1
|
Thomas HY, Ford Versypt AN. A mathematical model of glomerular fibrosis in diabetic kidney disease to predict therapeutic efficacy. Front Pharmacol 2024; 15:1481768. [PMID: 39525640 PMCID: PMC11543426 DOI: 10.3389/fphar.2024.1481768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Background Glomerular fibrosis is a tissue damage that occurs within the kidneys of chronic and diabetic kidney disease patients. Effective treatments are lacking, and the mechanism of glomerular damage reversal is poorly understood. Methods A mathematical model suitable for hypothesis-driven systems pharmacology of glomerular fibrosis in diabetes was developed from a previous model of interstitial fibrosis. The adapted model consists of a system of ordinary differential equations that models the complex disease etiology and progression of glomerular fibrosis in diabetes. Results Within the scope of the mechanism incorporated, advanced glycation end products (AGE)-matrix proteins that are modified due to high blood glucose-were identified as major contributors to the delay in the recovery from glomerular fibrosis after glucose control. The model predicted that inhibition of AGE production is not an effective approach for accelerating the recovery from glomerular fibrosis. Further, the model predicted that treatment breaking down accumulated AGE is the most productive at reversing glomerular fibrosis. The use of the model led to the identification that glucose control and aminoguanidine are ineffective treatments for reversing advanced glomerular fibrosis because they do not remove accumulated AGE. Additionally, using the model, a potential explanation was generated for the lack of efficacy of alagebrium in treating advanced glomerular fibrosis, which is due to the inability of alagebrium to reduce TGF- β . Impact Using the mathematical model, a mechanistic understanding of disease etiology and complexity of glomerular fibrosis in diabetes was illuminated, and then hypothesis-driven explanations for the lack of efficacy of different pharmacological agents for treating glomerular fibrosis were provided. This understanding can enable the development of more efficacious therapeutics for treating kidney damage in diabetes.
Collapse
Affiliation(s)
- Haryana Y. Thomas
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Ashlee N. Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
2
|
Attia H, Badr A, Alshehri O, Alsulaiman W, Alshanwani A, Alshehri S, Arafa M, Hasan I, Ali R. The Protective Effects of Vitamin B Complex on Diclofenac Sodium-Induced Nephrotoxicity: The Role of NOX4/RhoA/ROCK. Inflammation 2024; 47:1600-1615. [PMID: 38413451 DOI: 10.1007/s10753-024-01996-6] [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: 12/02/2023] [Revised: 02/04/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Diclofenac sodium (DIC) is a widely used non-steroidal anti-inflammatory drug. Unfortunately, its prolonged use is associated with nephrotoxicity due to oxidative stress, inflammation, and fibrosis. We aimed to investigate the nephroprotective effects of vitamin B complex (B1, B6, B12) against DIC-induced nephrotoxicity and its impact on NOX4/RhoA/ROCK, a pathway that plays a vital role in renal pathophysiology. Thirty-two Wistar rats were divided into four groups: (1) normal control; (2) vitamin B complex (16 mg/kg B1, 16 mg/kg B6, 0.16 mg/kg B12, intraperitoneal); (3) DIC (10 mg/kg, intramuscular); and (4) DIC plus vitamin B complex group. After 14 days, the following were assayed: serum renal biomarkers (creatinine, blood urea nitrogen, kidney injury molecule-1), oxidative stress, inflammatory (tumor necrosis factor-α, interleukin-6), and fibrotic (transforming growth factor-β) markers as well as the protein levels of NOX4, RhoA, and ROCK. Structural changes, inflammatory cell infiltration, and fibrosis were detected using hematoxylin and eosin and Masson trichrome stains. Compared to DIC, vitamin B complex significantly decreased the renal function biomarkers, markers of oxidative stress and inflammation, and fibrotic cytokines. Glomerular and tubular damage, inflammatory infiltration, and excessive collagen accumulation were also reduced. Protein levels of NOX4, RhoA, and ROCK were significantly elevated by DIC, and this elevation was ameliorated by vitamin B complex. In conclusion, vitamin B complex administration could be a renoprotective approach during treatment with DIC via, at least in part, suppressing the NOX4/RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box: 2454, Riyadh, 11495, Saudi Arabia.
| | - Amira Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box: 2454, Riyadh, 11495, Saudi Arabia
| | - Orjuwan Alshehri
- College of Pharmacy, King Saud University, Riyadh, 11495, Saudi Arabia
| | - Waad Alsulaiman
- College of Pharmacy, King Saud University, Riyadh, 11495, Saudi Arabia
| | - Aliah Alshanwani
- Department of Physiology, College of Medicine, King Saud University, Riyadh, 11495, Saudi Arabia
| | - Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box: 2454, Riyadh, 11495, Saudi Arabia
| | - Maha Arafa
- Pathology Department, College of Medicine, King Saud University, Riyadh, 11495, Saudi Arabia
| | - Iman Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box: 2454, Riyadh, 11495, Saudi Arabia
| | - Rehab Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P. O. Box: 2454, Riyadh, 11495, Saudi Arabia
| |
Collapse
|
3
|
Li X, Du H, Zhou H, Huang Y, Tang S, Yu C, Guo Y, Luo W, Gong Y. FOXL2 regulates RhoA expression to change actin cytoskeleton rearrangement in granulosa cells of chicken pre-ovulatory follicles†. Biol Reprod 2024; 111:391-405. [PMID: 38832713 DOI: 10.1093/biolre/ioae082] [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: 10/04/2023] [Revised: 04/04/2024] [Accepted: 06/01/2024] [Indexed: 06/05/2024] Open
Abstract
Forkhead box L2 (FOXL2) is an indispensable key regulator of female follicular development, and it plays important roles in the morphogenesis, proliferation, and differentiation of follicle granulosa cells, such as establishing normal estradiol signaling and regulating steroid hormone synthesis. Nevertheless, the effects of FOXL2 on granulosa cell morphology and the underlying mechanism remain unknown. Using FOXL2 ChIP-seq analysis, we found that FOXL2 target genes were significantly enriched in the actin cytoskeleton-related pathways. We confirmed that FOXL2 inhibited the expression of RhoA, a key gene for actin cytoskeleton rearrangement, by binding to TCATCCATCTCT in RhoA promoter region. In addition, FOXL2 overexpression in granulosa cells induced the depolymerization of F-actin and disordered the actin filaments, resulting in a slowdown in the expansion of granulosa cells, while FOXL2 silencing inhibited F-actin depolymerization and stabilized the actin filaments, thereby accelerating granulosa cell expansion. RhoA/ROCK pathway inhibitor Y-27632 exhibited similar effects to FOXL2 overexpression, even reversed the actin polymerization in FOXL2 silencing granulosa cells. This study revealed for the first time that FOXL2 regulated granulosa cell actin cytoskeleton by RhoA/ROCK pathway, thus affecting granulosa cell expansion. Our findings provide new insights for constructing the regulatory network of FOXL2 and propose a potential mechanism for facilitating rapid follicle expansion, thereby laying a foundation for further understanding follicular development.
Collapse
Affiliation(s)
- Xuelian Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Hongting Du
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Haobo Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Ying Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Shuixin Tang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Chengzhi Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yan Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Wei Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yanzhang Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| |
Collapse
|
4
|
Yang Z, Chen L, Huang Y, Dong J, Yan Q, Li Y, Qiu J, Li H, Zhao D, Liu F, Tang D, Dai Y. Proteomic profiling of laser capture microdissection kidneys from diabetic nephropathy patients. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1243:124231. [PMID: 38996754 DOI: 10.1016/j.jchromb.2024.124231] [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: 01/11/2024] [Revised: 05/23/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Diabetic nephropathy (DN) remains the primary cause of end-stage renal disease (ESRD), warranting equal attention and separate analysis of glomerular, tubular, and interstitial lesions in its diagnosis and intervention. This study aims to identify the specific proteomics characteristics of DN, and assess changes in the biological processes associated with DN. 5 patients with DN and 5 healthy kidney transplant donor control individuals were selected for analysis. The proteomic characteristics of glomeruli, renal tubules, and renal interstitial tissue obtained through laser capture microscopy (LCM) were studied using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Significantly, the expression of multiple heat shock proteins (HSPs), tubulins, and heterogeneous nuclear ribonucleoproteins (hnRNPs) in glomeruli and tubules was significantly reduced. Differentially expressed proteins (DEPs) in the glomerulus showed significant enrichment in pathways related to cell junctions and cell movement, including the regulation of actin cytoskeleton and tight junction. DEPs in renal tubules were significantly enriched in glucose metabolism-related pathways, such as glucose metabolism, glycolysis/gluconeogenesis, and the citric acid cycle. Moreover, the glycolysis/gluconeogenesis pathway was a co-enrichment pathway in both DN glomeruli and tubules. Notably, ACTB emerged as the most crucial protein in the protein-protein interaction (PPI) analysis of DEPs in both glomeruli and renal tubules. In this study, we delve into the unique proteomic characteristics of each sub-region of renal tissue. This enhances our understanding of the potential pathophysiological changes in DN, particularly the potential involvement of glycolysis metabolic disorder, glomerular cytoskeleton and cell junctions. These insights are crucial for further research into the identification of disease biomarkers and the pathogenesis of DN.
