1
|
Xu Q, Li J, Zhuo L, Gao H, Yang Y, Li W. RACGAP1 is a pivotal gene in lung adenocarcinoma-associated membranous nephropathy: Based on comprehensive bioinformatics analysis and machine learning. Int Immunopharmacol 2024; 139:112783. [PMID: 39068752 DOI: 10.1016/j.intimp.2024.112783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND This study performs a detailed bioinformatics and machine learning analysis to investigate the genetic foundations of membranous nephropathy (MN) in lung adenocarcinoma (LUAD). METHODS In this study, the gene expression profiles of MN microarray datasets (GSE99339) and LUAD dataset (GSE43767) were downloaded from the Gene Expression Omnibus database, common differentially expressed genes (DEGs) were obtained using the limma R package. The biological functions were analyzed with R Cluster Profiler package according to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Machine learning algorithms, including LASSO regression, support vector machine (SVM), Random Forest, and Boruta analysis, were applied to identify hubgenes linked to LUAD-associated MN. These genes' prognostic values were evaluated in the TCGA-LUAD cohort and validated through immunohistochemistry on renal biopsy specimens. RESULTS A total of 36 DEGs in common were identified for downstream analyses. Functional enrichment analysis highlighted the involvement of the Toll-like receptor 4 pathway and several immune recognition pathways in LUAD-associated MN. COL3A1, PSENEN, RACGAP1, and TNFRSF10B were identified as hub genes in LUAD-associated MN using machine learning algorithms. ROC analysis demonstrated their effective discrimination of MN with high accuracy. Survival analysis showed that lung adenocarcinoma patients with higher expression of these genes had significantly reduced overall survival. In patients with lung adenocarcinoma-associated MN, RACGAP1, COL3A1, PSENEN, and TNFRSF10B were higher expressed in the glomerular, especially RACGAP1, indicating an important role in the pathogenesis of LUAD-associated membranous nephropathy. CONCLUSIONS Our study underscores the critical role of RACGAP1, COL3A1, PSENEN, and TNFRSF10B in the development of LUAD-associated MN, providing important insights for future research and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Qianqian Xu
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Jiayi Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China; Department of Nephrology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Li Zhuo
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Hongmei Gao
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Yue Yang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China; Department of Nephrology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| |
Collapse
|
2
|
Lucero CM, Navarro L, Barros-Osorio C, Cáceres-Conejeros P, Orellana JA, Gómez GI. Activation of Pannexin-1 channels causes cell dysfunction and damage in mesangial cells derived from angiotensin II-exposed mice. Front Cell Dev Biol 2024; 12:1387234. [PMID: 38660621 PMCID: PMC11041381 DOI: 10.3389/fcell.2024.1387234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Chronic kidney disease (CKD) is a prevalent health concern associated with various pathological conditions, including hypertensive nephropathy. Mesangial cells are crucial in maintaining glomerular function, yet their involvement in CKD pathogenesis remains poorly understood. Recent evidence indicates that overactivation of Pannexin-1 (Panx1) channels could contribute to the pathogenesis and progression of various diseases. Although Panx1 is expressed in the kidney, its contribution to the dysfunction of renal cells during pathological conditions remains to be elucidated. This study aimed to investigate the impact of Panx1 channels on mesangial cell function in the context of hypertensive nephropathy. Using an Ang II-infused mouse model and primary mesangial cell cultures, we demonstrated that in vivo exposure to Ang II sensitizes cultured mesangial cells to show increased alterations when they are subjected to subsequent in vitro exposure to Ang II. Particularly, mesangial cell cultures treated with Ang II showed elevated activity of Panx1 channels and increased release of ATP. The latter was associated with enhanced basal intracellular Ca2+ ([Ca2+]i) and increased ATP-mediated [Ca2+]i responses. These effects were accompanied by increased lipid peroxidation and reduced cell viability. Crucially, all the adverse impacts evoked by Ang II were prevented by the blockade of Panx1 channels, underscoring their critical role in mediating cellular dysfunction in mesangial cells. By elucidating the mechanisms by which Ang II negatively impacts mesangial cell function, this study provides valuable insights into the pathogenesis of renal damage in hypertensive nephropathy.
Collapse
Affiliation(s)
- Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Laura Navarro
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Cristián Barros-Osorio
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Patricio Cáceres-Conejeros
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
3
|
Patyal P, Zhang X, Verma A, Azhar G, Wei JY. Inhibitors of Rho/MRTF/SRF Transcription Pathway Regulate Mitochondrial Function. Cells 2024; 13:392. [PMID: 38474356 PMCID: PMC10931493 DOI: 10.3390/cells13050392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
RhoA-regulated gene transcription by serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factors (MRTFs) signaling pathway has emerged as a promising therapeutic target for pharmacological intervention in multiple diseases. Altered mitochondrial metabolism is one of the major hallmarks of cancer, therefore, this upregulation is a vulnerability that can be targeted with Rho/MRTF/SRF inhibitors. Recent advances identified a novel series of oxadiazole-thioether compounds that disrupt the SRF transcription, however, the direct molecular target of these compounds is unclear. Herein, we demonstrate the Rho/MRTF/SRF inhibition mechanism of CCG-203971 and CCG-232601 in normal cell lines of human lung fibroblasts and mouse myoblasts. Further studies investigated the role of these molecules in targeting mitochondrial function. We have shown that these molecules hyperacetylate histone H4K12 and H4K16 and regulate the genes involved in mitochondrial function and dynamics. These small molecule inhibitors regulate mitochondrial function as a compensatory mechanism by repressing oxidative phosphorylation and increasing glycolysis. Our data suggest that these CCG molecules are effective in inhibiting all the complexes of mitochondrial electron transport chains and further inducing oxidative stress. Therefore, our present findings highlight the therapeutic potential of CCG-203971 and CCG-232601, which may prove to be a promising approach to target aberrant bioenergetics.
Collapse
Affiliation(s)
| | | | | | | | - Jeanne Y. Wei
- Donald W. Reynolds Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (P.P.); (X.Z.); (A.V.); (G.A.)
| |
Collapse
|
4
|
Zhang K, Fu Z, Zhang Y, Chen X, Cai G, Hong Q. The role of cellular crosstalk in the progression of diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1173933. [PMID: 37538798 PMCID: PMC10395826 DOI: 10.3389/fendo.2023.1173933] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/26/2023] [Indexed: 08/05/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the most common complications of diabetes, and its main manifestations are progressive proteinuria and abnormal renal function, which eventually develops end stage renal disease (ESRD). The pathogenesis of DN is complex and involves many signaling pathways and molecules, including metabolic disorders, genetic factors, oxidative stress, inflammation, and microcirculatory abnormalities strategies. With the development of medical experimental techniques, such as single-cell transcriptome sequencing and single-cell proteomics, the pathological alterations caused by kidney cell interactions have attracted more and more attention. Here, we reviewed the characteristics and related mechanisms of crosstalk among kidney cells podocytes, endothelial cells, mesangial cells, pericytes, and immune cells during the development and progression of DN and highlighted its potential therapeutic effects.
Collapse
|
5
|
Saleh MA, Shaaban AA, Talaat IM, Elmougy A, Adra SF, Ahmad F, Qaisar R, Elmoselhi AB, Abu-Gharbieh E, El-Huneidi W, Eladl MA, Shehatou G, Kafl HE. RhoA/ROCK inhibition attenuates endothelin-1-induced glomerulopathy in the rats. Life Sci 2023; 323:121687. [PMID: 37030613 DOI: 10.1016/j.lfs.2023.121687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/25/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023]
Abstract
Endothelin-1 (ET-1) contributes to the development of kidney diseases. However, the underlying molecular mechanism is largely undefined. Here we sought to investigate the potential role of ET-1 receptors, ETA and ETB in the regulation of increased glomerular permeability and underlying signaling pathways post-ET-1 infusion. Male Sprague-Dawley rats were infused with ET-1 (2 pmol/kg per minute, i.v.) for four weeks, and the effect on glomerular permeability to albumin (Palb) and albuminuria was measured. The selective ROCK-1/2 inhibitor, Y-27632, was administered to a separate group of rats to determine its effect on ET-1-induced Palb and albuminuria. The role of ETA and ETB receptors in regulating RhoA/ROCK activity was determined by incubating isolated glomeruli from normal rats with ET-1 and with selective ETA and ETB receptor antagonists. ET-1 infusion for four weeks significantly elevated Palb and albuminuria. Y-27632 significantly reduced the elevation of Palb and albuminuria. The activities of both RhoA and ROCK-1/2 were increased by ET-1 infusion. Selective ETB receptor antagonism had no effect on the elevated activity of both RhoA and ROCK-1/2 enzymes. Selective ETA receptor and combined ETA/ETB receptors blockade restored the activity of RhoA and ROCK-1/2 to normal levels. In addition, chronic ET-1 infusion increased the levels of glomerular inflammatory and fibrotic markers. These effects were all attenuated in rats following ROCK-1/2 inhibition. These observations suggest that ET-1 contributes to increased albuminuria, inflammation, and fibrosis by modulating the activity of the ETA-RhoA/ROCK-1/2 pathway. Selective ETA receptor blockade may represent a potential therapeutic strategy to limit glomerular injury and albuminuria in kidney disease.
Collapse
Affiliation(s)
- Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed A Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City 35712, Egypt
| | - Iman M Talaat
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Atef Elmougy
- Pediatric Nephrology Unit, Mansoura University Children's Hospital, Mansoura University, Mansoura 35516, Egypt
| | - Saryia F Adra
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Firdos Ahmad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
| | - Rizwan Qaisar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Adel B Elmoselhi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - George Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City 35712, Egypt
| | - Hoda E Kafl
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
6
|
Zhang S, Li X, Liu S, Zhang W, Li M, Qiao C. Research progress on the role of ET-1 in diabetic kidney disease. J Cell Physiol 2023; 238:1183-1192. [PMID: 37063089 DOI: 10.1002/jcp.31023] [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: 01/18/2023] [Revised: 03/16/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
Diabetic kidney disease (DKD) is one of the common complications of diabetes mellitus, which usually progresses to end-stage renal disease and causes great damage to the health of patients. Endothelin-1 (ET-1), a molecule closely associated with the progression of DKD, has increased expression in response to high glucose stimulation and is involved in hemodynamic changes, inflammation, glomerular and tubular dysfunction in the kidney, causing an increase in proteinuria and a decrease in glomerular filtration function, ultimately leading to glomerulosclerosis and renal failure. This paper aims to review the molecular level changes, regulatory mechanisms, and mechanisms of action of ET-1 under DKD, clinical trials of ET-1 receptor antagonists in recent years and current problems, to provide basic information and new research directions and ideas for the treatment of DKD and ET-1-related research.
Collapse
Affiliation(s)
- Shenghao Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaodan Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Siyu Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wanting Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meinuo Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chen Qiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Wang H, Wang J, Liu T, Leng Y, Yang W. Stem cell-derived exosomal MicroRNAs: Potential therapies in diabetic kidney disease. Biomed Pharmacother 2023; 164:114961. [PMID: 37257230 DOI: 10.1016/j.biopha.2023.114961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023] Open
Abstract
The diabetic kidney disease (DKD) is chronic kidney disease caused by diabetes and one of the most common comorbidities. It is often more difficult to treat end-stage renal disease once it develops because of its complex metabolic disorders, so early prevention and treatment are important. However, currently available DKD therapies are not ideal, and novel therapeutic strategies are urgently needed. The potential of stem cell therapies partly depends on their ability to secrete exosomes. More and more studies have shown that stem cell-derived exosomes take part in the DKD pathophysiological process, which may offer an effective therapy for DKD treatment. Herein, we mainly review potential therapies of stem cell-derived exosomes mainly stem cell-derived exosomal microRNAs in DKD, including their protective effects on mesangial cells, podocytes and renal tubular epithelial cells. Using this secretome as possible therapeutic drugs without potential carcinogenicity should be the focus of further research.
