulation of MIOX, RAGE and NF-κB, along with influx of monocytes into the tubulointerstitium, increased the expression of MCP-1, IL-6, and fibronectin and increased the generation of ROS. Such perturbations were abrogated with the concomitant treatment of inhibitors MIOX or RAGE (d-glucarate and FPS-ZM1). These studies support a role of AGE:RAGE interaction in the activation of PI3K-AKT pathway and upregulation of MIOX, with excessive generation of ROS, increased expression of NF-κB, inflammatory cytokines, TGF-β, and fibronectin. Collectively, these observations highlight the relevance of the biology of MIOX in the contribution toward tubulointerstitial injury in DN.
Collapse
Affiliation(s)
- Isha Sharma
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Rashmi S Tupe
- Biochemical Sciences Division, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth University , Pune , India
| | - Aryana K Wallner
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Yashpal S Kanwar
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
35
|
Sharma R, Waller AP, Agrawal S, Wolfgang KJ, Luu H, Shahzad K, Isermann B, Smoyer WE, Nieman MT, Kerlin BA. Thrombin-Induced Podocyte Injury Is Protease-Activated Receptor Dependent. J Am Soc Nephrol 2017; 28:2618-2630. [PMID: 28424276 PMCID: PMC5576925 DOI: 10.1681/asn.2016070789] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 03/16/2017] [Indexed: 12/29/2022] Open
Abstract
Nephrotic syndrome is characterized by massive proteinuria and injury of specialized glomerular epithelial cells called podocytes. Studies have shown that, whereas low-concentration thrombin may be cytoprotective, higher thrombin concentrations may contribute to podocyte injury. We and others have demonstrated that ex vivo plasma thrombin generation is enhanced during nephrosis, suggesting that thrombin may contribute to nephrotic progression. Moreover, nonspecific thrombin inhibition has been shown to decrease proteinuria in nephrotic animal models. We thus hypothesized that thrombin contributes to podocyte injury in a protease-activated receptor-specific manner during nephrosis. Here, we show that specific inhibition of thrombin with hirudin reduced proteinuria in two rat nephrosis models, and thrombin colocalized with a podocyte-specific marker in rat glomeruli. Furthermore, flow cytometry immunophenotyping revealed that rat podocytes express the protease-activated receptor family of coagulation receptors in vivo High-concentration thrombin directly injured conditionally immortalized human and rat podocytes. Using receptor-blocking antibodies and activation peptides, we determined that thrombin-mediated injury depended upon interactions between protease-activated receptor 3 and protease-activated receptor 4 in human podocytes, and between protease-activated receptor 1 and protease-activated receptor 4 in rat podocytes. Proximity ligation and coimmunoprecipitation assays confirmed thrombin-dependent interactions between human protease-activated receptor 3 and protease-activated receptor 4, and between rat protease-activated receptor 1 and protease-activated receptor 4 in cultured podocytes. Collectively, these data implicate thrombinuria as a contributor to podocyte injury during nephrosis, and suggest that thrombin and/or podocyte-expressed thrombin receptors may be novel therapeutic targets for nephrotic syndrome.
Collapse
Affiliation(s)
- Ruchika Sharma
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
- Division of Hematology, Oncology, and BMT, and
| | - Amanda P Waller
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
| | - Shipra Agrawal
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
| | - Katelyn J Wolfgang
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
| | - Hiep Luu
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
- Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Khurrum Shahzad
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University, Magdeburg, Germany
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan; and
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University, Magdeburg, Germany
| | - William E Smoyer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital
- Division of Nephrology, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Bryce A Kerlin
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital,
- Division of Hematology, Oncology, and BMT, and
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
36
|
Diabetic nephropathy - is this an immune disorder? Clin Sci (Lond) 2017; 131:2183-2199. [PMID: 28760771 DOI: 10.1042/cs20160636] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/04/2017] [Accepted: 07/10/2017] [Indexed: 02/06/2023]
Abstract
Chronic diabetes is associated with metabolic and haemodynamic stresses which can facilitate modifications to DNA, proteins and lipids, induce cellular dysfunction and damage, and stimulate inflammatory and fibrotic responses which lead to various types of renal injury. Approximately 30-40% of patients with diabetes develop nephropathy and this renal injury normally progresses in about a third of patients. Due to the growing incidence of diabetes, diabetic nephropathy is now the main cause of end-stage renal disease (ESRD) worldwide. Accumulating evidence from experimental and clinical studies has demonstrated that renal inflammation plays a critical role in determining whether renal injury progresses during diabetes. However, the immune response associated with diabetic nephropathy is considerably different to that seen in autoimmune kidney diseases or in acute kidney injury arising from episodes of ischaemia or infection. This review evaluates the role of the immune system in the development of diabetic nephropathy, including the specific contributions of leucocyte subsets (macrophages, neutrophils, mast cells, T and B lymphocytes), danger-associated molecular patterns (DAMPs), inflammasomes, immunoglobulin and complement. It also examines factors which may influence the development of the immune response, including genetic factors and exposure to other kidney insults. In addition, this review discusses therapies which are currently under development for targeting the immune system in diabetic nephropathy and indicates those which have proceeded into clinical trials.
