1
|
Patel K, Barter M, Soul J, Clark P, Proctor C, Clark I, Young D, Shanley DP. Systems analysis of miR-199a/b-5p and multiple miR-199a/b-5p targets during chondrogenesis. eLife 2024; 12:RP89701. [PMID: 39401064 PMCID: PMC11473111 DOI: 10.7554/elife.89701] [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] [Indexed: 10/15/2024] Open
Abstract
Changes in chondrocyte gene expression can contribute to the development of osteoarthritis (OA), and so recognition of the regulative processes during chondrogenesis can lead to a better understanding of OA. microRNAs (miRNAs) are key regulators of gene expression in chondrocytes/OA, and we have used a combined experimental, bioinformatic, and systems biology approach to explore the multiple miRNA-mRNA interactions that regulate chondrogenesis. A longitudinal chondrogenesis bioinformatic analysis identified paralogues miR-199a-5p and miR-199b-5p as pro-chondrogenic regulators. Experimental work in human cells demonstrated alteration of miR-199a-5p or miR-199b-5p expression led to significant inverse modulation of key chondrogenic genes and extracellular matrix production. miR-199a/b-5p targets FZD6, ITGA3 and CAV1 were identified by inhibition experiments and verified as direct targets by luciferase assay. The experimental work was used to generate and parameterise a multi-miRNA 14-day chondrogenesis kinetic model to be used as a repository for the experimental work and as a resource for further investigation of this system. This is the first multi-miRNA model of a chondrogenesis-based system, and highlights the complex relationships between regulatory miRNAs, and their target mRNAs.
Collapse
Affiliation(s)
- Krutik Patel
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Matt Barter
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Jamie Soul
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
- Computational Biology Facility, Faculty of Health and Life Sciences, University of LiverpoolLiverpoolUnited Kingdom
| | - Peter Clark
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Carole Proctor
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Ian Clark
- School of Biological Sciences, University of East AngliaNorwichUnited Kingdom
| | - David Young
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Daryl P Shanley
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| |
Collapse
|
2
|
Zarkada G, Chen X, Zhou X, Lange M, Zeng L, Lv W, Zhang X, Li Y, Zhou W, Liu K, Chen D, Ricard N, Liao JK, Kim YB, Benedito R, Claesson-Welsh L, Alitalo K, Simons M, Ju R, Li X, Eichmann A, Zhang F. Chylomicrons Regulate Lacteal Permeability and Intestinal Lipid Absorption. Circ Res 2023; 133:333-349. [PMID: 37462027 PMCID: PMC10530007 DOI: 10.1161/circresaha.123.322607] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Lymphatic vessels are responsible for tissue drainage, and their malfunction is associated with chronic diseases. Lymph uptake occurs via specialized open cell-cell junctions between capillary lymphatic endothelial cells (LECs), whereas closed junctions in collecting LECs prevent lymph leakage. LEC junctions are known to dynamically remodel in development and disease, but how lymphatic permeability is regulated remains poorly understood. METHODS We used various genetically engineered mouse models in combination with cellular, biochemical, and molecular biology approaches to elucidate the signaling pathways regulating junction morphology and function in lymphatic capillaries. RESULTS By studying the permeability of intestinal lacteal capillaries to lipoprotein particles known as chylomicrons, we show that ROCK (Rho-associated kinase)-dependent cytoskeletal contractility is a fundamental mechanism of LEC permeability regulation. We show that chylomicron-derived lipids trigger neonatal lacteal junction opening via ROCK-dependent contraction of junction-anchored stress fibers. LEC-specific ROCK deletion abolished junction opening and plasma lipid uptake. Chylomicrons additionally inhibited VEGF (vascular endothelial growth factor)-A signaling. We show that VEGF-A antagonizes LEC junction opening via VEGFR (VEGF receptor) 2 and VEGFR3-dependent PI3K (phosphatidylinositol 3-kinase)/AKT (protein kinase B) activation of the small GTPase RAC1 (Rac family small GTPase 1), thereby restricting RhoA (Ras homolog family member A)/ROCK-mediated cytoskeleton contraction. CONCLUSIONS Our results reveal that antagonistic inputs into ROCK-dependent cytoskeleton contractions regulate the interconversion of lymphatic junctions in the intestine and in other tissues, providing a tunable mechanism to control the lymphatic barrier.
Collapse
Affiliation(s)
- Georgia Zarkada
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Xun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuetong Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Martin Lange
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Lei Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Wenyu Lv
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yunhua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Weibin Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Keli Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Dongying Chen
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Nicolas Ricard
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - James K. Liao
- University of Arizona, College of Medicine, Banner University Medical Center, Tucson, AZ, 85724, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid E28029, Spain
| | - Lena Claesson-Welsh
- Uppsala University, Rudbeck, SciLifeLab and Beijer Laboratories, Department of Immunology, Genetics and Pathology, 751 85 Uppsala, Sweden
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum, University of Helsinki, Finland
| | - Michael Simons
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510-3221, USA
- INSERM U970, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| |
Collapse
|
3
|
Agarwal R, Iezhitsa I. Advances in targeting the extracellular matrix for glaucoma therapy: current updates. Expert Opin Ther Targets 2023; 27:1217-1229. [PMID: 38069479 DOI: 10.1080/14728222.2023.2293748] [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: 10/11/2023] [Accepted: 12/07/2023] [Indexed: 12/31/2023]
Abstract
INTRODUCTION Elevated intraocular pressure (IOP) is a well-recognized risk factor for development of primary open angle glaucoma (POAG), a leading cause of irreversible blindness. Ocular hypertension is associated with excessive extracellular matrix (ECM) deposition in trabecular meshwork (TM) resulting in increased aqueous outflow resistance and elevated IOP. Hence, therapeutic options targeting ECM remodeling in TM to lower IOP in glaucomatous eyes are of considerable importance. AREAS COVERED This paper discusses the complex process of ECM regulation in TM and explores promising therapeutic targets. The role of Transforming Growth Factor-β as a central player in ECM deposition in TM is discussed. We elaborate the key regulatory processes involved in its activation, release, signaling, and cross talk with other signaling pathways including Rho GTPase, Wnt, integrin, cytokines, and renin-angiotensin-aldosterone. Further, we summarize the therapeutic agents that have been explored to target ECM dysregulation in TM. EXPERT OPINION Targeting molecular pathways to reduce ECM deposition and/or enhance its degradation are of considerable significance for IOP lowering. Challenges lie in pinpointing specific targets and designing drug delivery systems to precisely interact with pathologically active/inactive signaling. Recent advances in monoclonal antibodies, fusion molecules, and vectored nanotechnology offer potential solutions.