Collapse
Affiliation(s)
- Zhiqian Yang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Liangmei Chen
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Yingxin Huang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; Department of Nephrology, Xiaolan People's Hospital of Zhongshan, 528400, China
| | - Jingjing Dong
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Qiang Yan
- Department of Organ Transplantation, 924 Hospital, Guilin 541002, China
| | - Ya Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Jing Qiu
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Haitao Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Da Zhao
- The First Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan 232001, Anhui, China
| | - Fanna Liu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China.
| | - Donge Tang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People' s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China.
| | - Yong Dai
- Comprehensive Health Industry Research Center, Taizhou Research Institute, Southern University of Science and Technology, Taizhou 317000, China; The First Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan 232001, Anhui, China.
| |
Collapse
|
5
|
Huyan T, Fan L, Zheng ZY, Zhao JH, Han ZR, Wu P, Ma Q, Du YQ, Shi YD, Gu CY, Li XJ, Wang WH, Zhang L, Tie L. ROCK1 inhibition improves wound healing in diabetes via RIPK4/AMPK pathway. Acta Pharmacol Sin 2024; 45:1477-1491. [PMID: 38538716 PMCID: PMC11192920 DOI: 10.1038/s41401-024-01246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/19/2024] [Indexed: 06/23/2024]
Abstract
Refractory wounds are a severe complication of diabetes mellitus that often leads to amputation because of the lack of effective treatments and therapeutic targets. The pathogenesis of refractory wounds is complex, involving many types of cells. Rho-associated protein kinase-1 (ROCK1) phosphorylates a series of substrates that trigger downstream signaling pathways, affecting multiple cellular processes, including cell migration, communication, and proliferation. The present study investigated the role of ROCK1 in diabetic wound healing and molecular mechanisms. Our results showed that ROCK1 expression significantly increased in wound granulation tissues in diabetic patients, streptozotocin (STZ)-induced diabetic mice, and db/db diabetic mice. Wound healing and blood perfusion were dose-dependently improved by the ROCK1 inhibitor fasudil in diabetic mice. In endothelial cells, fasudil and ROCK1 siRNA significantly elevated the phosphorylation of adenosine monophosphate-activated protein kinase at Thr172 (pThr172-AMPKα), the activity of endothelial nitric oxide synthase (eNOS), and suppressed the levels of mitochondrial reactive oxygen species (mtROS) and nitrotyrosine formation. Experiments using integrated bioinformatics analysis and coimmunoprecipitation established that ROCK1 inhibited pThr172-AMPKα by binding to receptor-interacting serine/threonine kinase 4 (RIPK4). These results suggest that fasudil accelerated wound repair and improved angiogenesis at least partially through the ROCK1/RIPK4/AMPK pathway. Fasudil may be a potential treatment for refractory wounds in diabetic patients.
Collapse
Affiliation(s)
- Tianru Huyan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Lu Fan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhong-Yuan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Jing-Hui Zhao
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Zhen-Ru Han
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Qun Ma
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Ya-Qin Du
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Yun-di Shi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Chun-Yan Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xue-Jun Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Wen-Hui Wang
- Department of Dermatology, Peking University Third Hospital, Beijing, 100191, China
| | - Long Zhang
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China.
| |
Collapse
|
6
|
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
|
7
|
Han L, Wang S, Li J, Zhao L, Zhou H. Urinary exosomes from patients with diabetic kidney disease induced podocyte apoptosis via microRNA-145-5p/Srgap2 and the RhoA/ROCK pathway. Exp Mol Pathol 2023; 134:104877. [PMID: 37952894 DOI: 10.1016/j.yexmp.2023.104877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease without early diagnostic and specific therapeutic approaches. Podocyte apoptosis and loss play important roles in the pathological process of DKD. This study aimed to explore whether urinary exosomes from type 2 diabetes patients with DKD could induce podocyte apoptosis and the underlying pathological mechanisms. The exosomes were isolated from the urine samples of patients with DKD (DKD-Exo). Later, they were taken up and internalized by MPC5 cells. MPC5 cells were co-cultured with DKD-Exo (45 μg/ml) for 24 h in the presence or absence of microRNA-145-5p (miR-145-5p) inhibitor, fasudil and pcDNA-Srgap2 transfection. MiR-145-5p and Srgap2 expression was evaluated using real-time quantitative PCR. The protein levels of Srgap2, Bcl-2, Bax, and cleaved caspase-3, as well as ROCK activity were determined using Western blotting. Cell apoptosis was measured using flow cytometry and the TUNEL assay. miR-145-5p expression in MPC5 cells exposed to DKD-Exo was markedly upregulated. miR-145-5p negatively regulated Srgap2 levels. Exposure of MPC5 cells to DKD-Exo reduced Srgap2 expression and activated ROCK, which was partly reversed by the presence of the miR-145-5p inhibitor or Srgap2 overexpression. The apoptosis of MPC5 cells exposed to DKD-Exo increased significantly, which was counteracted by the addition of the miR-145-5p inhibitor and fasudil. The results showed that urinary exosomal miR-145-5p from patients with DKD induced podocyte apoptosis by inhibiting Srgap2 and activating the RhoA/ROCK pathway, suggesting that urinary exosomal miR-145-5p is involved in the pathological process of DKD and could become a noninvasive diagnostic biomarker for DKD.
Collapse
Affiliation(s)
- Lulu Han
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Department of Endocrinology, the First Central Hospital of Baoding, Baoding 071000, China
| | - Shenghai Wang
- Department of Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Juan Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lulu Zhao
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
8
|
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
|
9
|
Njeim R, Alkhansa S, Fornoni A. Unraveling the Crosstalk between Lipids and NADPH Oxidases in Diabetic Kidney Disease. Pharmaceutics 2023; 15:pharmaceutics15051360. [PMID: 37242602 DOI: 10.3390/pharmaceutics15051360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious complication of diabetes mellitus and a leading cause of end-stage renal disease. Abnormal lipid metabolism and intrarenal accumulation of lipids have been shown to be strongly correlated with the development and progression of diabetic kidney disease (DKD). Cholesterol, phospholipids, triglycerides, fatty acids, and sphingolipids are among the lipids that are altered in DKD, and their renal accumulation has been linked to the pathogenesis of the disease. In addition, NADPH oxidase-induced production of reactive oxygen species (ROS) plays a critical role in the development of DKD. Several types of lipids have been found to be tightly linked to NADPH oxidase-induced ROS production. This review aims to explore the interplay between lipids and NADPH oxidases in order to provide new insights into the pathogenesis of DKD and identify more effective targeted therapies for the disease.
Collapse
Affiliation(s)
- Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sahar Alkhansa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
10
|
Proietti R, Giordani AS, Lorenzo CA. ROCK (RhoA/Rho Kinase) Activation in Atrial Fibrillation: Molecular Pathways and Clinical Implications. Curr Cardiol Rev 2023; 19:e171122210986. [PMID: 36625201 PMCID: PMC10280999 DOI: 10.2174/1573403x19666221117092951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 12/15/2022] Open
Abstract
Among the complex mechanisms of AF pathogenesis, intracellular calcium overload and oxidative stress play a major role, both triggered by inflammatory processes. The additional basic event taking place in AF is atrial fibrotic remodeling, again triggered by oxidative stress, which is determined by connexins rearrangement and differentiation of fibroblasts into active collagensecreting myofibroblasts. RhoA/ROCK system is the final pathway of a wide spectrum of molecular effectors such as Angiotensin II, platelet-derived growth factor, connective tissue growth factor and transforming growth factor β, that overall determine calcium dysregulation and pro-fibrotic remodeling. Both in experimental and clinical studies, RhoA/ROCK activation has been linked to superoxide ion production, fibrotic remodeling and connexins rearrangement, with important consequences for AF pathogenesis. ROCK pathway inhibition may therefore be a therapeutic or preventive target for special AF subgroups of patients.