Collapse
|
37
|
Role of albumin and its modifications in glomerular injury. Pflugers Arch 2017; 469:975-982. [PMID: 28735420 DOI: 10.1007/s00424-017-2029-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 02/06/2023]
Abstract
Albuminuria is both a characteristic hallmark and a known risk factor for progressive glomerular disease. Although the molecular basis for a potential causative role for albuminuria in progressive chronic kidney disease remains poorly understood, there have been several recent advances in our understanding of the role of albumin, and its molecular modifications, in the development and progression of glomerular disease. This review discusses recent findings related to the ability of albumin and its associated factors to directly induce podocyte and glomerular injury. Additional recent studies confirming the ability and mechanisms by which podocytes endocytose albumin are also discussed. Lastly, we present several known molecular modifications in the albumin molecule itself, as well as substances bound to it, which may be important and potentially clinically relevant mediators of albumin-induced glomerular injury. These recent findings may create entirely new opportunities to develop novel future therapies directed at albumin that could potentially help reduce podocyte and renal tubular injury and slow the progression of chronic glomerular disease.
Collapse
|
38
|
Motojima M, Kume T, Matsusaka T. Foxc1 and Foxc2 are necessary to maintain glomerular podocytes. Exp Cell Res 2017; 352:265-272. [PMID: 28223138 DOI: 10.1016/j.yexcr.2017.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/07/2017] [Accepted: 02/11/2017] [Indexed: 12/11/2022]
Abstract
Foxc1 and Foxc2 (Foxc1/2) are transcription factors involved in many biological processes. In adult kidneys, expression of Foxc1/2 is confined to the glomerular epithelial cells, i.e., podocytes. To bypass embryonic lethality of Foxc1/2 null mice, mice ubiquitously expressing inducible-Cre (ROSA26-CreERT2) or mice expressing Cre in podocytes (Nephrin-Cre) were mated with floxed-Foxc1 and floxed-Foxc2 mice. The CreERT2 was activated in adult mice by administrations of tamoxifen. Eight weeks after tamoxifen treatment, ROSA26-CreERT2; Foxc1+/flox; Foxc2flox/flox mice developed microalbuminuria, while ROSA26-Cre ERT2; Foxc1flox/flox; Foxc2+/flox mice had no microalbuminuria. The kidneys of conditional-Foxc1/2 null mice showed proteinaceous casts, protein reabsorption droplets in tubules and huge vacuoles in podocytes, indicating severe podocyte injury and massive proteinuria. Comparison of gene expression profiles revealed that Foxc1/2 maintain expression of genes necessary for podocyte function such as podocin and Cxcl12. In addition, mice with an innate podocyte-specific deletion of Foxc1/2 by Nephrin-Cre develop similar podocyte injury. These results demonstrate dose-dependence of Foxc1/2 gene in maintaining the podocyte with a more critical role for Foxc2 than Foxc1 and a critical role of Foxc1/2 in regulating expression of genes that maintain podocyte integrity.
Collapse
Affiliation(s)
- Masaru Motojima
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan.