Collapse
Affiliation(s)
- Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Wu JC, Wang XJ, Zhu JH, Huang XY, Liu M, Qiao Z, Zhang Y, Sun Y, Wang ZY, Zhan P, Zhang T, Hu HL, Liu H, Tang W, Yi F. GPR97 deficiency ameliorates renal interstitial fibrosis in mouse hypertensive nephropathy. Acta Pharmacol Sin 2023; 44:1206-1216. [PMID: 36635422 PMCID: PMC10203364 DOI: 10.1038/s41401-022-01041-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Hypertensive nephropathy (HTN) ranks as the second-leading cause of end-stage renal disease (ESRD). Accumulating evidence suggests that persistent hypertension injures tubular cells, leading to tubulointerstitial fibrosis (TIF), which is involved in the pathogenesis of HTN. G protein-coupled receptors (GPCRs) are implicated in many important pathological and physiological processes and act as important drug targets. In this study, we explored the intrarenal mechanisms underlying hypertension-associated TIF, and particularly, the potential role of GPR97, a member of the adhesion GPCR subfamily, in TIF. A deoxycorticosterone acetate (DOCA)/salt-induced hypertensive mouse model was used. We revealed a significantly upregulated expression of GPR97 in the kidneys, especially in renal tubules, of the hypertensive mice and 10 patients with biopsy-proven hypertensive kidney injury. GPR97-/- mice showed markedly elevated blood pressure, which was comparable to that of wild-type mice following DOCA/salt treatment, but dramatically ameliorated renal injury and TIF. In NRK-52E cells, we demonstrated that knockdown of GPR97 suppressed the activation of TGF-β signaling by disturbing small GTPase RhoA-mediated cytoskeletal reorganization, thus inhibiting clathrin-mediated endocytosis of TGF-β receptors and subsequent Smad activation. Collectively, this study demonstrates that GPR97 contributes to hypertension-associated TIF at least in part by facilitating TGF-β signaling, suggesting that GPR97 is a pivotal intrarenal factor for TIF progression under hypertensive conditions, and therapeutic strategies targeting GPR97 may improve the outcomes of patients with HTN.
Collapse
Affiliation(s)
- Ji-Chao Wu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xiao-Jie Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Jing-Han Zhu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xue-Ying Huang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Min Liu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Zhe Qiao
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yan Zhang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yu Sun
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Zi-Ying Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, 250012, China
| | - Hui-Li Hu
- Department of Systems Biomedicine and Research Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250012, China
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| |
Collapse
|
5
|
De Ieso ML, Kuhn M, Bernatchez P, Elliott MH, Stamer WD. A Role of Caveolae in Trabecular Meshwork Mechanosensing and Contractile Tone. Front Cell Dev Biol 2022; 10:855097. [PMID: 35372369 PMCID: PMC8969750 DOI: 10.3389/fcell.2022.855097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Polymorphisms in the CAV1/2 gene loci impart increased risk for primary open-angle glaucoma (POAG). CAV1 encodes caveolin-1 (Cav1), which is required for biosynthesis of plasma membrane invaginations called caveolae. Cav1 knockout mice exhibit elevated intraocular pressure (IOP) and decreased outflow facility, but the mechanistic role of Cav1 in IOP homeostasis is unknown. We hypothesized that caveolae sequester/inhibit RhoA, to regulate trabecular meshwork (TM) mechanosensing and contractile tone. Using phosphorylated myosin light chain (pMLC) as a surrogate indicator for Rho/ROCK activity and contractile tone, we found that pMLC was elevated in Cav1-deficient TM cells compared to control (131 ± 10%, n = 10, p = 0.016). Elevation of pMLC levels following Cav1 knockdown occurred in cells on a soft surface (137 ± 7%, n = 24, p < 0.0001), but not on a hard surface (122 ± 17%, n = 12, p = 0.22). In Cav1-deficient TM cells where pMLC was elevated, Rho activity was also increased (123 ± 7%, n = 6, p = 0.017), suggesting activation of the Rho/ROCK pathway. Cyclic stretch reduced pMLC/MLC levels in TM cells (69 ± 7% n = 9, p = 0.002) and in Cav1-deficient TM cells, although not significantly (77 ± 11% n = 10, p = 0.059). Treatment with the Cav1 scaffolding domain mimetic, cavtratin (1 μM) caused a reduction in pMLC (70 ± 5% n = 7, p = 0.001), as did treatment with the scaffolding domain mutant cavnoxin (1 μM) (82 ± 7% n = 7, p = 0.04). Data suggest that caveolae differentially regulate RhoA signaling, and that caveolae participate in TM mechanotransduction. Cav1 regulation of these key TM functions provide evidence for underlying mechanisms linking polymorphisms in the Cav1/2 gene loci with increased POAG risk.
Collapse
Affiliation(s)
- Michael L. De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| | - Megan Kuhn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Heart + Lung Innovation Centre, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Michael H. Elliott
- Department of Ophthalmology, Dean McGee Eye Institute University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Chen IT, Huang LT, Chen CC, Chen CM. Molecular mechanisms underlying hyperoxia-induced lung fibrosis. Pediatr Neonatol 2022; 63:109-116. [PMID: 35181258 DOI: 10.1016/j.pedneo.2021.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 11/24/2022] Open
Abstract
Supplemental oxygen is often used to treat newborns with respiratory disorders. Exposure to high concentration of oxygen and long-term oxygen causes inflammation and acute lung injury. The acute inflammatory phase is followed by a fibroproliferative repair phase, leading to lung fibrosis. Many infants with lung fibrosis develop significant respiratory morbidities including reactive airways dysfunction and obstructive lung disease during childhood. Despite the absence of effective treatments and the incomplete understanding regarding mechanisms underlying fibrosis, extensive literature regarding lung fibrosis from in vitro and in vivo hyperoxia-exposed models is available. In this review, we discuss molecular mediators and signaling pathways responsible for increased fibroblast proliferation and collagen production, excessive extracellular matrix accumulation, and eventually, lung fibrosis. We discuss each of these mediators separately to facilitate clear understanding as well as significant interactions occurring among these molecular mediators and signaling pathways.