Collapse
Affiliation(s)
- Han Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jiajia Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Tiejun Liu
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yan Leng
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
8
|
TNF-α Plus IL-1β Induces Opposite Regulation of Cx43 Hemichannels and Gap Junctions in Mesangial Cells through a RhoA/ROCK-Dependent Pathway. Int J Mol Sci 2022; 23:ijms231710097. [PMID: 36077498 PMCID: PMC9456118 DOI: 10.3390/ijms231710097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Connexin 43 (Cx43) is expressed in kidney tissue where it forms hemichannels and gap junction channels. However, the possible functional relationship between these membrane channels and their role in damaged renal cells remains unknown. Here, analysis of ethidium uptake and thiobarbituric acid reactive species revealed that treatment with TNF-α plus IL-1β increases Cx43 hemichannel activity and oxidative stress in MES-13 cells (a cell line derived from mesangial cells), and in primary mesangial cells. The latter was also accompanied by a reduction in gap junctional communication, whereas Western blotting assays showed a progressive increase in phosphorylated MYPT (a target of RhoA/ROCK) and Cx43 upon TNF-α/IL-1β treatment. Additionally, inhibition of RhoA/ROCK strongly antagonized the TNF-α/IL-1β-induced activation of Cx43 hemichannels and reduction in gap junctional coupling. We propose that activation of Cx43 hemichannels and inhibition of cell-cell coupling during pro-inflammatory conditions could contribute to oxidative stress and damage of mesangial cells via the RhoA/ROCK pathway.
Collapse
|
9
|
Rachubik P, Szrejder M, Rogacka D, Typiak M, Audzeyenka I, Kasztan M, Pollock DM, Angielski S, Piwkowska A. Insulin controls cytoskeleton reorganization and filtration barrier permeability via the PKGIα-Rac1-RhoA crosstalk in cultured rat podocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119301. [PMID: 35642843 DOI: 10.1016/j.bbamcr.2022.119301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Podocyte foot processes are an important cellular layer of the glomerular barrier that regulates glomerular permeability. Insulin via the protein kinase G type Iα (PKGIα) signaling pathway regulates the balance between contractility and relaxation (permeability) of the podocyte barrier by regulation of the actin cytoskeleton. This mechanism was shown to be disrupted in diabetes. Rho family guanosine-5'-triphosphates (GTPases) are dynamic modulators of the actin cytoskeleton and expressed in cells that form the glomerular filtration barrier. Thus, changes in Rho GTPase activity may affect glomerular permeability to albumin. The present study showed that Rho family GTPases control podocyte migration and permeability. Moreover these processes are regulated by insulin in PKGIα-dependent manner. Modulation of the PKGI-dependent activity of Rac1 and RhoA GTPases with inhibitors or small-interfering RNA impair glomerular permeability to albumin. We also demonstrated this mechanism in obese, insulin-resistant Zucker rats. We propose that PKGIα-Rac1-RhoA crosstalk is necessary in proper organization of the podocyte cytoskeleton and consequently the stabilization of glomerular architecture and regulation of filtration barrier permeability.
Collapse
Affiliation(s)
- Patrycja Rachubik
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Maria Szrejder
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Marlena Typiak
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Biology, Department of General and Medical Biochemistry, Gdańsk, Poland
| | - Irena Audzeyenka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland
| | - Małgorzata Kasztan
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefan Angielski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Department of Molecular Biotechnology, Gdańsk, Poland.
| |
Collapse
|
10
|
Fan X, Li X, Liu H, Xu F, Ji X, Chen Y, Li C. A ROCK1 Inhibitior Fasudil Alleviates Cardiomyocyte Apoptosis in Diabetic Cardiomyopathy by Inhibiting Mitochondrial Fission in a Type 2 Diabetes Mouse Model. Front Pharmacol 2022; 13:892643. [PMID: 35865967 PMCID: PMC9294374 DOI: 10.3389/fphar.2022.892643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes mellitus (DM) often involves cardiovascular complications; however, treatment regimens are limited. ROCK1 (rho-associated coiled-coil containing protein kinase 1) serves as a pathological factor in several diabetic complications. Herein, we aimed to explore the effect of Fasudil (a ROCK1 inhibitor) on the progress of cardiac dysfunction in type 2 DM (T2DM), and to explore the possible mechanisms. Type II diabetic mice models were established by inducing insulin resistance through a high-fat diet combined with low-dose streptozotocin (STZ) injection. NMCMs (neonatal mouse ventricular cardiac myocytes) in the control group were treated with 5.5 mM glucose, while those in the High Glucose (HG) group were treated with 33 mM glucose and 10 nmol/L insulin. In vivo, we found that type 2 diabetes enhanced the expression and activation of ROCK1 (p < 0.05). The ROCK1 inhibitor, Fasudil, prevented cardiac dysfunction, fibrosis, oxidative stress and myocardial ultrastructural disorders (p < 0.05) in the diabetic mice. In vitro, ROCK1 was upregulated in HG-induced cardiomyocytes, and ROCK1 inhibition using Fasudil reversed the increased apoptosis, consistent with in vivo results. Mechanistically, ROCK1 inhibition abrogated apoptosis, relieved mitochondrial fission, and efficiently attenuated the escalated production of reactive oxygen species in vitro and in vivo. The content of Ser616-phosphorylated dynamin-related protein 1 (Drp1) increased while ROCK1 led to apoptosis in HG-treated cardiomyocytes, which could be partly neutralized by ROCK1 inhibition with Fasudil, consistent with the in vivo results. Fasudil attenuated the cardiac dysfunction in diabetes by decreasing excessive mitochondrial fission via inhibiting Drp1 phosphorylation at serine 616.
Collapse
Affiliation(s)
- Xinhui Fan
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaoxing Li
- Department of Geriatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Huiruo Liu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaoping Ji
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Yuguo Chen, ; Chuanbao Li,
| | - Chuanbao Li
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Yuguo Chen, ; Chuanbao Li,
| |
Collapse
|
11
|
Paunas FTI, Finne K, Leh S, Marti HP, Berven F, Vikse BE. Proteomic signature of tubulointerstitial tissue predicts prognosis in IgAN. BMC Nephrol 2022; 23:118. [PMID: 35331167 PMCID: PMC8943973 DOI: 10.1186/s12882-022-02736-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND IgA nephropathy (IgAN) is associated with a significant risk of progression to kidney failure. Tubular atrophy is an established important risk factor for progressive disease, but few studies have investigated tubulointerstitial molecular markers and mechanisms of progression in IgAN. METHODS Based on data from the Norwegian Renal Registry, two groups were included: IgAN patients with (n = 9) or without (n = 18) progression to kidney failure during 10 years of follow-up. Tubulointerstitial tissue without discernible interstitial expansion or pronounced tubular alterations was microdissected, proteome was analysed using tandem mass spectrometry and relative protein abundances were compared between groups. RESULTS Proteome analyses quantified 2562 proteins with at least 2 unique peptides. Of these, 150 proteins had significantly different abundance between progressive and non-progressive IgAN patients, 67 were more abundant and 83 less abundant. Periostin was the protein with the highest fold change between progressive and non-progressive IgAN (fold change 8.75, p < 0.05) and periostin staining was also stronger in patients with progressive vs non-progressive IgAN. Reactome pathway analyses showed that proteins related to inflammation were more abundant and proteins involved in mitochondrial translation were significantly less abundant in progressive vs non-progressive patients. CONCLUSIONS Microdissection of tubulointerstitial tissue with only mild damage allowed for identification of proteome markers of early progressive IgAN. Periostin abundance showed promise as a novel and important risk marker of progression.
Collapse
Affiliation(s)
- Flavia Teodora Ioana Paunas
- Department of Medicine, Haugesund Hospital, Haugesund, Norway. .,Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Kenneth Finne
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Sabine Leh
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Frode Berven
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Bjørn Egil Vikse
- Department of Medicine, Haugesund Hospital, Haugesund, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
12
|
Freitas F, Attwell D. Pericyte-mediated constriction of renal capillaries evokes no-reflow and kidney injury following ischaemia. eLife 2022; 11:74211. [PMID: 35285797 PMCID: PMC8947765 DOI: 10.7554/elife.74211] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury is common, with ~13 million cases and 1.7 million deaths/year worldwide. A major cause is renal ischaemia, typically following cardiac surgery, renal transplant or severe haemorrhage. We examined the cause of the sustained reduction in renal blood flow ('no-reflow'), which exacerbates kidney injury even after an initial cause of compromised blood supply is removed. Adult male Sprague-Dawley rats, or NG2-dsRed male mice were used in this study. After 60 min kidney ischaemia and 30-60 min reperfusion, renal blood flow remained reduced, especially in the medulla, and kidney tubule damage was detected as Kim-1 expression. Constriction of the medullary descending vasa recta and cortical peritubular capillaries occurred near pericyte somata, and led to capillary blockages, yet glomerular arterioles and perfusion were unaffected, implying that the long-lasting decrease of renal blood flow contributing to kidney damage was generated by pericytes. Blocking Rho kinase to decrease pericyte contractility from the start of reperfusion increased the post-ischaemic diameter of the descending vasa recta capillaries at pericytes, reduced the percentage of capillaries that remained blocked, increased medullary blood flow and reduced kidney injury. Thus, post-ischaemic renal no-reflow, contributing to acute kidney injury, reflects pericytes constricting the descending vasa recta and peritubular capillaries. Pericytes are therefore an important therapeutic target for treating acute kidney injury.