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Taiji Matsusaka
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
39
|
Lan X, Lederman R, Eng JM, Shoshtari SSM, Saleem MA, Malhotra A, Singhal PC. Nicotine Induces Podocyte Apoptosis through Increasing Oxidative Stress. PLoS One 2016; 11:e0167071. [PMID: 27907022 PMCID: PMC5132002 DOI: 10.1371/journal.pone.0167071] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022] Open
Abstract
Background Cigarette smoking plays an important role in the progression of chronic kidney disease (CKD). Nicotine, one of the major components of cigarette smoking, has been demonstrated to increase proliferation of renal mesangial cells. In this study, we examined the effect of nicotine on podocyte injury. Methods To determine the expression of nicotinic acetylcholine receptors (nAChR subunits) in podocytes, cDNAs and cell lysate of cultured human podocytes were used for the expression of nAChR mRNAs and proteins, respectively; and mouse renal cortical sections were subjected to immunofluorescant staining. We also studied the effect of nicotine on podocyte nephrin expression, reactive oxygen species (ROS) generation (via DCFDA loading followed by fluorometric analysis), proliferation, and apoptosis (morphologic assays). We evaluated the effect of nicotine on podocyte downstream signaling including phosphorylation of ERK1/2, JNK, and p38 and established causal relationships by using respective inhibitors. We used nAChR antagonists to confirm the role of nicotine on podocyte injury. Results Human podocytes displayed robust mRNA and protein expression of nAChR in vitro studies. In vivo studies, mice renal cortical sections revealed co-localization of nAChRs along with synaptopodin. In vitro studies, nephrin expression in podocyte was decreased by nicotine. Nicotine stimulated podocyte ROS generation; nonetheless, antioxidants such as N-acetyl cysteine (NAC) and TEMPOL (superoxide dismutase mimetic agent) inhibited this effect of nicotine. Nicotine did not modulate proliferation but promoted apoptosis in podocytes. Nicotine enhanced podocyte phosphorylation of ERK1/2, JNK, and p38, and their specific inhibitors attenuated nicotine-induced apoptosis. nAChR antagonists significantly suppressed the effects of nicotine on podocyte. Conclusions Nicotine induces podocyte apoptosis through ROS generation and associated downstream MAPKs signaling. The present study provides insight into molecular mechanisms involved in smoking associated progression of chronic kidney disease.
Collapse
Affiliation(s)
- Xiqian Lan
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
- * E-mail: (XL); (PS)
| | - Rivka Lederman
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Judith M. Eng
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Seyedeh Shadafarin Marashi Shoshtari
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Moin A. Saleem
- Academic Renal Unit, Southmead Hospital, Bristol, United Kingdom
| | - Ashwani Malhotra
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Pravin C. Singhal
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
- * E-mail: (XL); (PS)
| |
Collapse
|
40
|
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
|
41
|
Pullen N, Fornoni A. Drug discovery in focal and segmental glomerulosclerosis. Kidney Int 2016; 89:1211-20. [PMID: 27165834 PMCID: PMC4875964 DOI: 10.1016/j.kint.2015.12.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 11/26/2022]
Abstract
Despite the high medical burden experienced by patients with focal segmental glomerulosclerosis, the etiology of the condition remains largely unknown. Focal segmental glomerulosclerosis is highly heterogeneous in clinical and morphologic manifestations. While this presents challenges for the development of new treatments, research investments over the last 2 decades have yielded a surfeit of potential avenues for therapeutic intervention. The development of many of those ideas and concepts into new therapies, however, has been very disappointing. Here, we describe some of the factors that have potentially contributed to the poor translational performance from this research investment, including the confidence we ascribe to a target, the conduct of experimental studies, and the availability of selective reagents to test hypotheses. We will discuss the significance of genetic and systems traits as well as other methods for reducing bias. We will analyze the limitations of a successful drug development. We will use specific examples hoping that these will guide a consensus for investment and drive greater translational quality. We hope that this substrate will serve to exemplify the tremendous opportunity for intervention as well as facilitate greater collaborative effort between industry, academia, and private foundations in promoting appropriate validation of these targets. Only then will we have achieved our goal for curative therapies for this devastating disease.
Collapse
Affiliation(s)
- Nick Pullen
- Pfizer Global Research & Development, Cambridge, Massachusetts, USA.
| | - Alessia Fornoni
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
42
|
Agrawal S, Chanley MA, Westbrook D, Nie X, Kitao T, Guess AJ, Benndorf R, Hidalgo G, Smoyer WE. Pioglitazone Enhances the Beneficial Effects of Glucocorticoids in Experimental Nephrotic Syndrome. Sci Rep 2016; 6:24392. [PMID: 27142691 PMCID: PMC4855145 DOI: 10.1038/srep24392] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/21/2016] [Indexed: 12/25/2022] Open
Abstract
Glucocorticoids are the primary therapy for nephrotic syndrome (NS), but have serious side effects and are ineffective in ~20-50% of patients. Thiazolidinediones have recently been suggested to be renoprotective, and to modulate podocyte glucocorticoid-mediated nuclear receptor signaling. We hypothesized that thiazolidinediones could enhance glucocorticoid efficacy in NS. We found that puromycin aminonucleoside-induced proteinuria in rats was significantly reduced by both high-dose glucocorticoids (79%) and pioglitazone (61%), but not low-dose glucocorticoids (25%). Remarkably, pioglitazone + low-dose glucocorticoids also reduced proteinuria (63%) comparably to high-dose glucocorticoids, whereas pioglitazone + high-dose glucocorticoids reduced proteinuria to almost control levels (97%). Molecular analysis revealed that both glucocorticoids and pioglitazone enhanced glomerular synaptopodin and nephrin expression, and reduced COX-2 expression, after injury. Furthermore, the glomerular phosphorylation of glucocorticoid receptor and Akt, but not PPARγ, correlated with treatment-induced reductions in proteinuria. Notably, clinical translation of these findings to a child with refractory NS by the addition of pioglitazone to the treatment correlated with marked reductions in both proteinuria (80%) and overall immunosuppression (64%). These findings together suggest that repurposing pioglitazone could potentially enhance the proteinuria-reducing effects of glucocorticoids during NS treatment.