Collapse
Affiliation(s)
- I-Ting Chen
- Division of Neonatology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Liang-Ti Huang
- Department of Pediatrics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Cheng Chen
- Division of Neonatology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
7
|
Activin A and Cell-Surface GRP78 Are Novel Targetable RhoA Activators for Diabetic Kidney Disease. Int J Mol Sci 2021; 22:ijms22062839. [PMID: 33799579 PMCID: PMC8000060 DOI: 10.3390/ijms22062839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of kidney failure. RhoA/Rho-associated protein kinase (ROCK) signaling is a recognized mediator of its pathogenesis, largely through mediating the profibrotic response. While RhoA activation is not feasible due to the central role it plays in normal physiology, ROCK inhibition has been found to be effective in attenuating DKD in preclinical models. However, this has not been evaluated in clinical studies as of yet. Alternate means of inhibiting RhoA/ROCK signaling involve the identification of disease-specific activators. This report presents evidence showing the activation of RhoA/ROCK signaling both in vitro in glomerular mesangial cells and in vivo in diabetic kidneys by two recently described novel pathogenic mediators of fibrosis in DKD, activins and cell-surface GRP78. Neither are present in normal kidneys. Activin inhibition with follistatin and neutralization of cell-surface GRP78 using a specific antibody blocked RhoA activation in mesangial cells and in diabetic kidneys. These data identify two novel RhoA/ROCK activators in diabetic kidneys that can be evaluated for their efficacy in inhibiting the progression of DKD.
Collapse
|
8
|
Mehta N, Li R, Zhang D, Soomro A, He J, Zhang I, MacDonald M, Gao B, Krepinsky JC. miR299a-5p promotes renal fibrosis by suppressing the antifibrotic actions of follistatin. Sci Rep 2021; 11:88. [PMID: 33420269 PMCID: PMC7794215 DOI: 10.1038/s41598-020-80199-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Abstract
Caveolin-1 (cav-1), an integral protein of the membrane microdomains caveolae, is required for synthesis of matrix proteins by glomerular mesangial cells (MC). Previously, we demonstrated that the antifibrotic protein follistatin (FST) is transcriptionally upregulated in cav-1 knockout MC and that its administration is protective against renal fibrosis. Here, we screened cav-1 wild-type and knockout MC for FST-targeting microRNAs in order to identity novel antifibrotic therapeutic targets. We identified that miR299a-5p was significantly suppressed in cav-1 knockout MC, and this was associated with stabilization of the FST 3'UTR. Overexpression and inhibition studies confirmed the role of miR299a-5p in regulating FST expression. Furthermore, the profibrotic cytokine TGFβ1 was found to stimulate the expression of miR299a-5p and, in turn, downregulate FST. Through inhibition of FST, miR299a-5p overexpression augmented, while miR299a-5p inhibition diminished TGFβ1 profibrotic responses, whereas miR299a-5p overexpression re-enabled cav-1 knockout MC to respond to TGFβ1. In vivo, miR299a-5p was upregulated in the kidneys of mice with chronic kidney disease (CKD). miR299a-5p inhibition protected these mice against renal fibrosis and CKD severity. Our data demonstrate that miR299a-5p is an important post-transcriptional regulator of FST, with its upregulation an important pathogenic contributor to renal fibrosis. Thus, miR299a-5p inhibition offers a potential novel therapeutic approach for CKD.
Collapse
Affiliation(s)
- Neel Mehta
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Asfia Soomro
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Juehua He
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Ivan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Melissa MacDonald
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Bo Gao
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada.
- St. Joseph's Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
9
|
Libring S, Shinde A, Chanda MK, Nuru M, George H, Saleh AM, Abdullah A, Kinzer-Ursem TL, Calve S, Wendt MK, Solorio L. The Dynamic Relationship of Breast Cancer Cells and Fibroblasts in Fibronectin Accumulation at Primary and Metastatic Tumor Sites. Cancers (Basel) 2020; 12:E1270. [PMID: 32429591 PMCID: PMC7281295 DOI: 10.3390/cancers12051270] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
In breast cancer (BC), tissue stiffening via fibronectin (FN) and collagen accumulation is associated with advanced disease progression at both the primary tumor and metastatic sites. Here, we evaluate FN production in 15 BC cell lines, representing a variety of subtypes, phenotypes, metastatic potentials, and chemotherapeutic sensitivities. We demonstrate that intracellular and soluble FN is initially lost during tumorigenic transformation but is rescued in all lines with epithelial-mesenchymal plasticity (EMP). Importantly, we establish that no BC cell line was able to independently organize a robust FN matrix. Non-transformed mammary epithelial cells were also unable to deposit FN matrices unless transglutaminase 2, a FN crosslinking enzyme, was overexpressed. Instead, BC cells manipulated the FN matrix production of fibroblasts in a phenotypic-dependent manner. In addition, varied accumulation levels were seen depending if the fibroblasts were conditioned to model paracrine signaling or endocrine signaling of the metastatic niche. In the former, fibroblasts conditioned by BC cultures with high EMP resulted in the largest FN matrix accumulation. In contrast, mesenchymal BC cells produced extracellular vesicles (EV) that resulted in the highest levels of matrix formation by conditioned fibroblasts. Overall, we demonstrate a dynamic relationship between tumor and stromal cells within the tumor microenvironment, in which the levels and fibrillarization of FN in the extracellular matrix are modulated during the particular stages of disease progression.
Collapse
Affiliation(s)
- Sarah Libring
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Aparna Shinde
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (A.S.); (A.A.)
| | - Monica K. Chanda
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Maryam Nuru
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Heather George
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Aya M. Saleh
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Ammara Abdullah
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (A.S.); (A.A.)
| | - Tamara L. Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
| | - Michael K. Wendt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (A.S.); (A.A.)
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; (S.L.); (M.K.C.); (M.N.); (H.G.); (A.M.S.); (T.L.K.-U.); (S.C.)
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
10
|
Lyu Y, Xu W, Zhang J, Li M, Xiang Q, Li Y, Tan T, Ou Q, Zhang J, Tian H, Xu JY, Jin C, Gao F, Wang J, Li W, Rong A, Lu L, Xu GT. Protein Kinase A Inhibitor H89 Attenuates Experimental Proliferative Vitreoretinopathy. Invest Ophthalmol Vis Sci 2020; 61:1. [PMID: 32031573 PMCID: PMC7325625 DOI: 10.1167/iovs.61.2.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 11/11/2019] [Indexed: 01/09/2023] Open
Abstract
PURPOSE This study aimed to explore the role of the protein kinase A (PKA) pathway in proliferative vitreoretinopathy (PVR) and the effect of the PKA inhibitor H89 on experimental PVR. METHODS Epiretinal membranes (ERMs) were acquired from PVR patients and analyzed by frozen-section immunofluorescence. An in vivo model was developed by intravitreal injecting rat eyes with ARPE-19 cells and platelet-rich plasma, and changes in eye structures and vision function were observed. An in vitro epithelial-mesenchymal transition (EMT) cell model was established by stimulating ARPE-19 cells with transforming growth factor (TGF)-β. Alterations in EMT-related genes and cell function were detected. Mechanistically, PKA activation and activity were explored to assess the relationship between TGF-β1 stimulation and the PKA pathway. The effect of H89 on the TGF-β-Smad2/3 pathway was detected. RNA sequencing was used to analyze gene expression profile changes after H89 treatment. RESULTS PKA was activated in human PVR membranes. In vivo, H89 treatment protected against structural changes in the retina and prevented decreases in electroretinogram b-wave amplitudes. In vitro, H89 treatment inhibited EMT-related gene alterations and partially reversed the functions of the cells. TGF-β-induced PKA activation was blocked by H89 pretreatment. H89 did not affect the phosphorylation or nuclear translocation of regulatory Smad2/3 but increased the expression of inhibitory Smad6. CONCLUSIONS PKA pathway activation is involved in PVR pathogenesis, and the PKA inhibitor H89 can effectively inhibit PVR, both in vivo and in vitro. Furthermore, the protective effect of H89 is related to an increase in inhibitory Smad6.