Collapse
Affiliation(s)
- Riccardo Proietti
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Andrea S. Giordani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Calò A. Lorenzo
- Department of Medicine (DIMED), Nephrology, Dialysis and Transplantation Unit, University of Padua and Azienda Ospedale Università di Padova, Padua, Italy
| |
Collapse
|
11
|
Trink J, Ahmed U, O'Neil K, Li R, Gao B, Krepinsky JC. Cell surface GRP78 regulates TGFβ1-mediated profibrotic responses via TSP1 in diabetic kidney disease. Front Pharmacol 2023; 14:1098321. [PMID: 36909183 PMCID: PMC9998550 DOI: 10.3389/fphar.2023.1098321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Introduction: Diabetic kidney disease (DKD) is the leading cause of kidney failure in North America, characterized by glomerular accumulation of extracellular matrix (ECM) proteins. High glucose (HG) induction of glomerular mesangial cell (MC) profibrotic responses plays a central role in its pathogenesis. We previously showed that the endoplasmic reticulum resident GRP78 translocates to the cell surface in response to HG, where it mediates Akt activation and downstream profibrotic responses in MC. Transforming growth factor β1 (TGFβ1) is recognized as a central mediator of HG-induced profibrotic responses, but whether its activation is regulated by cell surface GRP78 (csGRP78) is unknown. TGFβ1 is stored in the ECM in a latent form, requiring release for biological activity. The matrix glycoprotein thrombospondin 1 (TSP1), known to be increased in DKD and by HG in MC, is an important factor in TGFβ1 activation. Here we determined whether csGRP78 regulates TSP1 expression and thereby TGFβ1 activation by HG. Methods: Primary mouse MC were used. TSP1 and TGFβ1 were assessed using standard molecular biology techniques. Inhibitors of csGRP78 were: 1) vaspin, 2) the C-terminal targeting antibody C38, 3) siRNA downregulation of its transport co-chaperone MTJ-1 to prevent GRP78 translocation to the cell surface, and 4) prevention of csGRP78 activation by its ligand, active α2-macroglobulin (α2M*), with the neutralizing antibody Fα2M or an inhibitory peptide. Results: TSP1 transcript and promoter activity were increased by HG, as were cellular and ECM TSP1, and these required PI3K/Akt activity. Inhibition of csGRP78 prevented HG-induced TSP1 upregulation and deposition into the ECM. The HG-induced increase in active TGFβ1 in the medium was also inhibited, which was associated with reduced intracellular Smad3 activation and signaling. Overexpression of csGRP78 increased TSP-1, and this was further augmented in HG. Discussion: These data support an important role for csGRP78 in regulating HG-induced TSP1 transcriptional induction via PI3K/Akt signaling. Functionally, this enables TGFβ1 activation in response to HG, with consequent increase in ECM proteins. Means of inhibiting csGRP78 signaling represent a novel approach to preventing fibrosis in DKD.
Collapse
Affiliation(s)
- Jackie Trink
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Usman Ahmed
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Kian O'Neil
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Renzhong Li
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Bo Gao
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Joan C Krepinsky
- Division of Nephrology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
12
|
Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons. Int J Mol Sci 2022; 23:ijms232415936. [PMID: 36555574 PMCID: PMC9785367 DOI: 10.3390/ijms232415936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypertension is one of the most common risk factors for developing chronic cardiovascular diseases, including hypertensive nephropathy. Within the glomerulus, hypertension causes damage and activation of mesangial cells (MCs), eliciting the production of large amounts of vasoactive and proinflammatory agents. Accordingly, the activation of AT1 receptors by the vasoactive molecule angiotensin II (AngII) contributes to the pathogenesis of renal damage, which is mediated mostly by the dysfunction of intracellular Ca2+ ([Ca2+]i) signaling. Similarly, inflammation entails complex processes, where [Ca2+]i also play crucial roles. Deregulation of this second messenger increases cell damage and promotes fibrosis, reduces renal blood flow, and impairs the glomerular filtration barrier. In vertebrates, [Ca2+]i signaling depends, in part, on the activity of two families of large-pore channels: hemichannels and pannexons. Interestingly, the opening of these channels depends on [Ca2+]i signaling. In this review, we propose that the opening of channels formed by connexins and/or pannexins mediated by AngII induces the ATP release to the extracellular media, with the subsequent activation of purinergic receptors. This process could elicit Ca2+ overload and constitute a feed-forward mechanism, leading to kidney damage.
Collapse
|
13
|
Lee HE, Shim S, Choi Y, Bae YS. NADPH oxidase inhibitor development for diabetic nephropathy through water tank model. Kidney Res Clin Pract 2022; 41:S89-S98. [PMID: 35977907 PMCID: PMC9590298 DOI: 10.23876/j.krcp.21.269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 01/22/2024] Open
Abstract
Oxidative stress can cause generation of uncontrolled reactive oxygen species (ROS) and lead to cytotoxic damage to cells and tissues. Recently, it has been shown that transient ROS generation can serve as a secondary messenger in receptor-mediated cell signaling. Although excessive levels of ROS are harmful, moderated levels of ROS are essential for normal physiological function. Therefore, regulating cellular ROS levels should be an important concept for development of novel therapeutics for treating diseases. The overexpression and hyperactivation of NADPH oxidase (Nox) can induce high levels of ROS, which are strongly associated with diabetic nephropathy. This review discusses the theoretical basis for development of the Nox inhibitor as a regulator of ROS homeostasis to provide emerging therapeutic opportunities for diabetic nephropathy.
Collapse
Affiliation(s)
| | - Seunghwan Shim
- Institute of Life Science and Natural Resources, Korea University, Seoul, Republic of Korea
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yongseok Choi
- Institute of Life Science and Natural Resources, Korea University, Seoul, Republic of Korea
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yun Soo Bae
- Celros Biotech, Seoul, Republic of Korea
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Li X, Xu B, Wu J, Pu Y, Wan S, Zeng Y, Wang M, Luo L, Zhang F, Jiang Z, Xu Y. Maresin 1 Alleviates Diabetic Kidney Disease via LGR6-Mediated cAMP-SOD2-ROS Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7177889. [PMID: 35498124 PMCID: PMC9042615 DOI: 10.1155/2022/7177889] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Chronic hyperglycemia-induced inflammation is recognized as the most important pathophysiological process in diabetic kidney disease (DKD). As maresin 1 (MaR1) is an extensive anti-inflammatory lipid mediator, the present study investigated the protective role of MaR1 in the pathogenesis of DKD and its clinical relevance. METHODS Serum MaR1 concentrations were analyzed in 104 subjects with normal glucose tolerant, type 2 diabetes (T2DM), or DKD. Streptozotocin (STZ) together with high fat diet was used to induce male C57BL/6 J mice into diabetic mice which were treated with MaR1. Human renal tubule epithelial cells (HK-2 cells) were treated by high glucose for glucotoxicity cell model and transfected with LGR6 siRNA for knockdown with MaR1 added,and detected oxidative stress and inflammatory related factors. RESULTS Serum MaR1 concentrations were significant decreased in T2DM with or without kidney disease compared with normal participant and were lowest in patients with DKD. Serum MaR1 concentrations were negatively correlated with hemoglobin A1c (HbA1c), duration of diabetes, urinary albumin to creatinine ratio (UACR), neutrophil, and neutrophil-lymphocyte ratio and were positively correlated with high-density lipoprotein-cholesterol (HDL-C) and estimated glomerular filtration rate (eGFR). In mouse model, MaR1 injection alleviated hyperglycemia, UACR and the pathological progression of DKD. Interestingly, the renal expression of LGR6 was down-regulated in DKD and high glucose treated HK-2 cells but up-regulated by MaR1 treatment. Mechanistically, MaR1 alleviated inflammation via LGR6-mediated cAMP-SOD2 antioxidant pathway in DKD mice and high glucose treated HK-2 cells. CONCLUSIONS Our study demonstrates that decreased serum MaR1 levels were correlated with the development of DKD. MaR1 could alleviate DKD and glucotoxicity-induced inflammation via LGR6-mediated cAMP-SOD2 antioxidant pathway. Thus, our present findings identify MaR1 as a predictor and a potential therapeutic target for DKD.