Collapse
Affiliation(s)
- Felipe Freitas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
13
|
Wang F, Fan J, Pei T, He Z, Zhang J, Ju L, Han Z, Wang M, Xiao W. Effects of Shenkang Pills on Early-Stage Diabetic Nephropathy in db/db Mice via Inhibiting AURKB/RacGAP1/RhoA Signaling Pathway. Front Pharmacol 2022; 13:781806. [PMID: 35222021 PMCID: PMC8873791 DOI: 10.3389/fphar.2022.781806] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/17/2022] [Indexed: 12/22/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease, so there is an urgent need to suppress its development at early stage. Shenkang pills (SKP) are a hospital prescription selected and optimized from effective traditional Chinese medicinal formulas for clinical treatment of DN. In the present study, liquid chromatography-quadrupole-time of flight-mass spectrometry (LC-Q-TOF-MS) and total contents qualification were applied to generate a quality control standard of SKP. For verifying the therapeutic effects of SKP, db/db mice were administered intragastrically with SKP at a human-equivalent dose (1.82 g/kg) for 4 weeks. Moreover, the underlying mechanism of SKP were analyzed by the renal RNA sequencing and network pharmacology. LC-Q-TOF-MS identified 46 compounds in SKP. The total polysaccharide and organic acid content in SKP were 4.60 and 0.11 mg/ml, respectively, while the total flavonoid, saponin, and protein content were 0.25, 0.31, and 0.42 mg/ml, respectively. Treatment of SKP significantly reduced fasting blood glucose, improved renal function, and ameliorated glomerulosclerosis and focal foot processes effacement in db/db mice. In addition, SKP protected podocytes from injury by increasing nephrin and podocin expression. Furthermore, transcriptome analyses revealed that 430 and 288 genes were up and down-regulated in mice treated with SKP, relative to untreated controls. Gene ontology enrichment analysis revealed that the differentially expressed genes mainly involved in modulation of cell division and chromosome segregation. Weighted gene co-expression network analysis and network pharmacology analysis indicated that aurora kinase B (AURKB), Rac GTPase activating protein 1 (RacGAP1) and SHC binding, and spindle associated 1 (shcbp1) might be the core targets of SKP. This protein and Ras homolog family member A (RhoA) were found overexpression in db/db mice, but significantly decreased with SKP treatment. We conclude that SKP can effectively treat early-stage DN and improve renal podocyte dysfunction. The mechanism may involve down-regulation of the AURKB/RacGAP1/RhoA pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wei Xiao
- *Correspondence: Mingqing Wang, ; Wei Xiao,
| |
Collapse
|
14
|
Steichen C, Hervé C, Hauet T, Bourmeyster N. Rho GTPases in kidney physiology and diseases. Small GTPases 2022; 13:141-161. [PMID: 34138686 PMCID: PMC9707548 DOI: 10.1080/21541248.2021.1932402] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Rho family GTPases are molecular switches best known for their pivotal role in dynamic regulation of the actin cytoskeleton, but also of cellular morphology, motility, adhesion and proliferation. The prototypic members of this family (RhoA, Rac1 and Cdc42) also contribute to the normal kidney function and play important roles in the structure and function of various kidney cells including tubular epithelial cells, mesangial cells and podocytes. The kidney's vital filtration function depends on the structural integrity of the glomerulus, the proximal portion of the nephron. Within the glomerulus, the architecturally actin-based cytoskeleton podocyte forms the final cellular barrier to filtration. The glomerulus appears as a highly dynamic signalling hub that is capable of integrating intracellular cues from its individual structural components. Dynamic regulation of the podocyte cytoskeleton is required for efficient barrier function of the kidney. As master regulators of actin cytoskeletal dynamics, Rho GTPases are therefore of critical importance for sustained kidney barrier function. Dysregulated activities of the Rho GTPases and of their effectors are implicated in the pathogenesis of both hereditary and idiopathic forms of kidney diseases. Diabetic nephropathy is a progressive kidney disease that is caused by injury to kidney glomeruli. High glucose activates RhoA/Rho-kinase in mesangial cells, leading to excessive extracellular matrix production (glomerulosclerosis). This RhoA/Rho-kinase pathway also seems involved in the post-transplant hypertension frequently observed during treatment with calcineurin inhibitors, whereas Rac1 activation was observed in post-transplant ischaemic acute kidney injury.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
| | | | - Thierry Hauet
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
| | - Nicolas Bourmeyster
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
- Laboratoire STIM CNRS ERL 7003, Université de Poitiers, Poitiers Cédex, France
| |
Collapse
|
15
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
16
|
Ricciardi CA, Gnudi L. Kidney disease in diabetes: From mechanisms to clinical presentation and treatment strategies. Metabolism 2021; 124:154890. [PMID: 34560098 DOI: 10.1016/j.metabol.2021.154890] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
Metabolic and haemodynamic perturbations and their interaction drive the development of diabetic kidney disease (DKD) and its progression towards end stage renal disease (ESRD). Increased mitochondrial oxidative stress has been proposed as the central mechanism in the pathophysiology of DKD, but other mechanisms have been implicated. In parallel to increased oxidative stress, inflammation, cell apoptosis and tissue fibrosis drive the relentless progressive loss of kidney function affecting both the glomerular filtration barrier and the renal tubulointerstitium. Alteration of glomerular capillary autoregulation is at the basis of glomerular hypertension, an important pathogenetic mechanism for DKD. Clinical presentation of DKD can vary. Its classical presentation, often seen in patients with type 1 diabetes (T1DM), features hyperfiltration and albuminuria followed by progressive fall in renal function. Patients can often also present with atypical features characterised by progressive reduction in renal function without albuminuria, others in conjunction with non-diabetes related pathologies making the diagnosis, at times, challenging. Metabolic, lipid and blood pressure control with lifestyle interventions are crucial in reducing the progressive renal function decline seen in DKD. The prevention and management of DKD (and parallel cardiovascular disease) is a huge global challenge and therapies that target haemodynamic perturbations, such as inhibitors of the renin-angiotensin-aldosterone system (RAAS) and SGLT2 inhibitors, have been most successful.
Collapse
Affiliation(s)
| | - Luigi Gnudi
- School of Cardiovascular Medicine & Science, King's College London, London, UK.
| |
Collapse
|
17
|
Shu A, Du Q, Chen J, Gao Y, Zhu Y, Lv G, Lu J, Chen Y, Xu H. Catalpol ameliorates endothelial dysfunction and inflammation in diabetic nephropathy via suppression of RAGE/RhoA/ROCK signaling pathway. Chem Biol Interact 2021; 348:109625. [PMID: 34416245 DOI: 10.1016/j.cbi.2021.109625] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023]
Abstract
Catalpol is an iridoid glycoside compound isolated from the root of Rehmannia glutinosa, which has been reported to be a promising candidate for the treatment of diabetic diseases. The present study aimed at investigating the effects and potential mechanism of catalpol on endothelial dysfunction and inflammation in diabetic nephropathy (DN). We constructed DN mice and advanced glycation end products (AGEs)-induced mouse glomerular endothelial cells (mGECs) injury model. The results demonstrated that catalpol effectively improved renal pathology and decreased levels of urine protein, serum creatinine, and blood urea nitrogen in DN mice. Catalpol significantly reduced endothelial dysfunction and inflammatory infiltration of macrophages in DN mice and AGEs-induced mGECs. To further study the protective mechanism of catalpol, we transfected DN mice with recombinant adeno-associated virus expressing receptor of AGEs (RAGE) and intervened AGEs-induced mGECs with inhibitors. Catalpol reversed endothelial dysfunction and inflammation aggravated by RAGE overexpression in DN mice. Meanwhile, catalpol significantly inhibited the RAGE/Ras homolog gene family member A (RhoA)/Rho-associated kinase (ROCK) pathway in DN mice with RAGE overexpression. Moreover, the combination of catalpol with inhibitors of RAGE, RhoA and ROCK exerted stronger anti-endothelial dysfunction and anti-macrophage infiltration effects on AGEs-induced mGECs compared with catalpol alone. In short, this study indicated that catalpol could ameliorate endothelial dysfunction and inflammation via suppression of RAGE/RhoA/ROCK pathway, hereby delaying the progression of DN.
Collapse
Affiliation(s)
- Anmei Shu
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, 224005, China; Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Efficacy and Safety Evaluation of Traditional Chinese Medicine in Jiangsu Province, Nanjing, 210023, China.
| | - Qiu Du
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacy, Nanjing Hospital of Chinese Medicine Affiliate of Chinese Medicine, Nanjing, 210022, China.
| | - Jing Chen
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Efficacy and Safety Evaluation of Traditional Chinese Medicine in Jiangsu Province, Nanjing, 210023, China.
| | - Yuyan Gao
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Efficacy and Safety Evaluation of Traditional Chinese Medicine in Jiangsu Province, Nanjing, 210023, China.
| | - Yihui Zhu
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Efficacy and Safety Evaluation of Traditional Chinese Medicine in Jiangsu Province, Nanjing, 210023, China.
| | - Gaohong Lv
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jinfu Lu
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yuping Chen
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, 224005, China.
| | - Huiqin Xu
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Key Laboratory for Efficacy and Safety Evaluation of Traditional Chinese Medicine in Jiangsu Province, Nanjing, 210023, China.
| |
Collapse
|
18
|
Matoba K, Sekiguchi K, Nagai Y, Takeda Y, Takahashi H, Yokota T, Utsunomiya K, Nishimura R. Renal ROCK Activation and Its Pharmacological Inhibition in Patients With Diabetes. Front Pharmacol 2021; 12:738121. [PMID: 34557101 PMCID: PMC8454778 DOI: 10.3389/fphar.2021.738121] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine/threonine kinase with essential roles in cytoskeletal functions. Substantial evidence implicates ROCK as a critical regulator in the inception and progression of diabetic nephropathy through a mechanism involving mesangial fibrosis, podocyte apoptosis, and endothelial inflammation. Despite these experimental observations, human data is lacking. Here we show that the phosphorylated form of myosin phosphatase targeting subunit 1 (MYPT1), a ROCK substrate, was increased in both the glomerular and tubulointerstitial areas in patients with histologically confirmed diabetic nephropathy. We also conducted a retrospective pilot analysis of data from patients with diabetes to assess the renoprotective effects of fasudil, an ATP-competitive ROCK inhibitor licensed in Japan for the prevention of vasospasm following subarachnoid hemorrhage. Fifteen subjects (male, n = 8; female, n = 7; age 65.7 ± 14.7 years; body height, 161.1 ± 12.6 cm; body weight, 57.6 ± 13.7 kg; body mass index, 22.4 ± 3.7 kg/m2) were enrolled to evaluate blood pressure and the renal outcome after fasudil treatment. Of note, proteinuria was significantly reduced at the end of the fasudil treatment without affecting the blood pressure or estimated glomerular filtration rate. Taken together, these findings suggest that the administration of fasudil could be associated with a better renal outcome by inhibiting the ROCK activity in patients with diabetes.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kensuke Sekiguchi
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Takahashi
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tamotsu Yokota
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Department of Internal Medicine, Division of Diabetes, Metabolism, and Endocrinology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Gonzalez CD, Carro Negueruela MP, Nicora Santamarina C, Resnik R, Vaccaro MI. Autophagy Dysregulation in Diabetic Kidney Disease: From Pathophysiology to Pharmacological Interventions. Cells 2021; 10:2497. [PMID: 34572148 PMCID: PMC8469825 DOI: 10.3390/cells10092497] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 12/18/2022] Open
Abstract
Diabetic kidney disease (DKD) is a frequent, potentially devastating complication of diabetes mellitus. Several factors are involved in its pathophysiology. At a cellular level, diabetic kidney disease is associated with many structural and functional alterations. Autophagy is a cellular mechanism that transports intracytoplasmic components to lysosomes to preserve cellular function and homeostasis. Autophagy integrity is essential for cell homeostasis, its alteration can drive to cell damage or death. Diabetic kidney disease is associated with profound autophagy dysregulation. Autophagy rate and flux alterations were described in several models of diabetic kidney disease. Some of them are closely linked with disease progression and severity. Some antidiabetic agents have shown significant effects on autophagy. A few of them have also demonstrated to modify disease progression and improved outcomes in affected patients. Other drugs also target autophagy and are being explored for clinical use in patients with diabetic kidney disease. The modulation of autophagy could be relevant for the pharmacological treatment and prevention of this disease in the future. Therefore, this is an evolving area that requires further experimental and clinical research. Here we discuss the relationship between autophagy and Diabetic kidney disease and the potential value of autophagy modulation as a target for pharmacological intervention.
Collapse
Affiliation(s)
- Claudio D. Gonzalez
- Instituto de Bioquimica y Medicina Molecular Prof. Alberto Boveris (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina; (C.D.G.); (R.R.)
- Instituto Universitario del Centro de Educacion Medica e Investigacion Clinica (IUC-CEMIC-CONICET), Facultad de Medicina, Instituto Universitario CEMIC, Buenos Aires C1430 EFA, Argentina; (M.P.C.N.); (C.N.S.)
| | - María Paula Carro Negueruela
- Instituto Universitario del Centro de Educacion Medica e Investigacion Clinica (IUC-CEMIC-CONICET), Facultad de Medicina, Instituto Universitario CEMIC, Buenos Aires C1430 EFA, Argentina; (M.P.C.N.); (C.N.S.)
| | - Catalina Nicora Santamarina
- Instituto Universitario del Centro de Educacion Medica e Investigacion Clinica (IUC-CEMIC-CONICET), Facultad de Medicina, Instituto Universitario CEMIC, Buenos Aires C1430 EFA, Argentina; (M.P.C.N.); (C.N.S.)
| | - Roxana Resnik
- Instituto de Bioquimica y Medicina Molecular Prof. Alberto Boveris (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina; (C.D.G.); (R.R.)
| | - Maria I. Vaccaro
- Instituto de Bioquimica y Medicina Molecular Prof. Alberto Boveris (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina; (C.D.G.); (R.R.)