Collapse
Affiliation(s)
- Yali Lyu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Ophthalmology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jieping Zhang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Mengwen Li
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Qingyi Xiang
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Yao Li
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Tianhao Tan
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Caixia Jin
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Juan Wang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Weiye Li
- Department of Ophthalmology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ao Rong
- Department of Ophthalmology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Laboratory of Clinical Visual Science of Tongji Eye Institute, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
- Department of Regenerative Medicine, and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| |
Collapse
|
11
|
Haddad D, Al Madhoun A, Nizam R, Al-Mulla F. Role of Caveolin-1 in Diabetes and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9761539. [PMID: 32082483 PMCID: PMC7007939 DOI: 10.1155/2020/9761539] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/10/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
It is estimated that in 2017 there were 451 million people with diabetes worldwide. These figures are expected to increase to 693 million by 2045; thus, innovative preventative programs and treatments are a necessity to fight this escalating pandemic disorder. Caveolin-1 (CAV1), an integral membrane protein, is the principal component of caveolae in membranes and is involved in multiple cellular functions such as endocytosis, cholesterol homeostasis, signal transduction, and mechanoprotection. Previous studies demonstrated that CAV1 is critical for insulin receptor-mediated signaling, insulin secretion, and potentially the development of insulin resistance. Here, we summarize the recent progress on the role of CAV1 in diabetes and diabetic complications.
Collapse
Affiliation(s)
- Dania Haddad
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
12
|
The caveolin-1 regulated protein follistatin protects against diabetic kidney disease. Kidney Int 2019; 96:1134-1149. [DOI: 10.1016/j.kint.2019.05.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/27/2019] [Accepted: 05/30/2019] [Indexed: 01/30/2023]
|
13
|
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
|
14
|
Ramírez CM, Zhang X, Bandyopadhyay C, Rotllan N, Sugiyama MG, Aryal B, Liu X, He S, Kraehling JR, Ulrich V, Lin CS, Velazquez H, Lasunción MA, Li G, Suárez Y, Tellides G, Swirski FK, Lee WL, Schwartz MA, Sessa WC, Fernández-Hernando C. Caveolin-1 Regulates Atherogenesis by Attenuating Low-Density Lipoprotein Transcytosis and Vascular Inflammation Independently of Endothelial Nitric Oxide Synthase Activation. Circulation 2019; 140:225-239. [PMID: 31154825 DOI: 10.1161/circulationaha.118.038571] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Atherosclerosis is driven by synergistic interactions between pathological, biomechanical, inflammatory, and lipid metabolic factors. Our previous studies demonstrated that absence of caveolin-1 (Cav1)/caveolae in hyperlipidemic mice strongly inhibits atherosclerosis, which was attributed to activation of endothelial nitric oxide (NO) synthase (eNOS) and increased production of NO and reduced inflammation and low-density lipoprotein trafficking. However, the contribution of eNOS activation and NO production in the athero-protection of Cav1 and the exact mechanisms by which Cav1/caveolae control the pathogenesis of diet-induced atherosclerosis are still not clear. METHODS Triple-knockout mouse lacking expression of eNOS, Cav1, and Ldlr were generated to explore the role of NO production in Cav1-dependent athero-protective function. The effects of Cav1 on lipid trafficking, extracellular matrix remodeling, and vascular inflammation were studied both in vitro and in vivo with a mouse model of diet-induced atherosclerosis. The expression of Cav1 and distribution of caveolae regulated by flow were analyzed by immunofluorescence staining and transmission electron microscopy. RESULTS We found that absence of Cav1 significantly suppressed atherogenesis in Ldlr-/-eNOS-/- mice, demonstrating that athero-suppression is independent of increased NO production. Instead, we find that the absence of Cav1/caveolae inhibited low-density lipoprotein transport across the endothelium and proatherogenic fibronectin deposition and disturbed flow-mediated endothelial cell inflammation. Consistent with the idea that Cav1/caveolae may play a role in early flow-dependent inflammatory priming, distinct patterns of Cav1 expression and caveolae distribution were observed in athero-prone and athero-resistant areas of the aortic arch even in wild-type mice. CONCLUSIONS These findings support a role for Cav1/caveolae as a central regulator of atherosclerosis that links biomechanical, metabolic, and inflammatory pathways independently of endothelial eNOS activation and NO production.
Collapse
Affiliation(s)
- Cristina M Ramírez
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Chirosree Bandyopadhyay
- Cardiovascular Research Center, Department of Internal Medicine and Cell Biology (C.B., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Michael G Sugiyama
- Keenan Research Centre and Departments of Laboratory Medicine and Pathobiology, Biochemistry and Medicine, University of Toronto, ON, Canada (M.G.S., W.L.L.)
| | - Binod Aryal
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Xinran Liu
- Department of Cell Biology (X.L.), Yale University School of Medicine, New Haven, CT
| | - Shun He
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston (S.H., F.K.S.)
| | - Jan R Kraehling
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Victoria Ulrich
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Chin Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.S.L.)
| | - Heino Velazquez
- Section of Nephrology (H.V.), Yale University School of Medicine, New Haven, CT
| | - Miguel A Lasunción
- Cardiovascular Research Center, Department of Internal Medicine and Cell Biology (C.B., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Guangxin Li
- Departments of Cell Biology and Biomedical Engineering (G.L., G.T.), Yale University School of Medicine, New Haven, CT.,Department of Surgery (G.L., G.T.), Yale University School of Medicine, New Haven, CT
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - George Tellides
- Departments of Cell Biology and Biomedical Engineering (G.L., G.T.), Yale University School of Medicine, New Haven, CT.,Department of Surgery (G.L., G.T.), Yale University School of Medicine, New Haven, CT
| | - Filip K Swirski
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston (S.H., F.K.S.)