Collapse
Affiliation(s)
- Xinyue Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Butuo Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Jing Wu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Yueli Pu
- Department of Endocrinology and Metabolism, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengrong Wan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Yan Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Mei Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Lifang Luo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Fanjie Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Zongzhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan 646000, China
| |
Collapse
|
15
|
Zhu F, Li F, Deng L, Meng F, Liang Z. Protein Interaction Network Reconstruction with a Structural Gated Attention Deep Model by Incorporating Network Structure Information. J Chem Inf Model 2022; 62:258-273. [PMID: 35005980 DOI: 10.1021/acs.jcim.1c00982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Protein-protein interactions (PPIs) provide a physical basis of molecular communications for a wide range of biological processes in living cells. Establishing the PPI network has become a fundamental but essential task for a better understanding of biological events and disease pathogenesis. Although many machine learning algorithms have been employed to predict PPIs, with only protein sequence information as the training features, these models suffer from low robustness and prediction accuracy. In this study, a new deep-learning-based framework named the Structural Gated Attention Deep (SGAD) model was proposed to improve the performance of PPI network reconstruction (PINR). The improved predictive performances were achieved by augmenting multiple protein sequence descriptors, the topological features and information flow of the PPI network, which were further implemented with a gating mechanism to improve its robustness to noise. On 11 independent test data sets and one combined data set, SGAD yielded area under the curve values of approximately 0.83-0.93, outperforming other models. Furthermore, the SGAD ensemble can learn more characteristics information on protein pairs through a two-layer neural network, serving as a powerful tool in the exploration of PPI biological space.
Collapse
Affiliation(s)
- Fei Zhu
- School of Computer Science and Technology, Soochow University, Suzhou 215 006, China
| | - Feifei Li
- School of Computer Science and Technology, Soochow University, Suzhou 215 006, China
| | - Lei Deng
- School of Computer Science and Technology, Soochow University, Suzhou 215 006, China
| | - Fanwang Meng
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Zhongjie Liang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215 006, China
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Poitras TM, Munchrath E, Zochodne DW. Neurobiological Opportunities in Diabetic Polyneuropathy. Neurotherapeutics 2021; 18:2303-2323. [PMID: 34935118 PMCID: PMC8804062 DOI: 10.1007/s13311-021-01138-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/29/2022] Open
Abstract
This review highlights a selection of potential translational directions for the treatment of diabetic polyneuropathy (DPN) currently irreversible and without approved interventions beyond pain management. The list does not include all diabetic targets that have been generated over several decades of research but focuses on newer work. The emphasis is firstly on approaches that support the viability and growth of peripheral neurons and their ability to withstand a barrage of diabetic alterations. We include a section describing Schwann cell targets and finally how mitochondrial damage has been a common element in discussing neuropathic damage. Most of the molecules and pathways described here have not yet reached clinical trials, but many trials have been negative to date. Nonetheless, these failures clear the pathway for new thoughts over reversing DPN.
Collapse
Affiliation(s)
- Trevor M Poitras
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Easton Munchrath
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Douglas W Zochodne
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
19
|
Sousa-Lima I, Kim HJ, Jones J, Kim YB. Rho-Kinase as a Therapeutic Target for Nonalcoholic Fatty Liver Diseases. Diabetes Metab J 2021; 45:655-674. [PMID: 34610720 PMCID: PMC8497927 DOI: 10.4093/dmj.2021.0197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major public health problem and the most common form of chronic liver disease, affecting 25% of the global population. Although NAFLD is closely linked with obesity, insulin resistance, and type 2 diabetes mellitus, knowledge on its pathogenesis remains incomplete. Emerging data have underscored the importance of Rho-kinase (Rho-associated coiled-coil-containing kinase [ROCK]) action in the maintenance of normal hepatic lipid homeostasis. In particular, pharmacological blockade of ROCK in hepatocytes or hepatic stellate cells prevents the progression of liver diseases such as NAFLD and fibrosis. Moreover, mice lacking hepatic ROCK1 are protected against obesity-induced fatty liver diseases by suppressing hepatic de novo lipogenesis. Here we review the roles of ROCK as an indispensable regulator of obesity-induced fatty liver disease and highlight the key cellular pathway governing hepatic lipid accumulation, with focus on de novo lipogenesis and its impact on therapeutic potential. Consequently, a comprehensive understanding of the metabolic milieu linking to liver dysfunction triggered by ROCK activation may help identify new targets for treating fatty liver diseases such as NAFLD.
Collapse
Affiliation(s)
- Inês Sousa-Lima
- CEDOC-Chronic Disease Research Center, NOVA Medical School/ Faculty of Medical Sciences, New University of Lisbon, Lisbon, Portugal
| | - Hyun Jeong Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - John Jones
- Center for Neuroscience and Cell Biology, University of Coimbra, Marquis of Pombal Square, Coimbra, Portugal
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Corresponding author: Young-Bum Kim https://orcid.org/0000-0001-9471-6330 Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA E-mail:
| |
Collapse
|
20
|
Giordo R, Ahmed YMA, Allam H, Abusnana S, Pappalardo L, Nasrallah GK, Mangoni AA, Pintus G. EndMT Regulation by Small RNAs in Diabetes-Associated Fibrotic Conditions: Potential Link With Oxidative Stress. Front Cell Dev Biol 2021; 9:683594. [PMID: 34095153 PMCID: PMC8170089 DOI: 10.3389/fcell.2021.683594] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetes-associated complications, such as retinopathy, nephropathy, cardiomyopathy, and atherosclerosis, the main consequences of long-term hyperglycemia, often lead to organ dysfunction, disability, and increased mortality. A common denominator of these complications is the myofibroblast-driven excessive deposition of extracellular matrix proteins. Although fibroblast appears to be the primary source of myofibroblasts, other cells, including endothelial cells, can generate myofibroblasts through a process known as endothelial to mesenchymal transition (EndMT). During EndMT, endothelial cells lose their typical phenotype to acquire mesenchymal features, characterized by the development of invasive and migratory abilities as well as the expression of typical mesenchymal products such as α-smooth muscle actin and type I collagen. EndMT is involved in many chronic and fibrotic diseases and appears to be regulated by complex molecular mechanisms and different signaling pathways. Recent evidence suggests that small RNAs, in particular microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are crucial mediators of EndMT. Furthermore, EndMT and miRNAs are both affected by oxidative stress, another key player in the pathophysiology of diabetic fibrotic complications. In this review, we provide an overview of the primary redox signals underpinning the diabetic-associated fibrotic process. Then, we discuss the current knowledge on the role of small RNAs in the regulation of EndMT in diabetic retinopathy, nephropathy, cardiomyopathy, and atherosclerosis and highlight potential links between oxidative stress and the dyad small RNAs-EndMT in driving these pathological states.
Collapse
Affiliation(s)
- Roberta Giordo
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Yusra M. A. Ahmed
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Hilda Allam
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Salah Abusnana
- Department of Diabetes and Endocrinology, University Hospital Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Lucia Pappalardo
- Department of Biology, Chemistry and Environmental Studies, American University of Sharjah, Sharjah, United Arab Emirates
| | - Gheyath K. Nasrallah
- Department of Biomedical Sciences, College of Health Sciences Member of QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Arduino Aleksander Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Flinders Medical Centre, Adelaide, SA, Australia
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
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
|
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
|
23
|
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
|
24
|
Tian F, Wang X, Ni H, Feng X, Yuan X, Huang Q. The ginsenoside metabolite compound K stimulates glucagon-like peptide-1 secretion in NCI-H716 cells by regulating the RhoA/ROCKs/YAP signaling pathway and cytoskeleton formation. J Pharmacol Sci 2020; 145:88-96. [PMID: 33357784 DOI: 10.1016/j.jphs.2020.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
Ginsenoside Rb1 has been shown to have antidiabetic and anti-inflammatory effects. Its major metabolite, compound K (CK), can stimulate the secretion of glucagon-like peptide-1 (GLP1), a gastrointestinal hormone that plays a vital role in regulating glucose metabolism. However, the mechanism underlying the regulation of GLP1 secretion by compound K has not been fully explored. This study was designed to investigate whether CK ameliorates incretin impairment by regulating the RhoA/ROCKs/YAP signaling pathway and cytoskeleton formation in NCI-H716 cells. Using NCI-H716 cells as a model cell line for GLP1 secretion, we analyzed the effect of CK on the expression of RhoA/ROCK/YAP pathway components. Our results suggest that the effect of CK on GLP1 secretion depends on the anti-inflammatory effect of CK. We also demonstrated that CK can affect the RhoA/ROCK/YAP pathway, which is downstream of transforming growth factor β1 (TGFβ1), by maintaining the capacity of intestinal differentiation. In addition, this effect was mediated by regulating F/G-actin dynamics. These results provide not only the mechanistic insight for the effect of CK on intestinal L cells but also the molecular basis for the further development of CK as a potential therapeutic agent to treat type 2 diabetes mellitus (T2D).