- Instituto Universitario del Centro de Educacion Medica e Investigacion Clinica (IUC-CEMIC-CONICET), Facultad de Medicina, Instituto Universitario CEMIC, Buenos Aires C1430 EFA, Argentina; (M.P.C.N.); (C.N.S.)
| |
Collapse
|
20
|
Sharma S, Brown CE. Microvascular basis of cognitive impairment in type 1 diabetes. Pharmacol Ther 2021; 229:107929. [PMID: 34171341 DOI: 10.1016/j.pharmthera.2021.107929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/23/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
The complex computations of the brain require a constant supply of blood flow to meet its immense metabolic needs. Perturbations in blood supply, even in the smallest vascular networks, can have a profound effect on neuronal function and cognition. Type 1 diabetes is a prevalent and insidious metabolic disorder that progressively and heterogeneously disrupts vascular signalling and function in the brain. As a result, it is associated with an array of adverse vascular changes such as impaired regulation of vascular tone, pathological neovascularization and vasoregression, capillary plugging and blood brain barrier disruption. In this review, we highlight the link between microvascular dysfunction and cognitive impairment that is commonly associated with type 1 diabetes, with the aim of synthesizing current knowledge in this field.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
21
|
Chen L, Wu J, Hu B, Liu C, Wang H. The Role of Cell Division Autoantigen 1 (CDA1) in Renal Fibrosis of Diabetic Nephropathy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6651075. [PMID: 33997036 PMCID: PMC8102118 DOI: 10.1155/2021/6651075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023]
Abstract
The common kidney disease diabetic nephropathy (DN) accounts for significant morbidity and mortality in patients with diabetes, and its effective diagnosis in incipient stages is still lacking. Renal fibrosis is the main pathological feature of DN. Cell division autoantigen 1 (CDA1), a phosphorylated protein encoded by TSPYL2 on the X chromosome, plays a fibrogenic role by modulating the transforming growth factor-β (TGF-β) signaling, but the exact mechanism remains unclear. TGF-β signaling has been recognized as the key factor in promoting the development and progression of DN. At present, strict control of blood sugar and blood pressure can significantly lower the development and progression of DN in the early stages, and many studies have shown that blocking TGF-β signaling can delay the progress of DN. However, TGF-β is a multifunctional cytokine. Its direct intervention may result in increased side effects. Therefore, the targeted intervention of CDA1 not only can block the TGF-β signaling pathway but also can reduce these side effects. In this article, we review the main physiological roles of CDA1, with particular attention to its effect and potential mechanism in the renal fibrosis of DN.
Collapse
Affiliation(s)
- LinLin Chen
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Jiao Wu
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
| | - Bin Hu
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
| | - Changbai Liu
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Hu Wang
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Bai Y, Du Q, Zhang L, Li L, Tang L, Zhang W, Du R, Li P, Li L. Fasudil alleviated insulin resistance through promotion of proliferation, attenuation of cell apoptosis and inflammation and regulation of RhoA/Rho kinase/insulin/nuclear factor-κB signalling pathway in HTR-8/SVneo cells. J Pharm Pharmacol 2021; 73:1118-1127. [PMID: 33779714 DOI: 10.1093/jpp/rgab033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/10/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate the effects of fasudil on insulin resistance (IR) in HTR-8/SVneo cells. METHODS HTR-8/SVneo cells were treated with insulin or/and fasudil. Cell proliferation, apoptosis, inflammation and related signalling pathways were assessed. KEY FINDINGS Insulin treatment significantly enhanced the protein expressions of RhoA and Rho kinase (ROCK1 and ROCK2), but decreased glucose consumption. Administration of fasudil effectively promoted glucose uptake. Moreover, fasudil enhanced cell viability and the level of proliferating cell nuclear antigen (PCNA). Insulin-mediated cell apoptosis was inhibited by fasudil via the down-regulation of bax and cleaved-caspase-3, and the up-regulation of bcl-2. At the same time, fasudil led to the reduction of IL-1β, TNF-α, IL-6 and IL-8 mRNA levels in insulin-treated cells. In addition, RhoA, ROCK2 and phosphorylated myosin phosphatase target subunit-1 (p-MYPT-1) expressions were down-regulated by fasudil. Importantly, fasudil activated insulin receptor substrate-1 (IRS-1) through increasing p-IRS-1 (Tyr612) and p-Akt expressions. The nuclear NF-κB p65 and p-IκB-α levels were reduced via the administration of fasudil in insulin-treated cells. CONCLUSIONS Fasudil mitigated IR by the promotion of cell proliferation, inhibition of apoptosis and inflammation and regulation of RhoA/ROCK/insulin/NF-κB signalling pathway through in vitro studies.
Collapse
Affiliation(s)
- Yu Bai
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Qiang Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Le Zhang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Ling Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Lei Tang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Wei Zhang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Runyu Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Ping Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Ling Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
23
|
Activin A and Cell-Surface GRP78 Are Novel Targetable RhoA Activators for Diabetic Kidney Disease. Int J Mol Sci 2021; 22:ijms22062839. [PMID: 33799579 PMCID: PMC8000060 DOI: 10.3390/ijms22062839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of kidney failure. RhoA/Rho-associated protein kinase (ROCK) signaling is a recognized mediator of its pathogenesis, largely through mediating the profibrotic response. While RhoA activation is not feasible due to the central role it plays in normal physiology, ROCK inhibition has been found to be effective in attenuating DKD in preclinical models. However, this has not been evaluated in clinical studies as of yet. Alternate means of inhibiting RhoA/ROCK signaling involve the identification of disease-specific activators. This report presents evidence showing the activation of RhoA/ROCK signaling both in vitro in glomerular mesangial cells and in vivo in diabetic kidneys by two recently described novel pathogenic mediators of fibrosis in DKD, activins and cell-surface GRP78. Neither are present in normal kidneys. Activin inhibition with follistatin and neutralization of cell-surface GRP78 using a specific antibody blocked RhoA activation in mesangial cells and in diabetic kidneys. These data identify two novel RhoA/ROCK activators in diabetic kidneys that can be evaluated for their efficacy in inhibiting the progression of DKD.
Collapse
|
24
|
Huang C, Zhou Y, Huang H, Zheng Y, Kong L, Zhang H, Zhang Y, Wang H, Yang M, Xu X, Chen B. Islet Transplantation Reverses Podocyte Injury in Diabetic Nephropathy or Induced by High Glucose via Inhibiting RhoA/ROCK/NF- κB Signaling Pathway. J Diabetes Res 2021; 2021:9570405. [PMID: 33778085 PMCID: PMC7969114 DOI: 10.1155/2021/9570405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/26/2020] [Accepted: 12/21/2020] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Abnormal signaling pathways play a crucial role in the mechanisms of podocyte injury in diabetic nephropathy. They also affect the recovery of podocytes after islet transplantation (IT). However, the specific signaling abnormalities that affect the therapeutic effect of IT on podocytes remains unclear. The purpose of this study was to assess whether the RhoA/ROCK/NF-κB signaling pathway is related to podocyte restoration after IT. METHODS A mouse model of diabetic nephropathy was established in vivo using streptozotocin. The mice were then subsequently reared for 4 weeks after islet transplantation to determine the effect of IT. Islet cells, CCG-1423 (RhoA Inhibitor), and fasudil (ROCK inhibitor) were then cocultured with podocytes in vitro to assess their protective effects on podocyte injury induced by high glucose (HG). Protein expression levels of RhoA, ROCK1, synaptopodin, IL-6, and MCP-1 in kidney tissues were then measured using immunohistochemistry and Western blotting techniques. RESULTS Islet transplantation reduced the expression levels of RhoA/ROCK1 and that of related inflammatory factors such as IL-6 and MCP-1 in the kidney podocytes of diabetic nephropathy. In the same line, islet cells reduced the expression of RhoA, ROCK1, and pp65 in immortalized podocytes under high glucose (35.0 mmol/L glucose) conditions. CONCLUSIONS Islet transplantation can reverse podocyte injury in diabetes nephropathy by inhibiting the RhoA/ROCK1 signaling pathway. Islet cells have a strong protective effect on podocytes treated with high glucose (35.0 mmol/L glucose). Discovery of signaling pathways affecting podocyte recovery is helpful for individualized efficacy evaluation and targeted therapy of islet transplantation patients.
Collapse
Affiliation(s)
- Chongchu Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yi Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Hongjian Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yushu Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Lijun Kong
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Hewei Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yan Zhang
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical University, 325015 Wenzhou, Zhejiang Province, China
| | - Hongwei Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Mei Yang
- Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, 325015 Wenzhou, Zhejiang Province, China
| | - Xiaona Xu
- Operating Room, The First Affiliated Hospital of Wenzhou Medical University, 325015 Wenzhou, Zhejiang Province, China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| |
Collapse
|
25
|
Proteomics Analysis of Gastric Cancer Patients with Diabetes Mellitus. J Clin Med 2021; 10:jcm10030407. [PMID: 33494396 PMCID: PMC7866049 DOI: 10.3390/jcm10030407] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022] Open
Abstract
Proteomics is a powerful approach to study the molecular mechanisms of cancer. In this study, we aim to characterize the proteomic profile of gastric cancer (GC) in patients with diabetes mellitus (DM) type 2. Forty GC tissue samples including 19 cases from diabetic patients and 21 cases from individuals without diabetes (control group) were selected for the proteomics analysis. Gastric tissues were processed following the single-pot, solid-phase-enhanced sample preparation approach-SP3 and enzymatic digestion with trypsin. The resulting peptides were analyzed by LC-MS Liquid Chromatography-Mass Spectrometry (LC-MS). The comparison of protein expression levels between GC samples from diabetic and non-diabetic patients was performed by label-free quantification (LFQ). A total of 6599 protein groups were identified in the 40 samples. Thirty-seven proteins were differentially expressed among the two groups, with 16 upregulated and 21 downregulated in the diabetic cohort. Statistical overrepresentation tests were considered for different annotation sets including the Gene Ontology(GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome, and Disease functional databases. Upregulated proteins in the GC samples from diabetic patients were particularly enriched in respiratory electron transport and alcohol metabolic biological processes, while downregulated proteins were associated with epithelial cancers, intestinal diseases, and cell-cell junction cellular components. Taken together, these results support the data already obtained by previous studies that associate diabetes with metabolic disorders and diabetes-associated diseases, such as Alzheimer's and Parkinson's, and also provide valuable insights into seven GC-associated protein targets, claudin-3, polymeric immunoglobulin receptor protein, cadherin-17, villin-1, transglutaminase-2, desmoglein-2, and mucin-13, which warrant further investigation.