| | - Warren L Lee
- Keenan Research Centre and Departments of Laboratory Medicine and Pathobiology, Biochemistry and Medicine, University of Toronto, ON, Canada (M.G.S., W.L.L.)
| | - Martin A Schwartz
- Department of Cell Biology (M.A.S.), Yale University School of Medicine, New Haven, CT.,Departamento de Bioquímica-Investigación, Hospital Ramón y Cajal, IRyCIS, Madrid, Spain (M.A.L.).,CIBER de Fisiopatología de la Obesidad y Nutrición, ISCIII, Madrid, Spain (M.A.L.)
| | - William C Sessa
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program (C.M.R., X.Z., N.R., B.A., J.R.K., V.U., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (C.M.R., X.Z., N.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
15
|
Mehta N, Zhang D, Li R, Wang T, Gava A, Parthasarathy P, Gao B, Krepinsky JC. Caveolin-1 regulation of Sp1 controls production of the antifibrotic protein follistatin in kidney mesangial cells. Cell Commun Signal 2019; 17:37. [PMID: 30995923 PMCID: PMC6472091 DOI: 10.1186/s12964-019-0351-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND We previously showed that caveolin-1 (cav-1), an integral membrane protein, is required for the synthesis of matrix proteins by glomerular mesangial cells (MC). In a previous study to understand how cav-1 is involved in regulating matrix production, we had identified significant upregulation of the antifibrotic protein follistatin in cav-1 knockout MC. Follistatin inhibits the profibrotic effects of several members of the transforming growth factor beta superfamily, in particular the activins. Here, we characterize the molecular mechanism through which cav-1 regulates the expression of follistatin. METHODS Kidneys from cav-1 wild type and knockout (KO) mice were analyzed and primary cultures of MC from cav-1 wild-type and KO mice were utilized. FST promoter deletion constructs were generated to determine the region of the promoter important for mediating FST upregulation in cav-1 KO MC. siRNA-mediated down-regulation and overexpression of Sp1 in conjunction with luciferase activity assays, immunoprecipitation, western blotting and ChiP was used to assess the role of Sp1 in transcriptionally regulating FST expression. Pharmacologic kinase inhibitors and specific siRNA were used to determine the post-translational mechanism through which cav-1 affects Sp1 activity. RESULTS Our results establish that follistatin upregulation occurs at the transcript level. We identified Sp1 as the critical transcription factor regulating activation of the FST promoter in cav-1 KO MC through binding to a region within 123 bp of the transcription start site. We further determined that the lack of cav-1 increases Sp1 nuclear levels and transcriptional activity. This occurred through increased phosphoinositide 3-kinase (PI3K) activity and downstream protein kinase C (PKC) zeta-mediated phosphorylation and activation of Sp1. CONCLUSIONS These findings shed light on the transcriptional mechanism by which cav-1 represses the expression of a major antifibrotic protein, and can inform the development of novel antifibrotic treatment strategies.
Collapse
Affiliation(s)
- Neel Mehta
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Tony Wang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Agata Gava
- Physiological Sciences Graduate Program, Health Sciences Centre, Federal University of Espirito Santo, Vitoria, Brazil
| | | | - Bo Gao
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada. .,St. Joseph's Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
16
|
Hill LJ, Mead B, Thomas CN, Foale S, Feinstein E, Berry M, Blanch RJ, Ahmed Z, Logan A. TGF-β-induced IOP elevations are mediated by RhoA in the early but not the late fibrotic phase of open angle glaucoma. Mol Vis 2018; 24:712-726. [PMID: 30429640 PMCID: PMC6205807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 10/25/2018] [Indexed: 12/03/2022] Open
Abstract
Purpose Elevations in intraocular pressure (IOP) are associated with the development of glaucoma and loss of sight. High transforming growth factor-β (TGF-β) 1 levels in the eye's anterior chamber can lead to dysfunctional contractions through RhoA signaling in trabecular meshwork (TM) cells and IOP spikes. Sustained high TGF-β levels leads to TM fibrosis and sustained increases in IOP. We investigated whether inhibiting RhoA, using a siRNA-mediated RhoA (siRhoA), controls IOP by altering TM expression of fibrosis and contractility-related proteins in a rodent model of glaucoma. Methods TGF-β was injected intracamerally twice a week into adult Sprague Dawley rats, and IOP was recorded with tonometry. Animals were euthanized on day 7 and 35 with TM expression of fibrosis and contractility-related proteins, as well as survival of retinal ganglion cells (RGCs) assessed with immunohistochemistry. siRNA against RhoA or enhanced green fluorescent protein (EGFP) was also injected intracamerally into select animals. Successful RhoA knockdown was determined with quantitative reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry, and the effects of the knockdown on the parameters above analyzed. Results TGF-β caused increased TM contractile proteins and IOP spikes by day 7, sustained increases in IOP from day 15, and TM fibrosis at day 35. siRhoA abolished the transient 7 day IOP rise but not the later sustained IOP increase (due to fibrosis). At 35 days, TGF-β-related RGC loss was not prevented with siRhoA treatment. Conclusions We conclude that RhoA signaling mediates the early IOP rise induced by TM cellular changes associated with contractility but not the sustained IOP elevation caused by TM fibrosis. Thus, RhoA therapies offer a clinically relevant opportunity for IOP management, likely through the modulation of TM contractility, but appear to be ineffective in the amelioration of fibrosis.
Collapse
Affiliation(s)
- Lisa J Hill
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ben Mead
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Chloe N Thomas
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Simon Foale
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Elena Feinstein
- Research Division, Quark Pharmaceuticals, Ness Ziona, Israel
| | - Martin Berry
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Richard J Blanch
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Ann Logan
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
17
|
Zent J, Guo LW. Signaling Mechanisms of Myofibroblastic Activation: Outside-in and Inside-Out. Cell Physiol Biochem 2018; 49:848-868. [PMID: 30184544 DOI: 10.1159/000493217] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Myofibroblasts are central mediators of fibrosis. Typically derived from resident fibroblasts, myofibroblasts represent a heterogeneous population of cells that are principally defined by acquired contractile function and high synthetic ability to produce extracellular matrix (ECM). Current literature sheds new light on the critical role of ECM signaling coupled with mechanotransduction in driving myofibroblastic activation. In particular, transforming growth factor β1 (TGF-β1) and extra domain A containing fibronectin (EDA-FN) are thought to be the primary ECM signaling mediators that form and also induce positive feedback loops. The outside-in and inside-out signaling circuits are transmitted and integrated by TGF-β receptors and integrins at the cell membrane, ultimately perpetuating the abundance and activities of TGF-β1 and EDA-FN in the ECM. In this review, we highlight these conceptual advances in understanding myofibroblastic activation, in hope of revealing its therapeutic anti-fibrotic implications.