Collapse
Affiliation(s)
- Fengyuan Tian
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xi Wang
- Central Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, PR China.
| | - Haixiang Ni
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xiaohong Feng
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Xiao Yuan
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| | - Qi Huang
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, PR China.
| |
Collapse
|
25
|
Matoba K, Takeda Y, Nagai Y, Sekiguchi K, Yokota T, Utsunomiya K, Nishimura R. The Physiology, Pathology, and Therapeutic Interventions for ROCK Isoforms in Diabetic Kidney Disease. Front Pharmacol 2020; 11:585633. [PMID: 33101039 PMCID: PMC7545791 DOI: 10.3389/fphar.2020.585633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023] Open
Abstract
Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine/threonine kinase that was originally identified as RhoA interacting protein. A diverse array of cellular functions, including migration, proliferation, and phenotypic modulation, are orchestrated by ROCK through a mechanism involving cytoskeletal rearrangement. Mammalian cells express two ROCK isoforms: ROCK1 (Rho-kinase β/ROKβ) and ROCK2 (Rho-kinase α/ROKα). While both isoforms have structural similarities and are widely expressed across multiple tissues, investigations in gene knockout animals and cell-based studies have revealed distinct functions of ROCK1 and ROCK2. With respect to the kidney, inhibiting ROCK activity has proven effective for the preventing diabetic kidney disease (DKD) in both type 1 and type 2 diabetic rodent models. However, despite significant progress in the understanding of the renal ROCK biology over the past decade, the pathogenic roles of the ROCK isoforms is only beginning to be elucidated. Recent studies have demonstrated the involvement of renal ROCK1 in mitochondrial dynamics and cellular transdifferentiation, whereas ROCK2 activation leads to inflammation, fibrosis, and cell death in the diabetic kidney. This review provides a conceptual framework for dissecting the molecular underpinnings of ROCK-driven renal injury, focusing on the differences between ROCK1 and ROCK2.
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
| | - Kensuke Sekiguchi
- 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
|
26
|
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
|
27
|
Afsar B, Elsurer Afsar R, Sag AA, Kanbay A, Korkmaz H, Cipolla-Neto J, Covic A, Ortiz A, Kanbay M. Sweet dreams: therapeutic insights, targeting imaging and physiologic evidence linking sleep, melatonin and diabetic nephropathy. Clin Kidney J 2020; 13:522-530. [PMID: 32905249 PMCID: PMC7467577 DOI: 10.1093/ckj/sfz198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022] Open
Abstract
Melatonin is the main biochronologic molecular mediator of circadian rhythm and sleep. It is also a powerful antioxidant and has roles in other physiologic pathways. Melatonin deficiency is associated with metabolic derangements including glucose and cholesterol dysregulation, hypertension, disordered sleep and even cancer, likely due to altered immunity. Diabetic nephropathy (DN) is a key microvascular complication of both type 1 and 2 diabetes. DN is the end result of a complex combination of metabolic, haemodynamic, oxidative and inflammatory factors. Interestingly, these same factors have been linked to melatonin deficiency. This report will collate in a clinician-oriented fashion the mechanistic link between melatonin deficiency and factors contributing to DN.
Collapse
Affiliation(s)
- Baris Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Rengin Elsurer Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Asiye Kanbay
- Department of Pulmonary Medicine, Istanbul Medeniyet University School of Medicine, Istanbul, Turkey
| | - Hakan Korkmaz
- Division of Endocrinology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - José Cipolla-Neto
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Adrian Covic
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Nephrology Clinic, Dialysis and Renal Transplant Center, ‘C.I. PARHON’ University Hospital and ‘Grigore T. Popa’ University of Medicine, Iasi, Romania
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
28
|
Madhusudhan T, Ghosh S, Wang H, Dong W, Gupta D, Elwakiel A, Stoyanov S, Al-Dabet MM, Krishnan S, Biemann R, Nazir S, Zimmermann S, Mathew A, Gadi I, Rana R, Zeng-Brouwers J, Moeller MJ, Schaefer L, Esmon CT, Kohli S, Reiser J, Rezaie AR, Ruf W, Isermann B. Podocyte Integrin- β 3 and Activated Protein C Coordinately Restrict RhoA Signaling and Ameliorate Diabetic Nephropathy. J Am Soc Nephrol 2020; 31:1762-1780. [PMID: 32709711 DOI: 10.1681/asn.2019111163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (dNP), now the leading cause of ESKD, lacks efficient therapies. Coagulation protease-dependent signaling modulates dNP, in part via the G protein-coupled, protease-activated receptors (PARs). Specifically, the cytoprotective protease-activated protein C (aPC) protects from dNP, but the mechanisms are not clear. METHODS A combination of in vitro approaches and mouse models evaluated the role of aPC-integrin interaction and related signaling in dNP. RESULTS The zymogen protein C and aPC bind to podocyte integrin-β 3, a subunit of integrin-α v β 3. Deficiency of this integrin impairs thrombin-mediated generation of aPC on podocytes. The interaction of aPC with integrin-α v β 3 induces transient binding of integrin-β 3 with G α13 and controls PAR-dependent RhoA signaling in podocytes. Binding of aPC to integrin-β 3 via its RGD sequence is required for the temporal restriction of RhoA signaling in podocytes. In podocytes lacking integrin-β 3, aPC induces sustained RhoA activation, mimicking the effect of thrombin. In vivo, overexpression of wild-type aPC suppresses pathologic renal RhoA activation and protects against dNP. Disrupting the aPC-integrin-β 3 interaction by specifically deleting podocyte integrin-β 3 or by abolishing aPC's integrin-binding RGD sequence enhances RhoA signaling in mice with high aPC levels and abolishes aPC's nephroprotective effect. Pharmacologic inhibition of PAR1, the pivotal thrombin receptor, restricts RhoA activation and nephroprotects RGE-aPChigh and wild-type mice.Conclusions aPC-integrin-α v β 3 acts as a rheostat, controlling PAR1-dependent RhoA activation in podocytes in diabetic nephropathy. These results identify integrin-α v β 3 as an essential coreceptor for aPC that is required for nephroprotective aPC-PAR signaling in dNP.