Collapse
|
26
|
Hudlikar RR, Sargsyan D, Li W, Wu R, Zheng M, Kong AN. Epigenomic, Transcriptomic, and Protective Effect of Carotenoid Fucoxanthin in High Glucose-Induced Oxidative Stress in Mes13 Kidney Mesangial Cells. Chem Res Toxicol 2021; 34:713-722. [PMID: 33448797 DOI: 10.1021/acs.chemrestox.0c00235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic nephropathy (DN) is the major cause of kidney related diseases in patients induced by high glucose (HG) affecting around 40% of type 1 and 2 diabetic patients. It is characterized by excessive inflammation inducing factors, reactive oxygen species (ROS) overproduction, and potential epigenomic related changes. Fucoxanthin (FX), a carotenoid found in brown seaweed, has a structure which includes an allenic bond and a 5,6-monoepoxide in the molecule, with strong antioxidant and anti-inflammatory activity. However, understanding of the impact of FX on DN was lacking. In this study we tested the early effects of high glucose (HG) on mouse mesangial kidney Mes13 cells, a potential in vitro cell culture model of DN. Our results show that HG induced oxidative stress on kidney mesangial Mes13 cells, while FX treatment attenuates the oxidative stress by decreasing the ROS, demonstrated by flow cytometry. Next, we utilized next-generation sequencing (NGS) to profile the HG-induced early epigenomic and transcriptomic changes in this in vitro DN model and the protective effects of FX. Differentially expressed genes (DEGs) and differentially methylated regions (DMRs) were analyzed using R software in HG and FX treated groups. Differential regulation of signaling pathways was studied using Reactome Pathway Analysis in the comparison. DEG analysis shows that novel biomarkers with specific pathways, including interleukin regulation, Toll-like receptor pathway, and PKA phosphorylation pathways, were found to be modulated by the FX treatment. TGF β 1i1 (TGFB 1i1), MAP-3-kinase-13(MAP3K13) involved in crucial cellular processes including glucose metabolism, phosphodiesterase regulation was methylated in HG, which was demethylated with FX treatment. Integrated transcriptomic and CpG methylome analysis of DEGs and DMRs revealed that genes like adenylate cyclase (Adcy7), calponin 1 (CNN1), potassium voltage-gated channel interacting protein 2 (KCNIP2), phosphatidylinositol-4-phosphate 5-kinase type 1 β (PIP5K1B), and transmembrane protein with EGF-like and two follistatin-like domains 1 (TMEFF1), which were modulated by FX in HG-exposed Mes13 cells, potentially modulate ion channel transport and glucose metabolism. In summary, our current study shows that novel early epigenomic and transcriptomic biomarkers were altered during the disease progression of HG-induced DN and that FX modified these alterations potentially contributing to the protective effects of mesangial cells from the HG-induced oxidative stress and damage.
Collapse
Affiliation(s)
- Rasika R Hudlikar
- Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Davit Sargsyan
- Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Wenji Li
- Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Renyi Wu
- Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Meinizi Zheng
- Center for Phytochemical Epigenome Studies, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | | |
Collapse
|
27
|
Zhang Z, Liang W, Luo Q, Hu H, Yang K, Hu J, Chen Z, Zhu J, Feng J, Zhu Z, Chi Q, Ding G. PFKP Activation Ameliorates Foot Process Fusion in Podocytes in Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2021; 12:797025. [PMID: 35095764 PMCID: PMC8794994 DOI: 10.3389/fendo.2021.797025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glycolysis dysfunction is an important pathogenesis of podocyte injury in diabetic kidney disease (DKD). Foot process fusion of podocytes and increased albuminuria are markers of early DKD. Moreover, cytoskeletal remodeling has been found to be involved in the foot process fusion of podocytes. However, the connections between cytoskeletal remodeling and alterations of glycolysis in podocytes in DKD have not been clarified. METHODS mRNA sequencing of glomeruli obtained from db/db and db/m mice with albuminuria was performed to analyze the expression profiling of genes in glucose metabolism. Expressions of phosphofructokinase platelet type (PFKP) in the glomeruli of DKD patients were detected. Clotrimazole (CTZ) was used to explore the renal effects of PFKP inhibition in diabetic mice. Using Pfkp siRNA or recombinant plasmid to manipulate PFKP expression, the effects of PFKP on high glucose (HG) induced podocyte damage were assessed in vitro. The levels of fructose-1,6-bisphosphate (FBP) were measured. Targeted metabolomics was performed to observe the alterations of the metabolites in glucose metabolism after HG stimulation. Furthermore, aldolase type b (Aldob) siRNA or recombinant plasmid were applied to evaluate the influence of FBP level alteration on podocytes. FBP was directly added to podocyte culture media. Db/db mice were treated with FBP to investigate its effects on their kidney. RESULTS mRNA sequencing showed that glycolysis enzyme genes were altered, characterized by upregulation of upstream genes (Hk1, and Pfkp) and down-regulation of downstream genes of glycolysis (Pkm, and Ldha). Moreover, the expression of PFKP was increased in glomeruli of DKD patients. The CTZ group presented more severe renal damage. In vitro, the Pfkp siRNA group and ALDOB overexpression group showed much more induced cytoskeletal remodeling in podocytes, while overexpression of PFKP and suppression of ALDOB in vitro rescued podocytes from cytoskeletal remodeling through regulation of FBP levels and inhibition of the RhoA/ROCK1 pathway. Furthermore, targeted metabolomics showed FBP level was significantly increased in HG group compared with the control group. Exogenous FBP addition reduced podocyte cytoskeletal remodeling and renal damage of db/db mice. CONCLUSIONS These findings provide evidence that PFKP may be a potential target for podocyte injury in DN and provide a rationale for applying podocyte glycolysis enhancing agents in patients with DKD.
Collapse
Affiliation(s)
- Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- *Correspondence: Wei Liang, ; Guohua Ding,
| | - Qiang Luo
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Keju Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Jili Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
| | - Qingjia Chi
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- *Correspondence: Wei Liang, ; Guohua Ding,
| |
Collapse
|
28
|
Identification of C3 as a therapeutic target for diabetic nephropathy by bioinformatics analysis. Sci Rep 2020; 10:13468. [PMID: 32778679 PMCID: PMC7417539 DOI: 10.1038/s41598-020-70540-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of diabetic nephropathy is not completely understood, and the effects of existing treatments are not satisfactory. Various public platforms already contain extensive data for deeper bioinformatics analysis. From the GSE30529 dataset based on diabetic nephropathy tubular samples, we identified 345 genes through differential expression analysis and weighted gene coexpression correlation network analysis. GO annotations mainly included neutrophil activation, regulation of immune effector process, positive regulation of cytokine production and neutrophil-mediated immunity. KEGG pathways mostly included phagosome, complement and coagulation cascades, cell adhesion molecules and the AGE-RAGE signalling pathway in diabetic complications. Additional datasets were analysed to understand the mechanisms of differential gene expression from an epigenetic perspective. Differentially expressed miRNAs were obtained to construct a miRNA-mRNA network from the miRNA profiles in the GSE57674 dataset. The miR-1237-3p/SH2B3, miR-1238-5p/ZNF652 and miR-766-3p/TGFBI axes may be involved in diabetic nephropathy. The methylation levels of the 345 genes were also tested based on the gene methylation profiles of the GSE121820 dataset. The top 20 hub genes in the PPI network were discerned using the CytoHubba tool. Correlation analysis with GFR showed that SYK, CXCL1, LYN, VWF, ANXA1, C3, HLA-E, RHOA, SERPING1, EGF and KNG1 may be involved in diabetic nephropathy. Eight small molecule compounds were identified as potential therapeutic drugs using Connectivity Map.
Collapse
|
29
|
Rogacka D, Audzeyenka I, Piwkowska A. Regulation of podocytes function by AMP-activated protein kinase. Arch Biochem Biophys 2020; 692:108541. [PMID: 32781053 DOI: 10.1016/j.abb.2020.108541] [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: 05/19/2020] [Revised: 07/23/2020] [Accepted: 08/06/2020] [Indexed: 01/08/2023]
Abstract
Podocytes are unique, highly specialized, terminally differentiated cells that form an essential, integral part of the glomerular filter. These cells limit the outside border of the glomerular basement membrane, forming a tight barrier that prevents significant protein loss from the capillary space. The slit diaphragm formed by podocytes is crucial for maintaining glomerular integrity and function. They are the target of injury in many glomerular diseases, including hypertension and diabetes mellitus. Accumulating studies have revealed that AMP-activated protein kinase (AMPK), an essential cellular energy sensor, might play a fundamental role in regulating podocyte metabolism and function. AMPK participates in insulin signaling, therefore controls glucose uptake and podocytes insulin sensitivity. It is also involved in insulin-dependent cytoskeleton reorganization in podocytes, mediating glomerular albumin permeability. AMPK plays an important role in the regulation of autophagy/apoptosis processes, which influence podocytes viability. The present review aimed to highlight the molecular mechanisms associated with AMPK that are involved in the regulation of podocyte function in health and disease states.
Collapse
Affiliation(s)
- Dorota Rogacka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Irena Audzeyenka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| |
Collapse
|
30
|
Liu S, Rong G, Li X, Geng L, Zeng Z, Jiang D, Yang J, Wei Y. Diosgenin and GSK126 Produce Synergistic Effects on Epithelial-Mesenchymal Transition in Gastric Cancer Cells by Mediating EZH2 via the Rho/ROCK Signaling Pathway. Onco Targets Ther 2020; 13:5057-5067. [PMID: 32606728 PMCID: PMC7292386 DOI: 10.2147/ott.s237474] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/03/2020] [Indexed: 12/25/2022] Open
Abstract
Background Diosgenin, a natural steroidal saponin isolated from Trigonella foenum-graecum, has been reported to exert anti-cancer effects. Inhibitors of enhancer of zeste homology 2 (EZH2) have been widely used in treatment of cancers. However, the effects of combined treatment with diosgenin and an EZH2 inhibitor on gastric cancer (GC) cells, and the mechanism for those effects are not fully understood. Methods AGS and SGC-7901 gastric cancer cells were treated with diosgenin (0 to 8 μM), followed by treatment with either diosgenin or an EZH2 inhibitor, GSK126 alone. Afterwards, an EZH2 overexpression plasmid and Rho inhibitor, GSK429286A was involved in cells. Cell proliferation, cell cycle distribution, and cell apoptosis, migration, and invasion were examined by CCK-8 assays, flow cytometry, and transwell assays. Western blotting was performed to detect the relative levels of protein expression. Results Treatment with diosgenin alone caused a dose-dependent decrease in the cell viability, and combined treatment with an EZH2 inhibitor plus GSK126 caused a further significant decrease. A further analysis revealed that treatment with either diosgenin or GSK126 alone induced significant increases in G0/G1 cell cycle arrest and apoptosis, and combined treatment with both agents induced further increases in those parameters. In addition, combined treatment with diosgenin and GSK126 synergistically induced even stronger effects on impaired cell proliferation, G0/G1 phase arrest, and cell apoptosis when compared to treatment with either diosgenin or GSK126 treatment alone. At the molecular level, we demonstrated that inhibition of Rho/ROCK signaling by combined treatment with diosgenin and GSK126 could downregulate the expression of epithelial–mesenchymal transition (EMT)-related molecules. We also found that EZH2 overexpression reversed the anti-tumor effect of diosgenin by inducing cell survival, blocking G1-phase arrest, and promoted EMT. While, these biological properties were further reversed by GSK429286A. Conclusion Collectively, combined treatment with diosgenin and GSK126 produced even more significant effects on GC cell inhibition by targeting EZH2 via Rho/ROCK signaling-mediated EMT, which might be a therapeutic strategy for improving the poor therapeutic outcomes obtained with GSK126 monotherapy.