Collapse
Affiliation(s)
- Joshua Zent
- Medical Scientist Training Program, the Ohio State University, Columbus, Columbus, Ohio, USA
| | - Lian-Wang Guo
- Department of Surgery, Department of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, the Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
18
|
Morita T, Hayashi K. Tumor Progression Is Mediated by Thymosin-β4 through a TGFβ/MRTF Signaling Axis. Mol Cancer Res 2018; 16:880-893. [DOI: 10.1158/1541-7786.mcr-17-0715] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 11/16/2022]
|
19
|
Ptaschinski C, Lukacs NW. Acute and Chronic Inflammation Induces Disease Pathogenesis. MOLECULAR PATHOLOGY 2018:25-43. [DOI: 10.1016/b978-0-12-802761-5.00002-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Abstract
Transforming growth factor-β (TGF-β) may play a role in the pathogenesis of primary open-angle glaucoma (POAG). Elevated levels of TGF-β are found in the aqueous humor and in reactive optic nerve astrocytes in patients with glaucoma. In POAG, aqueous humor outflow resistance at the trabecular meshwork (TM) leads to increased intraocular pressure and retinal ganglion cell death. It is hypothesized that TGF-β increases outflow resistance by altering extracellular matrix homeostasis and cell contractility in the TM through interactions with other proteins and signaling molecules. TGF-β may also be involved in damage to the optic nerve head. Current available therapies for POAG focus exclusively on lowering intraocular pressure without addressing extracellular matrix homeostasis processes in the TM. The purpose of this review is to discuss possible therapeutic strategies targeting TGF-β in the treatment of POAG. Herein, we describe the current understanding of the role of TGF-β in POAG pathophysiology, and examine ways TGF-β may be targeted at the levels of production, activation, downstream signaling, and homeostatic regulation.
Collapse
|
21
|
Chen Y, Peng FF, Jin J, Chen HM, Yu H, Zhang BF. Src-mediated ligand release-independent EGFR transactivation involves TGF-β-induced Smad3 activation in mesangial cells. Biochem Biophys Res Commun 2017; 493:914-920. [PMID: 28943431 DOI: 10.1016/j.bbrc.2017.09.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 09/21/2017] [Indexed: 11/28/2022]
Abstract
A great deal of evidence highlighted the pathophysiologic importance of TGF-β1/Smad3 pathway in masangial extracellular matrix (ECM) accumulation, but some alternative signaling pathways are also involved. TGF-β was shown recently to induce rapid and transient epidermal-like growth factor receptor (EGFR) transactivation and subsequent fibronectin expression via heparin-binding epidermal-like growth factors (HB-EGF) release and binding in mesangial cells, which is independent of Smad2 activation. However, whether TGF-β could induce persistent EGFR transactivation remains to be identified. The present study demonstrates that in addition to transient EGFR transactivation, TGF-β1 can also induce continuous EGFR transactivation by a non-ligand-dependent pathway in rat mesangial cells. This sustained EGFR transactivation is mainly due to Src kinase-mediated persistent EGFR tyrosine phosphorylation at Y845 rather than Y1173. TGF-β1-induced early Smad3 phosphorylation is independent of transient EGFR transactivation and ERK1/2 activation initiated by HB-EGF release, whereas Src-mediated chronic EGFR transactivation and ERK1/2 activation participate in Smad3 activation in a relatively modest and delayed manner. Therefore, the present study further clarifies the mechanisms of EGFR transactivation in the TGF-β-initiated ECM upregulation and raises the possibility that targeting EGFR may provide a viable alternative strategy for inhibiting TGF-β in chronic kidney disease.
Collapse
Affiliation(s)
- Yan Chen
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Fang-Fang Peng
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Jing Jin
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Hong-Min Chen
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Hong Yu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China
| | - Bai-Fang Zhang
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, PR China.
| |
Collapse
|
22
|
Matoba K, Kawanami D, Nagai Y, Takeda Y, Akamine T, Ishizawa S, Kanazawa Y, Yokota T, Utsunomiya K. Rho-Kinase Blockade Attenuates Podocyte Apoptosis by Inhibiting the Notch Signaling Pathway in Diabetic Nephropathy. Int J Mol Sci 2017; 18:ijms18081795. [PMID: 28820432 PMCID: PMC5578183 DOI: 10.3390/ijms18081795] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
Podocyte apoptosis is a key process in the onset of diabetic nephropathy. A significant body of evidence shows that the Notch signaling pathway plays a central role in this process. We found that Rho-kinase mediates transforming growth factor β (TGF-β)-induced Notch ligand Jag1 expression. Importantly, TGF-β-mediated podocyte apoptosis was attenuated by Rho-kinase inhibition. Mechanistically, Rho-kinase regulated Jag1 induction via the extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK) but not Smad pathways. Consistently, the Rho-kinase inhibitor fasudil prevented albuminuria and the urinary excretion of nephrin in db/db mice and reduced the prevalence of podocyte apoptosis and Jag1 expression. Finally, the expression of Jag1 and apoptosis markers such as Bax and cyclin-dependent kinase inhibitor 1A (CDKN1A) was decreased in podocytes derived from db/db mice treated with fasudil. The present study provides evidence that Rho-kinase plays a key role in podocyte apoptosis. Rho-kinase is an attractive therapeutic target for diabetic nephropathy.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Daiji Kawanami
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Yosuke Nagai
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Yusuke Takeda
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Tomoyo Akamine
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Sho Ishizawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Kazunori Utsunomiya
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| |
Collapse
|
23
|
Liu D, Liu Y, Xia Z, Dong H, Yi Z. Reactive oxygen species modulator 1 regulates oxidative stress and induces renal and pulmonary fibrosis in a unilateral ureteral obstruction rat model and in HK‑2 cells. Mol Med Rep 2017; 16:4855-4862. [PMID: 28791399 PMCID: PMC5647032 DOI: 10.3892/mmr.2017.7161] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 05/19/2017] [Indexed: 02/06/2023] Open
Abstract
Renal interstitial fibrosis (RIF) is the main process that leads to renal failure. It is necessary to investigate the mechanism of RIF and identify appropriate methods of regulating it. Furthermore, unilateral ureteral obstruction is a frequently used model for the study of RIF. The morphological damage associated with kidney and lung dysfunction was detected using histopathological experiments. Subsequently, high expression of reactive oxygen species (ROS) modulator 1 (ROMO1) and ROS was measured in blood serum. In addition, epithelial‑mesenchymal transition marker, transforming growth factor β (TGF‑β) and mothers against decapentaplegic homolog 2/3 expression was evaluated using the reverse transcription‑quantitative polymerase chain reaction and western blotting. All serious symptoms were relieved to a certain extent following oxidation inhibitor intervention using three common antioxidants. HK‑2 cells were treated with H2O2 to cause oxidative stress, and ROMO1 and fibrosis marker expression increased; however, activation was suppressed byROMO1 knockout. The present study provides evidence that the expression of ROMO1 induces ROS production and activates the TGF‑β signaling pathway. It may be concluded that ROMO1 helps to provide a molecular basis for improved clinical intervention and prognosis of patients.