Collapse
Affiliation(s)
- Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany .,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Sanchita Ghosh
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Hongjie Wang
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Dong
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Dheerendra Gupta
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Ahmed Elwakiel
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Stoyan Stoyanov
- German Center for Neurodegenerative Diseases, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Moh'd Mohanad Al-Dabet
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany.,Department of Medical Laboratories, Faculty of Health Sciences, American University of Madaba, Amman, Jordan
| | - Shruthi Krishnan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Ronald Biemann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Sumra Nazir
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Silke Zimmermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Akash Mathew
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Ihsan Gadi
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Rajiv Rana
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Jinyang Zeng-Brouwers
- Institute of Pharmacology, University Hospital and Goethe University, Frankfurt, Germany
| | - Marcus J Moeller
- Division of Nephrology and Immunology, University Hospital of the Rheinisch-Westfälische Technische Hochschule, Aachen University of Technology, Aachen, Germany
| | - Liliana Schaefer
- Institute of Pharmacology, University Hospital and Goethe University, Frankfurt, Germany
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Shrey Kohli
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Alireza R Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany .,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
Metabolic memory and diabetic nephropathy: Beneficial effects of natural epigenetic modifiers. Biochimie 2020; 170:140-151. [DOI: 10.1016/j.biochi.2020.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/13/2020] [Indexed: 01/04/2023]
|
34
|
Shi Y, Wang C, Zhou X, Li Y, Ma Y, Zhang R, Li R. Downregulation of PTEN promotes podocyte endocytosis of lipids aggravating obesity-related glomerulopathy. Am J Physiol Renal Physiol 2020; 318:F589-F599. [PMID: 31813249 DOI: 10.1152/ajprenal.00392.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With the increasing prevalence of obesity in adults worldwide, the incidence of obesity-related glomerulopathy (ORG) has increased yearly, becoming one of the leading causes of end-stage renal disease. Studies have demonstrated significant correlations between hyperlipidemia and impaired renal function in patients with ORG, indicating that hyperlipidemia causes damage in kidney cells. In podocytes, the endocytosis of lipids triggers an intracellular oxidative stress response that disrupts cellular integrity, resulting in proteinuria and glomerular sclerosis. However, the specific molecular mechanisms through which podocytes endocytose lipids remain unclear. Here, we demonstrated the enhanced endocytosis of lipids by podocytes from patients with ORG. This response was associated with decreased expression of phosphatase and tensin homolog (PTEN). In vitro silencing of PTEN promoted the endocytosis of low-density lipoprotein in mouse podocytes. Conversely, overexpression of PTEN inhibited the endocytosis of lipoproteins in podocytes. PTEN directly dephosphorylates and activates the actin-depolymerizing factor cofilin-1, leading to depolymerization of filamentous actin (F-actin), which is necessary for endocytosis. Notably, inhibition of PTEN resulted in the phosphorylation and inactivation of cofilin-1, leading to F-actin formation that enhanced the endocytosis of lipoproteins in podocytes. When hyperlipidemia was induced in mice with podocyte-specific deletion of PTEN, these mice recapitulated the major pathophysiological features of ORG. Thus, PTEN downregulation in podocytes may contribute to the pathogenesis of ORG.
Collapse
Affiliation(s)
- Yuanyuan Shi
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
- Nephrology Division, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chen Wang
- Pathology Division, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoshuang Zhou
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Yafeng Li
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuehong Ma
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Zhang
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| | - Rongshan Li
- Nephrology Division, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
35
|
Sharma P, Roy K. ROCK-2-selective targeting and its therapeutic outcomes. Drug Discov Today 2020; 25:446-455. [DOI: 10.1016/j.drudis.2019.11.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/08/2019] [Accepted: 11/30/2019] [Indexed: 01/21/2023]
|
36
|
Xie F, Lei J, Ran M, Li Y, Deng L, Feng J, Zhong Y, Li J. Attenuation of Diabetic Nephropathy in Diabetic Mice by Fasudil through Regulation of Macrophage Polarization. J Diabetes Res 2020; 2020:4126913. [PMID: 32685556 PMCID: PMC7345603 DOI: 10.1155/2020/4126913] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/11/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
Inflammation and fibrosis induced by hyperglycemia are considered to play a critical role in the pathogenesis of diabetic nephropathy. As macrophage polarization may determine the severity and progression of inflammation, regulation of macrophage polarization may be an effective method to treat diabetic complications. Fasudil, a potent Rho-kinase inhibitor, reportedly exhibits anti-inflammatory activity. However, whether fasudil reduces hyperglycemia-induced diabetic nephropathy via regulation of macrophage polarization remains unclear. In this study, we investigate the effect of fasudil on diabetic nephropathy in streptozotocin-induced type 1 diabetic mice. Our data showed that fasudil significantly decreased urinary protein and serum creatinine in diabetic mice, whereas it had no effect on the body weight and blood glucose. We also found increased M1-type macrophages and related proinflammatory cytokines, adverse fibrosis in renal tissue of diabetic mice. Interestingly, treatment of diabetic mice with fasudil increased the number of M2-type macrophages and related anti-inflammatory cytokines, which attenuated renal injury in diabetic mice. Taken together, the results of this study suggest that fasudil could slow the progression of diabetic nephropathy. The possible mechanism might be associated with its induction of M2 macrophage polarization and the reduction of M1 macrophage polarization and inflammation.
Collapse
Affiliation(s)
- Fajiang Xie
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jiesen Lei
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Maoxia Ran
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Yan Li
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Feng
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yi Zhong
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiafu Li
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The goal of this review is to review the role that renal parenchymal lipid accumulation plays in contributing to diabetic kidney disease (DKD), specifically contributing to the mitochondrial dysfunction observed in glomerular renal cells in the context of DKD development and progression. RECENT FINDINGS Mitochondrial dysfunction has been observed in experimental and clinical DKD. Recently, Ayanga et al. demonstrate that podocyte-specific deletion of a protein involved in mitochondrial dynamics protects from DKD progression. Furthermore, our group has recently shown that ATP-binding cassette A1 (a protein involved in cholesterol and phospholipid efflux) is significantly reduced in clinical and experimental DKD and that genetic or pharmacological induction of ABCA1 is sufficient to protect from DKD. ABCA1 deficiency in podocytes leads to mitochondrial dysfunction observed with alterations of mitochondrial lipids, in particular, cardiolipin (a mitochondrial-specific phospholipid). However, through pharmacological reduction of cardiolipin peroxidation DKD progression is reverted. Lipid metabolism is significantly altered in the diabetic kidney and renders cellular components, such as the podocyte, susceptible to injury leading to worsened DKD progression. Dysfunction of the lipid metabolism pathway can also lead to mitochondrial dysfunction and mitochondrial lipid alteration. Future research aimed at targeting mitochondrial lipids content and function could prove to be beneficial for the treatment of DKD.
Collapse
Affiliation(s)
- G Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL, USA.
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
38
|
Zhao L, Li Y, Wang Y, Qiao Z, Miao Z, Yang J, Huang L, Tian C, Li L, Chen D, Yang S. Discovery of 4H-Chromen-4-one Derivatives as a New Class of Selective Rho Kinase (ROCK) Inhibitors, which Showed Potent Activity in ex Vivo Diabetic Retinopathy Models. J Med Chem 2019; 62:10691-10710. [DOI: 10.1021/acs.jmedchem.9b01143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
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
|
40
|
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
|
41
|
Ducasa GM, Mitrofanova A, Mallela SK, Liu X, Molina J, Sloan A, Pedigo CE, Ge M, Santos JV, Hernandez Y, Kim JJ, Maugeais C, Mendez AJ, Nair V, Kretzler M, Burke GW, Nelson RG, Ishimoto Y, Inagi R, Banerjee S, Liu S, Szeto HH, Merscher S, Fontanesi F, Fornoni A. ATP-binding cassette A1 deficiency causes cardiolipin-driven mitochondrial dysfunction in podocytes. J Clin Invest 2019; 129:3387-3400. [PMID: 31329164 PMCID: PMC6668702 DOI: 10.1172/jci125316] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Fibroblasts from patients with Tangier disease carrying ATP-binding cassette A1 (ABCA1) loss-of-function mutations are characterized by cardiolipin accumulation, a mitochondrial-specific phospholipid. Suppression of ABCA1 expression occurs in glomeruli from patients with diabetic kidney disease (DKD) and in human podocytes exposed to DKD sera collected prior to the development of DKD. We demonstrated that siRNA ABCA1 knockdown in podocytes led to reduced oxygen consumption capabilities associated with alterations in the oxidative phosphorylation (OXPHOS) complexes and with cardiolipin accumulation. Podocyte-specific deletion of Abca1 (Abca1fl/fl) rendered mice susceptible to DKD, and pharmacological induction of ABCA1 improved established DKD. This was not mediated by free cholesterol, as genetic deletion of sterol-o-acyltransferase-1 (SOAT1) in Abca1fl/fl mice was sufficient to cause free cholesterol accumulation but did not cause glomerular injury. Instead, cardiolipin mediates ABCA1-dependent susceptibility to podocyte injury, as inhibition of cardiolipin peroxidation with elamipretide improved DKD in vivo and prevented ABCA1-dependent podocyte injury in vitro and in vivo. Collectively, we describe a pathway definitively linking ABCA1 deficiency to cardiolipin-driven mitochondrial dysfunction. We demonstrated that this pathway is relevant to DKD and that ABCA1 inducers or inhibitors of cardiolipin peroxidation may each represent therapeutic strategies for the treatment of established DKD.