Collapse
Affiliation(s)
- Shanshan Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Guihong Rong
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Xia Li
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Lijun Geng
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Zhineng Zeng
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Dongxiang Jiang
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Jun Yang
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Yesheng Wei
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| |
Collapse
|
31
|
Matoba K, Takeda Y, Nagai Y, Kanazawa Y, Kawanami D, Yokota T, Utsunomiya K, Nishimura R. ROCK Inhibition May Stop Diabetic Kidney Disease. JMA J 2020; 3:154-163. [PMID: 33150249 PMCID: PMC7590381 DOI: 10.31662/jmaj.2020-0014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease and is strongly associated with cardiovascular mortality. Given the pandemic of obesity and diabetes, the elucidation of the molecular underpinnings of DKD and establishment of effective therapy are urgently required. Studies over the past decade have identified the activated renin-angiotensin system (RAS) and hemodynamic changes as important therapeutic targets. However, given the residual risk observed in patients treated with RAS inhibitors and/or sodium glucose co-transporter 2 inhibitors, the involvement of other molecular machinery is likely, and the elucidation of such pathways represents fertile ground for the development of novel strategies. Rho-kinase (ROCK) is a serine/threonine kinase that is under the control of small GTPase protein Rho. Many fundamental cellular processes, including migration, proliferation, and survival are orchestrated by ROCK through a mechanism involving cytoskeletal reorganization. From a pathological standpoint, several analyses provide compelling evidence supporting the hypothesis that ROCK is an important regulator of DKD that is highly pertinent to cardiovascular disease. In cell-based studies, ROCK is activated in response to a diverse array of external stimuli associated with diabetes, and renal ROCK activity is elevated in the context of type 1 and 2 diabetes. Experimental studies have demonstrated the efficacy of pharmacological or genetic inhibition of ROCK in the prevention of diabetes-related histological and functional abnormalities in the kidney. Through a bird’s eye view of ROCK in renal biology, the present review provides a conceptual framework that may be widely applicable to the pathological processes of multiple organs and illustrate novel therapeutic promise in diabetology.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Zou HH, Wang L, Zheng XX, Xu GS, Shen Y. Endothelial cells secreted endothelin-1 augments diabetic nephropathy via inducing extracellular matrix accumulation of mesangial cells in ETBR -/- mice. Aging (Albany NY) 2020; 11:1804-1820. [PMID: 30926764 PMCID: PMC6461170 DOI: 10.18632/aging.101875] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/10/2019] [Indexed: 01/26/2023]
Abstract
Endothelin B receptor (ETBR) deficiency may contribute to the progression of diabetic nephropathy (DN) in a streptozotocin (STZ) model, but the underlying mechanism is not fully revealed. In this study, STZ-diabetic ETBR-/- mice was characterized by increased serum creatinine and urinary albumin, enhanced glomerulosclerosis, and upregulated ET-1 expression compared with STZ-diabetic WT mice. In vitro, HG conditioned media (CM) of ETBR-/- GENs promoted mesangial cell proliferation and upregulated ECM-related proteins, and ET-1 knockout in GENs or inhibition of ET-1/ETAR in mesangial cell suppressed mesangial cell proliferation and collagen IV formation. In addition, ET-1 was over-expressed in ETBR-/- GENs and was regulated by NF-kapapB pathway. ET-1/ETBR suppressed NF-kappaB to modulate ET-1 in GENs. Furthermore, ET-1/ETAR promoted RhoA/ROCK pathway in mesangial cells, and accelerated mesangial cell proliferation and ECM accumulation. Finally, in vivo experiments proved inhibition of NF-kappaB pathway ameliorated DN in ETBR-/- mice. These results suggest that in HG-exposed ETBR-/- GENs, suppression of ET-1 binding to ETBR activated NF-kappaB pathway, thus to secrete large amount of ET-1. Due to the communication between GENs and mesangial cells in diabetes, ET-1 binding to ETAR in mesangial cell promoted RhoA/ROCK pathway, thus to accelerate mesangial cell proliferation and ECM accumulation.
Collapse
Affiliation(s)
- Hong-Hong Zou
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Wang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Xu Zheng
- Division of Renal Diseases and Hypertension, Department of Medicine, The George Washington University, Washington, DC 20052, USA
| | - Gao-Si Xu
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
33
|
Sharma D, Kumar Tekade R, Kalia K. Kaempferol in ameliorating diabetes-induced fibrosis and renal damage: An in vitro and in vivo study in diabetic nephropathy mice model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153235. [PMID: 32563017 DOI: 10.1016/j.phymed.2020.153235] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/03/2020] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Kaempferol is a natural polyflavonol that has gained considerable attention as antidiabetic therapeutics. Recent reports emphasize the role of hyperglycemia and RhoA/Rho Kinase activity in the pathogenesis of diabetic nephropathy (DN). This study aims to evaluate the GLP-1 and insulin release along with RhoA/Rho Kinase inhibition pertaining to the anti-fibrotic and reno-protective effects of Kaempferol in DN. METHODS The effect of Kaempferol on GLP-1 and insulin release along with underlying mechanisms (Ca2+ and cAMP levels) in GLUTag and MIN6 cells as well as in their co-culture has been evaluated. Further, the effect of Kaempferol on GLP-1 and insulin release was evaluated under in-vivo circumstances in the DN C57BL/6 mouse model. Histology and fibrosis specific staining was performed to study the renal injuries and fibrosis, while the expression of mRNA and protein of interest was evaluated by RT-PCR and western blot analysis. RESULTS Kaempferol treatment promoted the GLP-1 and insulin release, which was accompanied by increased intracellular levels of cAMP and Ca2+ in GLUTag and MIN6 cells. In agreement with in vitro studies, Kaempferol also increased the release of GLP-1 and insulin in the DN mouse model. Notably, Kaempferol showed the potential to ameliorate the histological changes as well as renal fibrosis while decreasing the expression levels of DN markers including TGF-β1, CTGF, fibronectin, collagen IV, IL-1β, RhoA, ROCK2, and p-MYPT1 in DN kidney tissues. A rise in the expression of E-cadherin and nephrin was also noted in the same study. CONCLUSION This study establishes that Kaempferol ameliorates renal injury and fibrosis by enhancing the release of GLP-1, insulin, and inhibition of RhoA/Rho Kinase. This study recommends Kaempferol for further clinical trials to be developed as novel therapeutics for improving the renal function in DN patients.
Collapse
Affiliation(s)
- Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Rakesh Kumar Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
34
|
Kravets I, Mallipattu SK. The Role of Podocytes and Podocyte-Associated Biomarkers in Diagnosis and Treatment of Diabetic Kidney Disease. J Endocr Soc 2020; 4:bvaa029. [PMID: 32232184 PMCID: PMC7093089 DOI: 10.1210/jendso/bvaa029] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
Diabetic kidney disease (DKD) is an important public health problem. Podocyte injury is a central event in the mechanism of DKD development. Podocytes are terminally differentiated, highly specialized glomerular visceral epithelial cells critical for the maintenance of the glomerular filtration barrier. Although potential mechanisms by which diabetic milieu contributes to irreversible loss of podocytes have been described, identification of markers that prognosticate either the development of DKD or the progression to end-stage kidney disease (ESKD) have only recently made it to the forefront. Currently, the most common marker of early DKD is microalbuminuria; however, this marker has significant limitations: not all diabetic patients with microalbuminuria will progress to ESKD and as many as 30% of patients with DKD have normal urine albumin levels. Several novel biomarkers indicating glomerular or tubular damage precede microalbuminuria, suggesting that the latter develops when significant kidney injury has already occurred. Because podocyte injury plays a key role in DKD pathogenesis, identification of markers of early podocyte injury or loss may play an important role in the early diagnosis of DKD. Such biomarkers in the urine include podocyte-released microparticles as well as expression of podocyte-specific markers. Here, we review the mechanisms by which podocyte injury contributes to DKD as well as key markers that have been recently implicated in the development and/or progression of DKD and might serve to identify individuals that require earlier preventative care and treatment in order to slow the progression to ESKD.
Collapse
Affiliation(s)
- Igor Kravets
- Division of Endocrinology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
- Renal Section, Northport VA Medical Center, Northport, NY
| |
Collapse
|
35
|
Dorotea D, Koya D, Ha H. Recent Insights Into SREBP as a Direct Mediator of Kidney Fibrosis via Lipid-Independent Pathways. Front Pharmacol 2020; 11:265. [PMID: 32256356 PMCID: PMC7092724 DOI: 10.3389/fphar.2020.00265] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/24/2020] [Indexed: 12/17/2022] Open
Abstract
Sterol regulatory-element binding proteins (SREBPs) are classical regulators of cellular lipid metabolism in the kidney and other tissues. SREBPs are currently recognized as versatile transcription factors involved in a myriad of cellular processes. Meanwhile, SREBPs have been recognized to mediate lipotoxicity, contributing to the progression of kidney diseases. SREBP1 has been shown to bind to the promoter region of TGFβ, a major pro-fibrotic signaling mechanism in the kidney. Conversely, TGFβ activates SREBP1 transcriptional activity suggesting a positive feedback loop of SREBP1 in TGFβ signaling. Public ChIP-seq data revealed numerous non-lipid transcriptional targets of SREBPs that plausibly play roles in progressive kidney disease and fibrosis. This review provides new insights into SREBP as a mediator of kidney fibrosis via lipid-independent pathways.
Collapse
Affiliation(s)
- Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Daisuke Koya
- Department of Internal Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
36
|
Jihua C, Cai C, Xubin B, Yue Y. Effects of Dexmedetomidine on the RhoA /ROCK/ Nox4 Signaling Pathway in Renal Fibrosis of Diabetic Rats. Open Med (Wars) 2019; 14:890-898. [PMID: 31844679 PMCID: PMC6884926 DOI: 10.1515/med-2019-0105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Objective To investigate the effects and mechanisms of dexmedetomidine (Dex) on model rats of diabetic nephropathy (DN). Methods Rats were divided into NC, model, Dex-L (1μg/ kg), Dex-M (5μg/kg) and Dex-H (10μg/kg) groups. Rats in all groups except in the NC group were injected with streptozotocin (STZ) combined with right nephrectomy. Rats in Dex (1, 5 and 10μg/kg) groups received gavage with Dex (1, 5 and 10μg/kg). After 4 weeks, rats were sacrificed and kidneys were collected. HE staining was performed for a renal injury. Masson staining was applied to detect the fibrotic accumulation in rat kidney. Radioimmunoassay was used to test the renal function. Immunohistochemical method was used to detect protein expressions of RhoA, p-MYPT and Nox4 in rat kidney. Results Compared with the NC group, the levels of urine microalbumin in protein, α1-MG and β2-MG, renal fibrotic accumulation, RhoA, p-MYPT, Nox4 and α-SMA in model group increased significantly (P<0.001, respectively). Compared with the model group, Dex low, medium and high groups improved the deposition of renal fiber in rats, inhibited the expression levels of microalbumin, α1-MG and β2-MG in urine and decreased expression of RhoA, p-MYPT, Nox4 and α-SMA proteins (P<0.05, P<0.01). Conclusion Dex is possible to inhibit the expression of α-SMA and renal fibrous substance deposition in rat kidney via RhoA/ROCK/Nox4 signaling pathway, thereby reducing early kidney damage in model rats.
Collapse
Affiliation(s)
- Chen Jihua
- Department of anesthesiology, Fenghua District People's Hospital, Ningbo City, Zhejiang Province, Philippines
| | - Chen Cai
- Department of anesthesiology, Fenghua District People's Hospital, Ningbo City, Zhejiang Province, Philippines
| | - Bao Xubin
- Department of anesthesiology, Fenghua District People's Hospital, Ningbo City, Zhejiang Province, Philippines
| | - Yu Yue
- Department of anesthesiology, Fenghua District People's Hospital, Ningbo City, Zhejiang Province, Philippines
| |
Collapse
|
37
|
Kim DY, Kang MK, Kim YH, Lee EJ, Oh H, Kim SI, Oh SY, Kang YH. Eucalyptol Ameliorates Dysfunction of Actin Cytoskeleton Formation and Focal Adhesion Assembly in Glucose-Loaded Podocytes and Diabetic Kidney. Mol Nutr Food Res 2019; 63:e1900489. [PMID: 31483951 DOI: 10.1002/mnfr.201900489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/06/2019] [Indexed: 01/19/2023]
Abstract
SCOPE Podocytes are a component of glomerular filtration barrier with interdigitating foot processes. The podocyte function depends on the dynamics of actin cytoskeletal and focal adhesion crucial for foot process structure. This study investigates the renoprotective effects of eucalyptol on the F-actin cytoskeleton formation and focal adhesion assembly in glucose-loaded podocytes and diabetic kidneys. METHODS AND RESULTS Eucalyptol at 1-20 µm reverses the reduction of cellular level of F-actin, ezrin, cortactin, and Arp2/3 in 33 mm glucose-loaded mouse podocytes, and oral administration of 10 mg kg-1 eucalyptol elevates tissue levels of actin cytoskeletal proteins reduced in db/db mouse kidneys. Eucalyptol inhibits podocyte morphological changes, showing F-actin cytoskeleton formation in cortical regions and agminated F-actin along the cell periphery. Eucalyptol induces focal adhesion proteins of paxillin, vinculin, talin1, FAK, and Src in glucose-exposed podocytes and diabetic kidneys. Additionally, GTP-binding Rac1, Cdc42, Rho A, and ROCK are upregulated in glucose-stimulated podocytes and diabetic kidneys, which is attenuated by supplying eucalyptol. Rho A gene depletion partially diminishes GSK3β induction of podocytes by glucose. CONCLUSION Eucalyptol ameliorates F-actin cytoskeleton formation and focal adhesion assembly through blockade of the Rho signaling pathway, entailing partial involvement of GSK3β, which may inhibit barrier dysfunction of podocytes and resultant proteinuria.