Collapse
Affiliation(s)
- Donghai Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ying Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Zhenkun Xia
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Haiyun Dong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Zhuwen Yi
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
24
|
Ojiaku CA, Yoo EJ, Panettieri RA. Transforming Growth Factor β1 Function in Airway Remodeling and Hyperresponsiveness. The Missing Link? Am J Respir Cell Mol Biol 2017; 56:432-442. [PMID: 27854509 DOI: 10.1165/rcmb.2016-0307tr] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of asthma includes a complex interplay among airway inflammation, hyperresponsiveness, and remodeling. Current evidence suggests that airway structural cells, including bronchial smooth muscle cells, myofibroblasts, fibroblasts, and epithelial cells, mediate all three aspects of asthma pathogenesis. Although studies show a connection between airway remodeling and changes in bronchomotor tone, the relationship between the two remains unclear. Transforming growth factor β1 (TGF-β1), a growth factor elevated in the airway of patients with asthma, plays a role in airway remodeling and in the shortening of various airway structural cells. However, the role of TGF-β1 in mediating airway hyperresponsiveness remains unclear. In this review, we summarize the literature addressing the role of TGF-β1 in airway remodeling and shortening. Through our review, we aim to further elucidate the role of TGF-β1 in asthma pathogenesis and the link between airway remodeling and airway hyperresponsiveness in asthma and to define TGF-β1 as a potential therapeutic target for reducing asthma morbidity and mortality.
Collapse
Affiliation(s)
- Christie A Ojiaku
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Edwin J Yoo
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Reynold A Panettieri
- 2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Diabetic nephropathy, a major microvascular complication of diabetes and the most common cause of end-stage renal disease, is characterized by prominent accumulation of extracellular matrix. The membrane microdomains caveolae, and their integral protein caveolin-1, play critical roles in the regulation of signal transduction. In this review we discuss current knowledge of the contribution of caveolin-1/caveolae to profibrotic signaling and the pathogenesis of diabetic kidney disease, and assess its potential as a therapeutic target. RECENT FINDINGS Caveolin (cav)-1 is key to facilitating profibrotic signal transduction induced by several stimuli known to be pathogenic in diabetic nephropathy, including the most prominent factors hyperglycemia and angiotensin II. Phosphorylation of cav-1 on Y14 is an important regulator of these responses. In vivo studies support a pathogenic role for caveolae in the progression of diabetic nephropathy. Targeting caveolin-1/caveolae would enable inhibition of multiple profibrotic pathways, representing a novel and potentially potent therapeutic option for diabetic nephropathy.
Collapse
Affiliation(s)
- Richard Van Krieken
- Department of Medicine, Division of Nephrology, St. Joseph's Hospital, McMaster University, 50 Charlton Ave E, T3311, Hamilton, ON, L8N 4A6, Canada
| | - Joan C Krepinsky
- Department of Medicine, Division of Nephrology, St. Joseph's Hospital, McMaster University, 50 Charlton Ave E, T3311, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
26
|
Roussa E, Speer JM, Chudotvorova I, Khakipoor S, Smirnov S, Rivera C, Krieglstein K. The membrane trafficking and functionality of the K+-Cl- co-transporter KCC2 is regulated by TGF-β2. J Cell Sci 2016; 129:3485-98. [PMID: 27505893 PMCID: PMC5047681 DOI: 10.1242/jcs.189860] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/02/2016] [Indexed: 02/02/2023] Open
Abstract
Functional activation of the neuronal K(+)-Cl(-) co-transporter KCC2 (also known as SLC12A5) is a prerequisite for shifting GABAA responses from depolarizing to hyperpolarizing during development. Here, we introduce transforming growth factor β2 (TGF-β2) as a new regulator of KCC2 membrane trafficking and functional activation. TGF-β2 controls membrane trafficking, surface expression and activity of KCC2 in developing and mature mouse primary hippocampal neurons, as determined by immunoblotting, immunofluorescence, biotinylation of surface proteins and KCC2-mediated Cl(-) extrusion. We also identify the signaling pathway from TGF-β2 to cAMP-response-element-binding protein (CREB) and Ras-associated binding protein 11b (Rab11b) as the underlying mechanism for TGF-β2-mediated KCC2 trafficking and functional activation. TGF-β2 increases colocalization and interaction of KCC2 with Rab11b, as determined by 3D stimulated emission depletion (STED) microscopy and co-immunoprecipitation, respectively, induces CREB phosphorylation, and enhances Rab11b gene expression. Loss of function of either CREB1 or Rab11b suppressed TGF-β2-dependent KCC2 trafficking, surface expression and functionality. Thus, TGF-β2 is a new regulatory factor for KCC2 functional activation and membrane trafficking, and a putative indispensable molecular determinant for the developmental shift of GABAergic transmission.