Collapse
Affiliation(s)
- G. Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
- Department of Surgery, University of Miami, Miami, Florida, USA
| | - Shamroop K. Mallela
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Xiaochen Liu
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | | | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Javier Varona Santos
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Yanio Hernandez
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Cyrille Maugeais
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Armando J. Mendez
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Viji Nair
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - George W. Burke
- Department of Surgery, University of Miami, Miami, Florida, USA
| | | | - Yu Ishimoto
- Division of CKD Pathophysiology, University of Tokyo, Tokyo, Japan
| | - Reiko Inagi
- Division of CKD Pathophysiology, University of Tokyo, Tokyo, Japan
| | | | - Shaoyi Liu
- Social Profit Network Research Lab, Alexandria LaunchLabs, New York, New York, USA
| | - Hazel H. Szeto
- Social Profit Network Research Lab, Alexandria LaunchLabs, New York, New York, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| | - Flavia Fontanesi
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension/ Drug Discovery Center, Department of Medicine, University of Miami, Miami, Florida, USA
| |
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
|
Nabi R, Alvi SS, Saeed M, Ahmad S, Khan MS. Glycation and HMG-CoA Reductase Inhibitors: Implication in Diabetes and Associated Complications. Curr Diabetes Rev 2019; 15:213-223. [PMID: 30246643 DOI: 10.2174/1573399814666180924113442] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/17/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Diabetes Mellitus (DM) acts as an absolute mediator of cardiovascular risk, prompting the prolonged occurrence, size and intricacy of atherosclerotic plaques via enhanced Advanced Glycation Endproducts (AGEs) formation. Moreover, hyperglycemia is associated with enhanced glyco-oxidized and oxidized Low-Density Lipoprotein (LDL) possessing greater atherogenicity and decreased the ability to regulate HMG-CoA reductase (HMG-R). Although aminoguanidine (AG) prevents the AGE-induced protein cross-linking due to its anti-glycation potential, it exerts several unusual pharmaco-toxicological effects thus restraining its desirable therapeutic effects. HMG-R inhibitors/ statins exhibit a variety of beneficial impacts in addition to the cholesterol-lowering effects. OBJECTIVE Inhibition of AGEs interaction with receptor for AGEs (RAGE) and glyco-oxidized-LDL by HMG-R inhibitors could decrease LDL uptake by LDL-receptor (LDL-R), regulate cholesterol synthesis via HMG-R, decrease oxidative and inflammatory stress to improve the diabetes-associated complications. CONCLUSION Current article appraises the pathological AGE-RAGE concerns in diabetes and its associated complications, mainly focusing on the phenomenon of both circulatory AGEs and those accumulating in tissues in diabetic nephropathy, diabetic neuropathy, and diabetic retinopathy, discussing the potential protective role of HMG-R inhibitors against diabetic complications.
Collapse
Affiliation(s)
- Rabia Nabi
- Clinical Biochemistry & Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, 226026, U.P, India
| | - Sahir Sultan Alvi
- Clinical Biochemistry & Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, 226026, U.P, India
| | - Mohammad Saeed
- Department of Clinical Lab. Sciences, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Saheem Ahmad
- Laboratory of Glycation Biology and Metabolic Disorders, Department of Biosciences, Integral University Lucknow, 226026, U.P, India
| | - Mohammad Salman Khan
- Clinical Biochemistry & Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, 226026, U.P, India
| |
Collapse
|
44
|
Liu S, Zhao M, Zhou Y, Wang C, Yuan Y, Li L, Bresette W, Chen Y, Cheng J, Lu Y, Liu J. Resveratrol exerts dose-dependent anti-fibrotic or pro-fibrotic effects in kidneys: A potential risk to individuals with impaired kidney function. PHYTOMEDICINE 2018; 57:223-235. [PMID: 30785018 DOI: 10.1016/j.phymed.2018.12.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Renal fibrosis is the pathological feature of chronic kidney disease (CKD) which leads to end-stage renal disease (ESRD) and renal failure. Resveratrol [3,5,4'-trihydroxy-trans-stilbene (RSV)] has shown benefits for metabolic diseases and anti-cancer therapy, but its potential risk on renal health has not been fully evaluated. PURPOSE To investigate the global effects of RSV on renal fibrosis in human tubular epithelial cell (TEC) line HK-2, and in mice with unilateral ureteral obstruction (UUO). METHODS A TGF-β-induced in vitro model of epithelial-mesenchymal transition (EMT) in TEC was established. The effects of RSV on cell viability, pro-fibrotic factors, oxidative stress, mitochondria function, and underlying pathway proteins were analyzed. In vivo, the effects of RSV on renal function and fibrosis were assayed in UUO mice. RESULTS Our results showed that low concentrations of RSV (5-20 μM) decreased TGF-β-induced EMT via Sirt1-dependent deacetylation of Smad3/Smad4 mechanism. By contrast, long-term (72 h) exposure to high concentrations of RSV (≥ 40 μM) promoted EMT in HK-2 cells via mitochondrial oxidative stress and ROCK1-mediated disordered cytoskeleton remodeling. In vivo, low-dose treatment of RSV (≤ 25 mg/kg) partly improved renal function, whereas high-dose treatment of RSV (≥ 50 mg/kg) lost its anti-fibrotic role and even aggravated renal fibrosis. However, mice with UUO were more susceptible to high RSV-induced renal injury than normal mice. CONCLUSION Dependent on dose, RSV activated either anti-fibrotic or pro-fibrotic effects in kidneys. The risk of RSV consumption in individuals with impaired kidney function should be carefully considered.
Collapse
Affiliation(s)
- Shuyun Liu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Zhao
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yijie Zhou
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chengshi Wang
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - William Bresette
- Center for Metabolic and Vascular Biology, School for Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Scottsdale, AZ, United States
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China; Center for Metabolic and Vascular Biology, School for Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Scottsdale, AZ, United States.
| |
Collapse
|
45
|
Wei C, Banu K, Garzon F, Basgen JM, Philippe N, Yi Z, Liu R, Choudhuri J, Fribourg M, Liu T, Cumpelik A, Wong J, Khan M, Das B, Keung K, Salem F, Campbell KN, Kaufman L, Cravedi P, Zhang W, O'Connell PJ, He JC, Murphy B, Menon MC. SHROOM3-FYN Interaction Regulates Nephrin Phosphorylation and Affects Albuminuria in Allografts. J Am Soc Nephrol 2018; 29:2641-2657. [PMID: 30341149 PMCID: PMC6218856 DOI: 10.1681/asn.2018060573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/14/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We previously showed that the presence of a CKD-associated locus in SHROOM3 in a donor kidney results in increased expression of SHROOM3 (an F-actin-binding protein important for epithelial morphogenesis, via rho-kinase [ROCK] binding); this facilitates TGF-b signaling and allograft fibrosis. However, other evidence suggests Shroom3 may have a protective role in glomerular development. METHODS We used human data, Shroom3 knockdown podocytes, and inducible shRNA-mediated knockdown mice to study the role of Shroom3 in adult glomeruli. RESULTS Expression data from the Nephroseq database showed glomerular and nonglomerular SHROOM3 had opposing associations with renal function in CKD biopsy samples. In human allografts, homozygosity at rs17319721, the SHROOM3 locus linked with lower GFR, was associated with reduced albuminuria by 2 years after transplant. Although our previous data showed reduced renal fibrosis with tubular Shroom3 knockdown, this study found that glomerular but not tubular Shroom3 knockdown induced albuminuria. Electron microscopy revealed diffuse foot process effacement, and glomerular RNA-sequencing showed enrichment of tyrosine kinase signaling and podocyte actin cytoskeleton pathways in knockdown mice. Screening SHROOM3-interacting proteins identified FYN (a src-kinase) as a candidate.We confirmed the interaction of endogenous SHROOM3 with FYN in human podocytes via a critical Src homology 3-binding domain, distinct from its ROCK-binding domain. Shroom3-Fyn interaction was required in vitro and in vivo for activation of Fyn kinase and downstream nephrin phosphorylation in podocytes. SHROOM3 knockdown altered podocyte morphology, cytoskeleton, adhesion, and migration. CONCLUSIONS We demonstrate a novel mechanism that may explain SHROOM3's dichotomous associations in glomerular versus nonglomerular compartments in CKD.