Collapse
Affiliation(s)
- Dong Yeon Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Min-Kyung Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Yun-Ho Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Eun-Jung Lee
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Hyeongjoo Oh
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Soo-Il Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Su Yeon Oh
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| | - Young-Hee Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, 24252, Korea
| |
Collapse
|
38
|
Role of a RhoA/ROCK-Dependent Pathway on Renal Connexin43 Regulation in the Angiotensin II-Induced Renal Damage. Int J Mol Sci 2019; 20:ijms20184408. [PMID: 31500276 PMCID: PMC6770162 DOI: 10.3390/ijms20184408] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022] Open
Abstract
In various models of chronic kidney disease, the amount and localization of Cx43 in the nephron is known to increase, but the intracellular pathways that regulate these changes have not been identified. Therefore, we proposed that: "In the model of renal damage induced by infusion of angiotensin II (AngII), a RhoA/ROCK-dependent pathway, is activated and regulates the abundance of renal Cx43". In rats, we evaluated: 1) the time-point where the renal damage induced by AngII is no longer reversible; and 2) the involvement of a RhoA/ROCK-dependent pathway and its relationship with the amount of Cx43 in this irreversible stage. Systolic blood pressure (SBP) and renal function (urinary protein/urinary creatinine: Uprot/UCrea) were evaluated as systemic and organ outcomes, respectively. In kidney tissue, we also evaluated: 1) oxidative stress (amount of thiobarbituric acid reactive species), 2) inflammation (immunoperoxidase detection of the inflammatory markers ED-1 and IL-1β), 3) fibrosis (immune detection of type III collagen; Col III) and 4) activity of RhoA/ROCK (amount of phosphorylated MYPT1; p-MYPT1). The ratio Uprot/UCrea, SBP, oxidative stress, inflammation, amount of Cx43 and p-MYPT1 remained high 2 weeks after suspending AngII treatment in rats treated for 4 weeks with AngII. These responses were not observed in rats treated with AngII for less than 4 weeks, in which all measurements returned spontaneously close to the control values after suspending AngII treatment. Rats treated with AngII for 6 weeks and co-treated for the last 4 weeks with Fasudil, an inhibitor of ROCK, showed high SBP but did not present renal damage or increased amount of renal Cx43. Therefore, renal damage induced by AngII correlates with the activation of RhoA/ROCK and the increase in Cx43 amounts and can be prevented by inhibitors of this pathway.
Collapse
|
39
|
Nagai Y, Matoba K, Kawanami D, Takeda Y, Akamine T, Ishizawa S, Kanazawa Y, Yokota T, Utsunomiya K, Nishimura R. ROCK2 regulates TGF-β-induced expression of CTGF and profibrotic genes via NF-κB and cytoskeleton dynamics in mesangial cells. Am J Physiol Renal Physiol 2019; 317:F839-F851. [PMID: 31364374 DOI: 10.1152/ajprenal.00596.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The small GTPase Rho and its effector Rho kinase (ROCK) are involved in the pathogenesis of diabetic kidney disease. Rho kinase has two isoforms: ROCK1 and ROCK2. However, it remains unclear which is mainly involved in the progression of diabetic glomerulosclerosis and the regulation of profibrotic mediators. Glomeruli isolated from type 2 diabetic db/db mice demonstrated increased gene expression of transforming growth factor (TGF)-β and its downstream profibrotic mediators. Chemical inhibition of ROCK suppressed the expression of profibrotic mediators in both isolated glomeruli and cultured mesangial cells. An investigation of mechanisms underlying this observation revealed activated ROCK functions through the phosphorylation of JNK and Erk and the nuclear translocation of NF-κB via actin dynamics. Knockdown by siRNA against ROCK1 and ROCK2 showed that ROCK2 but not ROCK1 controls this fibrotic machinery. Further in vivo experiments showed that ROCK2 activity in the renal cortex of db/db mice was elevated compared with control db/m mice. Importantly, oral administration of ROCK2 inhibitor attenuated renal ROCK2 activity, albuminuria, and glomerular fibrosis in db/db mice. These observations indicate that ROCK2 is a key player in the development of diabetic renal injury. Glomerular ROCK2 may be a potential therapeutic target for the treatment of diabetic kidney disease.
Collapse
Affiliation(s)
- Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomoyo Akamine
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Sho Ishizawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Jiang C, Wang Y, Jin Q, Zhang D, Gao M, Yao N, Yin Z, Zhang J, Ma S. Cyclocarya paliurus Triterpenoids Improve Diabetes-Induced Hepatic Inflammation via the Rho-Kinase-Dependent Pathway. Front Pharmacol 2019; 10:811. [PMID: 31404259 PMCID: PMC6669819 DOI: 10.3389/fphar.2019.00811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/24/2019] [Indexed: 12/19/2022] Open
Abstract
This study aimed to assess the effects of triterpene extract of Cyclocarya paliurus (Batal.) Iljinskaja (CPT) on diabetes-induced hepatic inflammation and to unveil the underlying mechanisms. Diabetes in db/db mice was alleviated after CPT administration, as assessed by the oral glucose tolerance test. In addition, treatment with CPT dramatically reduced serum insulin, aspartate amino-transaminase, alanine aminotransferase, triglyceride, and total cholesterol amounts. Besides, serum levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α were also reduced after CPT administration. Western blot analysis revealed that CPT treatment significantly reversed the protein expression levels of Rho, ROCK1, ROCK2, p-P65, p-IκBα, p-IKKα, and p-IKKβ in liver samples obtained from db/db mice. Upon palmitic acid stimulation, the protective effects of CPT on the liver were further assessed in HepG2 and LO2 cells, and no appreciable cytotoxic effects were found. Therefore, these findings indicate that CPT alleviates liver inflammation via Rho-kinase signaling. Chemical compounds evaluated in this report: Metformin (PubChem CID: 4091); Fasudil (PubChem CID: 3547); Palmitic acid (PubChem CID: 985).
Collapse
Affiliation(s)
- Cuihua Jiang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China.,Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yiting Wang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Nan Yao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhiqi Yin
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
41
|
Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: from the periphery to the brain. J Neurochem 2019; 150:648-665. [PMID: 31106417 DOI: 10.1111/jnc.14725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly apparent that disorders of the brain microvasculature contribute to many neurological disorders. In recent years it has become clear that a major player in these events is the capillary pericyte which, in the brain, is now known to control the blood-brain barrier, regulate blood flow, influence immune cell entry and be crucial for angiogenesis. In this review we consider the under-explored possibility that peripheral diseases which affect the microvasculature, such as hypertension, kidney disease and diabetes, produce central nervous system (CNS) dysfunction by mechanisms affecting capillary pericytes within the CNS. We highlight how cellular messengers produced peripherally can act via signalling pathways within CNS pericytes to reshape blood vessels, restrict blood flow or compromise blood-brain barrier function, thus causing neuronal dysfunction. Increased understanding of how renin-angiotensin, Rho-kinase and PDGFRβ signalling affect CNS pericytes may suggest novel therapeutic approaches to reducing the CNS effects of peripheral disorders.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - David Attwell
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| |
Collapse
|
42
|
Purvis GSD, Collino M, Loiola RA, Baragetti A, Chiazza F, Brovelli M, Sheikh MH, Collotta D, Cento A, Mastrocola R, Aragno M, Cutrin JC, Reutelingsperger C, Grigore L, Catapano AL, Yaqoob MM, Norata GD, Solito E, Thiemermann C. Identification of AnnexinA1 as an Endogenous Regulator of RhoA, and Its Role in the Pathophysiology and Experimental Therapy of Type-2 Diabetes. Front Immunol 2019; 10:571. [PMID: 30972066 PMCID: PMC6446914 DOI: 10.3389/fimmu.2019.00571] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Annexin A1 (ANXA1) is an endogenously produced anti-inflammatory protein, which plays an important role in the pathophysiology of diseases associated with chronic inflammation. We demonstrate that patients with type-2 diabetes have increased plasma levels of ANXA1 when compared to normoglycemic subjects. Plasma ANXA1 positively correlated with fatty liver index and elevated plasma cholesterol in patients with type-2 diabetes, suggesting a link between aberrant lipid handling, and ANXA1. Using a murine model of high fat diet (HFD)-induced insulin resistance, we then investigated (a) the role of endogenous ANXA1 in the pathophysiology of HFD-induced insulin resistance using ANXA1−/− mice, and (b) the potential use of hrANXA1 as a new therapeutic approach for experimental diabetes and its microvascular complications. We demonstrate that: (1) ANXA1−/− mice fed a HFD have a more severe diabetic phenotype (e.g., more severe dyslipidemia, insulin resistance, hepatosteatosis, and proteinuria) compared to WT mice fed a HFD; (2) treatment of WT-mice fed a HFD with hrANXA1 attenuated the development of insulin resistance, hepatosteatosis and proteinuria. We demonstrate here for the first time that ANXA1−/− mice have constitutively activated RhoA. Interestingly, diabetic mice, which have reduced tissue expression of ANXA1, also have activated RhoA. Treatment of HFD-mice with hrANXA1 restored tissue levels of ANXA1 and inhibited RhoA activity, which, in turn, resulted in restoration of the activities of Akt, GSK-3β and endothelial nitric oxide synthase (eNOS) secondary to re-sensitization of IRS-1 signaling. We further demonstrate in human hepatocytes that ANXA1 protects against excessive mitochondrial proton leak by activating FPR2 under hyperglycaemic conditions. In summary, our data suggest that (a) ANXA1 is a key regulator of RhoA activity, which restores IRS-1 signal transduction and (b) recombinant human ANXA1 may represent a novel candidate for the treatment of T2D and/or its complications.