Collapse
Affiliation(s)
- S Agrawal
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - M A Chanley
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - D Westbrook
- James and Connie Maynard Children's Hospital, Greenville, NC, USA
| | - X Nie
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - T Kitao
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - A J Guess
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - R Benndorf
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - G Hidalgo
- James and Connie Maynard Children's Hospital, Greenville, NC, USA.,Department of Pediatrics, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - W E Smoyer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
43
|
Yu J, Gong W, Wu Y, Li S, Cui Y, Ma Y, Zhang Y, Yang G, Huang S, Jia Z, Zhang A. mPGES-1-derived PGE2 contributes to adriamycin-induced podocyte injury. Am J Physiol Renal Physiol 2016; 310:F492-8. [PMID: 26739892 DOI: 10.1152/ajprenal.00499.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022] Open
Abstract
Podocyte damage is a common pathological feature in many types of glomerular diseases and is involved in the occurrence and progression of kidney disease. However, the pathogenic mechanisms leading to podocyte injury are still uncertain. The present study was undertaken to investigate the role of microsomal PGE synthase (mPGES)-1 in adriamycin (ADR)-induced podocyte injury as well as the underlying mechanism. In both mouse kidneys and in vitro podocytes, application of ADR remarkably enhanced mPGES-1 expression in line with a stimulation of cyclooxygenase-2. Interestingly, inhibition of mPGES-1 with a small interfering RNA approach significantly attenuated ADR-induced downregualtion of podocin and nephrin. Moreover, ADR-induced podocyte apoptosis was also markedly blocked in parallel with blunted caspase-3 induction. In agreement with the improvement of cell phenotypic alteration and apoptosis, the enhanced inflammatory markers of IL-1β and TNF-α were also significantly suppressed by mPGES-1 silencing. More importantly, in mPGES-1-deficient mice, albuminuria induced by ADR showed a remarkable attenuation in line with decreased urinary output of PGE2 and TNF-α, highly suggesting an in vivo role of mPGES-1 in mediating podocyte injury. In summary, findings from the present study offered the first evidence demonstrating a pathogenic role of mPGES-1 in mediating ADR-induced podocyte injury possibly via triggering an inflammatory response.
Collapse
Affiliation(s)
- Jing Yu
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Wei Gong
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Yimei Wu
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Shuzhen Li
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Yiyun Cui
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Yifei Ma
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Guangrui Yang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; and
- Nanjing Key Laboratory of Pediatrics, Nanjing, China
| |
Collapse
|
44
|
Bassi R, Fornoni A, Doria A, Fiorina P. CTLA4-Ig in B7-1-positive diabetic and non-diabetic kidney disease. Diabetologia 2016; 59:21-29. [PMID: 26409459 PMCID: PMC5003171 DOI: 10.1007/s00125-015-3766-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/19/2015] [Indexed: 01/10/2023]
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease in the Western world. Standard treatments have ultimately proven ineffective in blocking DKD progression, thus necessitating the design of new therapies to complement glycaemic and blood pressure control. High glucose levels upregulate the immune-related molecule B7-1 in podocytes, and such an event may play a relevant role in DKD onset, suggesting that B7-1 is a suitable therapeutic target for DKD. CTLA4-Ig is a clinically available fusion protein, approved for the treatment of some autoimmune diseases, which binds B7-1 and blocks its signalling. We have previously demonstrated that CTLA4-Ig restores the physiological structure and cellular motility of podocytes challenged with high glucose in vitro and abrogates the onset of proteinuria in murine models of DKD in vivo. Notably, these beneficial effects occurred independently of any systemic immunological effects of CTLA4-Ig. While the expression of B7-1 on podocytes raises questions regarding the very nature of the podocyte as we know it, the preliminary positive effect of CTLA4-Ig on proteinuria in preclinical models and the evidence of B7-1 expression in kidney biopsies of diabetic individuals suggest a potential novel indication for CTLA4-Ig in DKD. Nonetheless, recent reports of problems with detecting podocyte B7-1 and of inconsistent therapeutic efficacy of CTLA4-Ig in proteinuric patients highlight the necessity to establish uniformly accepted protocols for the detection of B7-1 and underline the need for randomised trials with CTLA4-Ig in kidney diseases.