Collapse
Affiliation(s)
- Chengguo Wei
- Division of Nephrology, Department of Medicine and
| | - Khadija Banu
- Division of Nephrology, Department of Medicine and
| | | | - John M Basgen
- Morphometry and Stereology Laboratory, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | | | - Zhengzi Yi
- Division of Nephrology, Department of Medicine and
| | - Ruijie Liu
- Division of Nephrology, Department of Medicine and
| | | | - Miguel Fribourg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tong Liu
- Center for Advanced Proteomics, Department of Biochemistry and Molecular Biology, New Jersey Medical School, Newark, New Jersey; and
| | | | - Jenny Wong
- Division of Nephrology, Department of Medicine and
| | - Mubeen Khan
- Division of Nephrology, Department of Medicine and
| | - Bhaskar Das
- Division of Nephrology, Department of Medicine and
| | - Karen Keung
- Renal Unit, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Fadi Salem
- Division of Nephrology, Department of Medicine and
| | | | | | | | - Weijia Zhang
- Division of Nephrology, Department of Medicine and
| | - Philip J O'Connell
- Renal Unit, University of Sydney at Westmead Hospital, Sydney, Australia
| | | | | | | |
Collapse
|
46
|
Role of MicroRNAs in Renal Parenchymal Diseases-A New Dimension. Int J Mol Sci 2018; 19:ijms19061797. [PMID: 29914215 PMCID: PMC6032378 DOI: 10.3390/ijms19061797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 11/18/2022] Open
Abstract
Since their discovery in 1993, numerous microRNAs (miRNAs) have been identified in humans and other eukaryotic organisms, and their role as key regulators of gene expression is still being elucidated. It is now known that miRNAs not only play a central role in the processes that ensure normal development and physiology, but they are often dysregulated in various diseases. In this review, we present an overview of the role of miRNAs in normal renal development and physiology, in maladaptive renal repair after injury, and in the pathogenesis of renal parenchymal diseases. In addition, we describe methods used for their detection and their potential as therapeutic targets. Continued research on renal miRNAs will undoubtedly improve our understanding of diseases affecting the kidneys and may also lead to new therapeutic agents.
Collapse
|
47
|
Tang H, Zhu M, Zhao G, Fu W, Shi Z, Ding Y, Tang X, Guo D. Loss of CLOCK under high glucose upregulates ROCK1-mediated endothelial to mesenchymal transition and aggravates plaque vulnerability. Atherosclerosis 2018; 275:58-67. [PMID: 29860109 DOI: 10.1016/j.atherosclerosis.2018.05.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/19/2018] [Accepted: 05/23/2018] [Indexed: 10/16/2022]
Abstract
BACKGROUND AND AIMS Carotid atherosclerotic plaque is one of the main sources of ischemic stroke, and endothelial-to-mesenchymal transition (EndMT) is a major feature of atherosclerosis. Rho-associated coiled-coil-containing protein kinase 1 (ROCK1) activation, stimulated by high glucose, plays an important role in EndMT, and circadian locomotor output cycles protein kaput (Clock) deficiency leads to hyperglycemia and enhanced atherosclerosis in ClockΔ19/Δ19apolipoprotein E (ApoE)-/- mice. These findings point to a mechanism whereby CLOCK exerts a protective effect against EndMT and atherosclerotic plaque accumulation. METHODS Cultured human umbilical vein endothelial cells (HUVECs) were stimulated with 66 mM glucose for 120 h to induce EndMT. The expression of CLOCK and ROCK1 was assayed, as were their effects on EndMT. We also conducted molecular and morphometric examination of carotid artery plaques from patients with carotid artery stenosis to assess the clinical relevance of these findings. RESULTS Upon EndMT, HUVECs exhibited decreased CLOCK expression and increased ROCK1 expression. Notably, CLOCK silencing increased high glucose-induced EndMT, migration ability, and ROCK1 activation, while overexpressing CLOCK attenuated these characteristics. Moreover, inhibition of ROCK1 largely blocked EndMT induced by high-glucose or transforming growth factor (TGF)-β1 but failed to rescue the reduced CLOCK expression. The vulnerability of human carotid artery plaque was strongly correlated with loss of CLOCK expression, activation of TGF-β/ROCK1 signaling, and the extent of EndMT. CONCLUSIONS The data indicate that loss of protective endothelial CLOCK expression aggravates TGF-β/ROCK1-modulated EndMT progression, which contributes to the vulnerability of human carotid plaque.
Collapse
Affiliation(s)
- Hanfei Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengjiao Zhu
- Department of Orthodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Gefei Zhao
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenyu Shi
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Ding
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Daqiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Xiao YH, He XY, Han Q, Yang F, Zhou SX. Atorvastatin prevents glomerular extracellular matrix formation by interfering with the PKC signaling pathway. Mol Med Rep 2018; 17:6441-6448. [PMID: 29532876 PMCID: PMC5928626 DOI: 10.3892/mmr.2018.8724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 01/25/2018] [Indexed: 01/21/2023] Open
Abstract
Platelet-activating factor (PAF) promotes glomerular extracellular matrix (ECM) deposition, primarily through activation of the protein kinase C (PKC) pathway. The present study was designed to investigate whether atorvastatin, which mediates a protective effect against glomerular ECM deposition and diabetic neuropathy, may interfere with the PKC‑transforming growth factor‑β1 (TGF‑β1) pathway in a model of human mesangial cells (HMCs) exposed to a high glucose (HG) and lysophosphatidylcholine (LPC) environment. HMCs were divided into three treatment groups: Control, high glucose and lysophosphatidylcholine (HG+LPC), and HG+LPC+atorvastatin. Cells were cultured for 24 h. The levels of the ECM‑associated molecules collagen IV (Col IV) and fibronectin (Fn) in the supernatant were detected using an ELISA kit. PKC‑β1, TGF‑β1 and PAF‑receptor gene expression was detected by reverse transcription‑quantitative polymerase chain reaction. PKC‑β1 and TGF‑β1 protein expression was detected by western blotting, and the subcellular localization of PKC‑β1 was assessed using immunofluorescence. The results indicated that atorvastatin may reduce the secretion of ECM components (Fn and Col IV) in HMCs in a HG and LPC environment, by inhibiting the increase in PAF secretion and the activation of the PKC‑TGF‑β1 signaling pathway.
Collapse
Affiliation(s)
- Yan-Hua Xiao
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Xiao-Yun He
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Qing Han
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Fan Yang
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Su-Xian Zhou
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| |
Collapse
|
49
|
Gao Z, Zhu W, Zhang H, Li Z, Cui T. The influence of fasudil on renal proximal tubular cell epithelial-mesenchymal transition induced by parathormone. Ren Fail 2018; 39:575-581. [PMID: 28741985 PMCID: PMC6446168 DOI: 10.1080/0886022x.2017.1349677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND Renal fibrosis is a common pathway through which a variety of chronic kidney diseases progress to end-stage renal disease. Epithelial-mesenchymal transition (EMT) of renal proximal tubular cells is one of the most important factors in renal fibrosis. This study investigates if fasudil could influence EMT of renal proximal tubular cells. METHODS HK-2 cells in passage 3-4 were used for all experiments. The cells were divided into five groups and treated with different concentrations of PTH and then observe cellular morphological changes at 0, 24 and 48 h using an inverted microscope and investigate the expression of the epithelial cell marker E-cadherin and the renal fibroblast marker α-smooth muscle actin (α-SMA). RESULTS PTH significantly induced EMT, fasudil-inhibited EMT induced by PTH to different degrees, and the inhibitory effect of fasudil was most pronounced at 20 μmol/L. CONCLUSION Monitoring PTH levels, early prevention and control of hyperparathyroidism and reducing the concentration of PTH are important means to improve prognosis and delay the progression of chronic kidney disease. Fasudil can restrain EMT induced by PTH; this conclusion provides experimental data for the application of fasudil in the clinical prevention and treatment of renal fibrosis.
Collapse
Affiliation(s)
- Ziqing Gao
- a Department of Ultrasound , the Fifth Affiliated Hospital of Sun Yat-Sen University , Zhuhai , China
| | - Weiping Zhu
- b Department of Nephrology , the Fifth Affiliated Hospital of Sun Yat-Sen University , Zhuhai , China
| | - Hua Zhang
- b Department of Nephrology , the Fifth Affiliated Hospital of Sun Yat-Sen University , Zhuhai , China
| | - Zhonghe Li
- b Department of Nephrology , the Fifth Affiliated Hospital of Sun Yat-Sen University , Zhuhai , China
| | - Tongxia Cui
- b Department of Nephrology , the Fifth Affiliated Hospital of Sun Yat-Sen University , Zhuhai , China
| |
Collapse
|
50
|
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
|