Collapse
Affiliation(s)
- Gareth S D Purvis
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Rodrigo A Loiola
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Martina Brovelli
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy.,Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy
| | - Madeeha H Sheikh
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Alessia Cento
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Raffaella Mastrocola
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Manuela Aragno
- Department of Molecular Biotechnology and Sciences for the Health, University of Turin, Turin, Italy
| | - Juan C Cutrin
- Department of Molecular Biotechnology and Sciences for the Health, University of Turin, Turin, Italy
| | - Chris Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute, Maastricht University, Maastricht, Netherlands
| | - Liliana Grigore
- Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy.,IRCCS Multimedica, Lombardy, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Magdi M Yaqoob
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy.,Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy
| | - Egle Solito
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - Christoph Thiemermann
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
43
|
Sun W, Gao Y, Ding Y, Cao Y, Chen J, Lv G, Lu J, Yu B, Peng M, Xu H, Sun Y. Catalpol ameliorates advanced glycation end product-induced dysfunction of glomerular endothelial cells via regulating nitric oxide synthesis by inducible nitric oxide synthase and endothelial nitric oxide synthase. IUBMB Life 2019; 71:1268-1283. [PMID: 30861639 DOI: 10.1002/iub.2032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
Abstract
Catalpol (Cat.) is an iridoid glucoside extracted from the root of Rehmannia glutinosa Libosch. In this study, we investigated whether Cat. could protect the mouse glomerular endothelial cells against the deleterious effect induced by advanced glycation end products (AGEs) and explored potential mechanisms. We found that 10 μM Cat. showed a protective effect on dead cells stimulated by AGEs. Cat. significantly decreased the expression of p-NF-κBp65 and inducible nitric oxide synthase (iNOS) and increased the expression of phosphorylated-endothelial nitric oxide synthase (p-eNOS; Ser1177), PI3K, p-Akt (Thr308), and total-Akt. Moreover, Cat. restored the integrity of glomerular endothelial barrier by increasing endothelial tight gap junction protein and ameliorated the endothelial hyperpermeability induced by AGEs via modulating the nitric oxide (NO) production. Additionally, Cat. attenuated the massive release of NO induced by AGEs, inhibiting the macrophage infiltration by modulating the NO production, accompanied by the decrease in the release of monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1 in vitro. Therefore, Cat. ameliorated AGEs-induced endothelial dysfunction via inhibiting the NF-κB/iNOS pathway and activating the PI3K/Akt/eNOS pathway. © 2019 IUBMB Life, 71(9):1268-1283, 2019.
Collapse
Affiliation(s)
- Weixiang Sun
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yuyan Gao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yushi Ding
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Ying Cao
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Jing Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Gaohong Lv
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jinfu Lu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Bin Yu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Meilin Peng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Huiqin Xu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| | - Yun Sun
- Department of Pharmacology, School of Pharmacy, Hanlin College, Nanjing University of Chinese Medicine, Taizhou, People's Republic of China
| |
Collapse
|
44
|
Increased levels of serum pigment epithelium-derived factor aggravate proteinuria via induction of podocyte actin rearrangement. Int Urol Nephrol 2018; 51:359-367. [PMID: 30536192 PMCID: PMC6394770 DOI: 10.1007/s11255-018-2026-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 11/03/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE To assess the role of serum pigment epithelium-derived factor (PEDF) in the occurrence and development of proteinuria and renal dysfunction and determine its relevant signaling pathway. METHODS We analyzed serum PEDF, creatinine, the urinary albumin-to-creatinine ratio, and renal morphology of normal or streptozotocin (STZ)-induced diabetic mice, before and after treatment with PEDF. In vitro, podocytes were stimulated with PEDF under normal or high-glucose conditions; permeability was measured by the transwell assay with fluorescein isothiocyanate (FITC)-dextran; and F-actin cytoskeleton was analyzed by phalloidin staining. Apoptosis was assessed by flow cytometry. RhoA activity and ROCK1, ZO-1, nephrin, and podocin levels were detected by Western blotting. RESULTS Diabetic mice exhibited a high serum PEDF level. In vivo, elevated serum PEDF led to proteinuria, increased serum creatinine, and podocyte foot process fusion in normal or diabetic mice. In vitro, both high-glucose and PEDF stimulation activated the RhoA/ROCK1 pathway in podocytes and promoted cell permeability, F-actin rearrangement, and apoptosis. Inhibition of RhoA/ROCK1 alleviated the damage from these effects. CONCLUSIONS Elevated serum PEDF aggravates the development of proteinuria and renal dysfunction by inducing F-actin arrangement, foot process fusion, and apoptosis of podocytes in both normal and diabetic mice, and this effect may be mediated by activation of the RhoA/ROCK1 pathway.
Collapse
|
45
|
Utility of curcumin for the treatment of diabetes mellitus: Evidence from preclinical and clinical studies. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
46
|
Sharma D, Gondaliya P, Tiwari V, Kalia K. Kaempferol attenuates diabetic nephropathy by inhibiting RhoA/Rho-kinase mediated inflammatory signalling. Biomed Pharmacother 2018; 109:1610-1619. [PMID: 30551415 DOI: 10.1016/j.biopha.2018.10.195] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022] Open
Abstract
RhoA/Rho-associated coiled-coil forming protein serine/threonine kinase (ROCK) has appeared as a potential therapeutic target in numerous diseases, because of its preventing action on various enzymes providing antioxidant and cytoprotective action. Progression and pathophysiology of diabetic nephropathy have also shown potential involvement of oxidative stress and inflammatory pathways. In the present study, we investigated the effect of kaempferol on hyperglycemia-induced activation of RhoA kinase and associated inflammatory signaling cascade. Currently there is only small literature available on the mechanism of anti-diabetic and nephroprotective action of this compound, which creates a void. Therefore, we focused here on the investigation of molecular mechanisms for kaempferol by means of in vitro testing, using rat (NRK-52E) and human renal tubular epithelial cells (RPTEC). Our findings suggest that kaempferol inhibits hyperglycemia-induced activation of RhoA and decreased oxidative stress, pro-inflammatory cytokines (TNF-α and IL-1β) and fibrosis (TGF-β1 expression, extracellular matrix protein expression) in NRK-52E and RPTEC cells. Therefore, kaempferol can be used as a potential therapeutic for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Piyush Gondaliya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Vinod Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
47
|
FOXO1 inhibition potentiates endothelial angiogenic functions in diabetes via suppression of ROCK1/Drp1-mediated mitochondrial fission. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2481-2494. [DOI: 10.1016/j.bbadis.2018.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/30/2018] [Accepted: 04/08/2018] [Indexed: 12/22/2022]
|
48
|
Petit AP, Garcia-Petit C, Bueren-Calabuig JA, Vuillard LM, Ferry G, Boutin JA. A structural study of the complex between neuroepithelial cell transforming gene 1 (Net1) and RhoA reveals a potential anticancer drug hot spot. J Biol Chem 2018; 293:9064-9077. [PMID: 29695506 DOI: 10.1074/jbc.ra117.001123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/18/2018] [Indexed: 12/20/2022] Open
Abstract
The GTPase RhoA is a major player in many different regulatory pathways. RhoA catalyzes GTP hydrolysis, and its catalysis is accelerated when RhoA forms heterodimers with proteins of the guanine nucleotide exchange factor (GEF) family. Neuroepithelial cell transforming gene 1 (Net1) is a RhoA-interacting GEF implicated in cancer, but the structural features supporting the RhoA/Net1 interaction are unknown. Taking advantage of a simple production and purification process, here we solved the structure of a RhoA/Net1 heterodimer with X-ray crystallography at 2-Å resolution. Using a panel of several techniques, including molecular dynamics simulations, we characterized the RhoA/Net1 interface. Moreover, deploying an extremely simple peptide-based scanning approach, we found that short peptides (penta- to nonapeptides) derived from the protein/protein interaction region of RhoA could disrupt the RhoA/Net1 interaction and thereby diminish the rate of nucleotide exchange. The most inhibitory peptide, EVKHF, spanning residues 102-106 in the RhoA sequence, displayed an IC50 of ∼100 μm without further modifications. The peptides identified here could be useful in further investigations of the RhoA/Net1 interaction region. We propose that our structural and functional insights might inform chemical approaches for transforming the pentapeptide into an optimized pseudopeptide that antagonizes Net1-mediated RhoA activation with therapeutic anticancer potential.
Collapse
Affiliation(s)
- Alain-Pierre Petit
- From the Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery and
| | - Christel Garcia-Petit
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom and
| | | | - Laurent M Vuillard
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| | - Gilles Ferry
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| | - Jean A Boutin
- Pôle d'Expertise Biotechnologie, Chimie, Biologie, Institut de Recherches SERVIER, 78290 Croissy-sur-Seine, France
| |
Collapse
|
49
|
Angiotensin II-Induced Mesangial Cell Damaged Is Preceded by Cell Membrane Permeabilization Due to Upregulation of Non-Selective Channels. Int J Mol Sci 2018; 19:ijms19040957. [PMID: 29570626 PMCID: PMC5979336 DOI: 10.3390/ijms19040957] [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] [Received: 02/16/2018] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023] Open
Abstract
Connexin43 (Cx43), pannexin1 (Panx1) and P2X7 receptor (P2X7R) are expressed in kidneys and are known to constitute a feedforward mechanism leading to inflammation in other tissues. However, the possible functional relationship between these membrane channels and their role in damaged renal cells remain unknown. In the present work, we found that MES-13 cells, from a cell line derived from mesangial cells, stimulated with angiotensin II (AngII) developed oxidative stress (OS, thiobarbituric acid reactive species (TBARS) and generated pro-inflammatory cytokines (ELISA; IL-1β and TNF-α). The membrane permeability increased progressively several hours before the latter outcome, which was a response prevented by Losartan, indicating the involvement of AT1 receptors. Western blot analysis showed that the amount of phosphorylated MYPT (a substrate of RhoA/ROCK) and Cx43 increased progressively and in parallel in cells treated with AngII, a response followed by an increase in the amount in Panx1 and P2X7R. Greater membrane permeability was partially explained by opening of Cx43 hemichannels (Cx43 HCs) and Panx1 channels (Panx1 Chs), as well as P2X7Rs activation by extracellular ATP, which was presumably released via Cx HCs and Panx1 Chs. Additionally, inhibition of RhoA/ROCK blocked the progressive increase in membrane permeability, and the remaining response was explained by the other non-selective channels. The rise of activity in the RhoA/ROCK-dependent pathway, as well as in Cx HCs, P2X7R, and to a minor extent in Panx1 Chs led to higher amounts of TBARS and pro-inflammatory cytokines. We propose that AngII-induced mesangial cell damage could be effectively inhibited by concomitantly inhibiting the RhoA/ROCK-dependent pathway and one or more non-selective channel(s) activated through this pathway.
Collapse
|
50
|
Wang Q, Yang X, Xu Y, Shen Z, Cheng H, Cheng F, Liu X, Wang R. RhoA/Rho-kinase triggers epithelial-mesenchymal transition in mesothelial cells and contributes to the pathogenesis of dialysis-related peritoneal fibrosis. Oncotarget 2018; 9:14397-14412. [PMID: 29581852 PMCID: PMC5865678 DOI: 10.18632/oncotarget.24208] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 12/05/2017] [Indexed: 12/22/2022] Open
Abstract
Peritoneal fibrosis (PF) with associated peritoneal dysfunction is almost invariably observed in long-term peritoneal dialysis (PD) patients. Advanced glycation end products (AGEs) are pro-oxidant compounds produced in excess during the metabolism of glucose and are present in high levels in standard PD solutions. The GTPase RhoA has been implicated in PF, but its specific role remains poorly understood. Here, we studied the effects of RhoA/Rho-kinase signaling in AGEs-induced epithelial-mesenchymal transition (EMT) in human peritoneal mesothelial cells (HPMCs), and evaluated morphological and molecular changes in a rat model of PD-related PF. Activation of RhoA/Rho-kinase and activating protein-1 (AP-1) was assessed in HPMCs using pull-down and electrophoretic mobility shift assays, respectively, while expression of transforming growth factor-β, fibronectin, α-smooth muscle actin, vimentin, N-cadherin, and E-cadherin expression was assessed using immunohistochemistry and western blot. AGEs exposure activated Rho/Rho-kinase in HPMCs and upregulated EMT-related genes via AP-1. These changes were prevented by the Rho-kinase inhibitors fasudil and Y-27632, and by the AP-1 inhibitor curcumin. Importantly, fasudil normalized histopathological and molecular alterations and preserved peritoneal function in rats. These data support the therapeutic potential of Rho-kinase inhibitors in PD-related PF.
Collapse
Affiliation(s)
- Qinglian Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiaowei Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Ying Xu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhenwei Shen
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Hongxia Cheng
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Fajuan Cheng
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiang Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|