Collapse
Affiliation(s)
- Roberto Bassi
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave., Enders Building, Boston, MA, 02115, USA
- Department of Transplant Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA, USA
| | - Paolo Fiorina
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave., Enders Building, Boston, MA, 02115, USA.
- Department of Transplant Medicine, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
45
|
Inoue K, Ishibe S. Podocyte endocytosis in the regulation of the glomerular filtration barrier. Am J Physiol Renal Physiol 2015; 309:F398-405. [PMID: 26084928 DOI: 10.1152/ajprenal.00136.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Severe defects in the glomerular filtration barrier result in nephrotic syndrome, which is characterized by massive proteinuria. The podocyte, a specialized epithelial cell with interdigitating foot processes separated by a slit diaphragm, plays a vital role in regulating the passage of proteins from the capillary lumen to Bowman's space. Recent findings suggest a critical role for endocytosis in podocyte biology as highlighted by genetic mouse models of disease and human genetic mutations that result in the loss of the integrity of the glomerular filtration barrier. In vitro podocyte studies have also unraveled a plethora of constituents that are differentially internalized to maintain homeostasis. These observations provide a framework and impetus for understanding the precise regulation of podocyte endocytic machinery in both health and disease.
Collapse
Affiliation(s)
- Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
46
|
Morita T, Nakano D, Kitada K, Morimoto S, Ichihara A, Hitomi H, Kobori H, Shiojima I, Nishiyama A. Chelation of dietary iron prevents iron accumulation and macrophage infiltration in the type I diabetic kidney. Eur J Pharmacol 2015; 756:85-91. [PMID: 25820160 DOI: 10.1016/j.ejphar.2015.03.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 03/12/2015] [Accepted: 03/17/2015] [Indexed: 11/24/2022]
Abstract
We previously reported that the functional deletion of p21, a cyclin-dependent kinase inhibitor, in mice attenuated renal cell senescence in streptozotocin (STZ)-induced type 1 diabetic mice. In the present study, we investigated the effect of iron chelation on renal cell senescence and inflammation in the type 1 diabetic kidney. STZ-treated mice showed increase in iron accumulation, tubular cell senescence and macrophage infiltration at week 28 in the kidney. Administering deferasirox, which removes only dietary iron, significantly attenuated iron accumulation in proximal tubules and the number of infiltrating F4/80-positive cells without effecting blood glucose, hematocrit or hemoglobin levels. In contrast however, deferasirox did not influence renal cell senescence. The lack of p21 decreased the renal tubular iron accumulation and did not change tubular cell senescence. Interestingly, the STZ-treated animals showed an increase in p16, another cyclin-dependent kinase inhibitor. The results suggest that type 1 diabetes increases renal tubular iron accumulation and macrophage infiltration through a p21-dependent mechanism, and that the chelation of dietary iron attenuates these responses.
Collapse
Affiliation(s)
- Tatsuyori Morita
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan; The Second Department of Internal Medicine, Kansai Medical University, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan.
| | - Kento Kitada
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Hirofumi Hitomi
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan
| | - Hiroyuki Kobori
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan
| | - Ichiro Shiojima
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, 1750-1 Miki, Kita, Kagawa, Japan
| |
Collapse
|
47
|
Role of COX-2/mPGES-1/prostaglandin E2 cascade in kidney injury. Mediators Inflamm 2015; 2015:147894. [PMID: 25729216 PMCID: PMC4333324 DOI: 10.1155/2015/147894] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/19/2015] [Indexed: 12/26/2022] Open
Abstract
COX-2/mPGES-1/PGE2 cascade plays critical roles in modulating many physiological and pathological actions in different organs. In the kidney, this cascade is of high importance in regulating fluid metabolism, blood pressure, and renal hemodynamics. Under some disease conditions, this cascade displays various actions in response to the different pathological insults. In the present review, the roles of this cascade in the pathogenesis of kidney injuries including diabetic and nondiabetic kidney diseases and acute kidney injuries were introduced and discussed. The new insights from this review not only increase the understanding of the pathological role of the COX-2/mPGES-1/PGE2 pathway in kidney injuries, but also shed new light on the innovation of the strategies for the treatment of kidney diseases.
Collapse
|