Collapse
Affiliation(s)
- Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Jan Manuel Speer
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Ilona Chudotvorova
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Shokoufeh Khakipoor
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Sergei Smirnov
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki FIN-00014, Finland
| | - Claudio Rivera
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki FIN-00014, Finland
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| |
Collapse
|
27
|
Choi HJ, Do KH, Park JH, Kim J, Yu M, Park SH, Moon Y. Early Epithelial Restitution by Nonsteroidal Anti-Inflammatory Drug–Activated Gene 1 Counteracts Intestinal Ulcerative Injuries. THE JOURNAL OF IMMUNOLOGY 2016; 197:1415-24. [DOI: 10.4049/jimmunol.1501784] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 06/03/2016] [Indexed: 01/07/2023]
|
28
|
Echarri A, Del Pozo MA. Caveolae - mechanosensitive membrane invaginations linked to actin filaments. J Cell Sci 2015; 128:2747-58. [PMID: 26159735 DOI: 10.1242/jcs.153940] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An essential property of the plasma membrane of mammalian cells is its plasticity, which is required for sensing and transmitting of signals, and for accommodating the tensional changes imposed by its environment or its own biomechanics. Caveolae are unique invaginated membrane nanodomains that play a major role in organizing signaling, lipid homeostasis and adaptation to membrane tension. Caveolae are frequently associated with stress fibers, a major regulator of membrane tension and cell shape. In this Commentary, we discuss recent studies that have provided new insights into the function of caveolae and have shown that trafficking and organization of caveolae are tightly regulated by stress-fiber regulators, providing a functional link between caveolae and stress fibers. Furthermore, the tension in the plasma membrane determines the curvature of caveolae because they flatten at high tension and invaginate at low tension, thus providing a tension-buffering system. Caveolae also regulate multiple cellular pathways, including RhoA-driven actomyosin contractility and other mechanosensitive pathways, suggesting that caveolae could couple mechanotransduction pathways to actin-controlled changes in tension through their association with stress fibers. Therefore, we argue here that the association of caveolae with stress fibers could provide an important strategy for cells to deal with mechanical stress.
Collapse
Affiliation(s)
- Asier Echarri
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Miguel A Del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
29
|
Blocking VEGF/Caveolin-1 signaling contributes to renal protection of fasudil in streptozotocin-induced diabetic rats. Acta Pharmacol Sin 2015; 36:831-40. [PMID: 25937636 DOI: 10.1038/aps.2015.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/10/2014] [Indexed: 01/02/2023]
Abstract
AIM RhoA/ROCK signaling plays an important role in diabetic nephropathy, and ROCK inhibitor fasudil exerts nephroprotection in experimental diabetic nephropathy. In this study we investigated the molecular mechanisms underlying the protective actions of fasudil in a rat model of diabetic nephropathy. METHODS Streptozotocin (STZ)-induced diabetic rats, to which fasudil or a positive control drug enalapril were orally administered for 8 months. Metabolic parameters and blood pressure were assessed during the treatments. After the rats were euthanized, kidney samples were collected for histological and molecular biological studies. VEGF, VEGFR1, VEGFR2 and fibronectin expression, and Src and caveolin-1 phosphorylation in the kidneys were assessed using RT-PCR, Western blot and immunohistochemistry assays. The association between VEGFR2 and caveolin-1 was analyzed with immunoprecipitation. RESULTS Chronic administration of fasudil (30 and 100 mg·kg(-1)·d(-1)) or enalapril (10 mg/kg, bid) significantly attenuated the glomerular sclerosis and albuminuria in the diabetic rats. Furthermore, fasudil treatment prevented the upregulation of VEGF, VEGFR1, VEGFR2 and fibronectin, and the increased association between VEGFR2 and caveolin-1 in the renal cortices, and partially blocked Src activation and caveolin-1 phosphorylation on tyrosine 14 in the kidneys, whereas enalapril treatment had no effects on the VEGFR2/Src/caveolin-1 signaling pathway. CONCLUSION Fasudil exerts protective actions in STZ-induced diabetic nephropathy by blocking the VEGFR2/Src/caveolin-1 signaling pathway and fibronectin upregulation. Thus, VEGFR2 may be a potential therapeutic target for the treatment of diabetic nephropathy.
Collapse
|
30
|
Hamzeh MT, Sridhara R, Alexander LD. Cyclic stretch-induced TGF-β1 and fibronectin expression is mediated by β1-integrin through c-Src- and STAT3-dependent pathways in renal epithelial cells. Am J Physiol Renal Physiol 2014; 308:F425-36. [PMID: 25477471 DOI: 10.1152/ajprenal.00589.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Extracellular matrix (ECM) proteins, including fibronectin, may contribute to the early development and progression of renal interstitial fibrosis associated with chronic renal disease. Recent studies showed that β1-integrin is associated with the development of renal fibrosis in a murine model of unilateral ureteral obstruction (UUO). However, the molecular events responsible for β1-integrin-mediated signaling, following UUO, have yet to be determined. In this study, we investigated the mechanism by which mechanical stretch, an in vitro model for chronic obstructive nephropathy, regulates fibronectin and transforming growth factor-β1 (TGF-β1) expression in cultured human proximal tubular epithelium (HK-2) cells. Mechanical stretch upregulated fibronectin and TGF-β1 expression and activated signal transducer and transcription factor 3 (STAT3) in a time-dependent manner. Stretch-induced fibronectin and TGF-β1 were suppressed by a STAT3 inhibitor, S3I-201, and by small interfering RNA (siRNA) targeting human STAT3 (STAT3 siRNA). Similarly, fibronectin and TGF-β1 expression and STAT3 activation induced by mechanical stretch were suppressed by the Src family kinase inhibitor PP2 and by transfection of HK-2 cells with a dominant-negative mutant of c-Src (DN-Src), whereas PP3, an inactive analog of PP2, had no significant effect. Furthermore, mechanical stretch resulted in increased β1-integrin mRNA and protein levels in HK-2 cells. Furthermore, neutralizing antibody against β1-integrin and silencing of β1-integrin expression with siRNAs resulted in decreased c-Src and STAT3 activation and TGF-β1 and fibronectin expression evoked by mechanical stretch. This work demonstrates, for the first time, a role for β1-integrin in stretch-induced renal fibrosis through the activation of c-Src and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Mona T Hamzeh
- Department of Biology, Division of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan
| | - Rashmi Sridhara
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| | - Larry D Alexander
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| |
Collapse
|
31
|
Liu Y, Lu S, Zhang Y, Wang X, Kong F, Liu Y, Peng L, Fu Y. Role of caveolae in high glucose and TGF-β₁ induced fibronectin production in rat mesangial cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8381-8390. [PMID: 25674202 PMCID: PMC4314031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/26/2014] [Indexed: 06/04/2023]
Abstract
Accumulation of extracellular matrix (ECM) in glomerular mesangium correlates with loss of renal function in diabetic nephropathy. However, the mechanisms underlying are still incompletely known. In the present study, we explored the role of caveolae in ECM production in rat mesangial cells (MCs) stimulated by high glucose or transforming growth factor-β1 (TGF-β1), and investigated the possible mechanisms. High glucose (HG) or TGF-β1 significantly increased collagen-1 and fibronectin expression at both mRNA and protein levels in time- course dependent manners, and simultaneously induced caveolin-1 tyrosine phosphorylation. Disruption of caveolae with Methyl-β-cyclodextrin (β-MCD) prevented HG and TGF-β1 induced caveolin-1 tyrosine phosphorylation, and attenuated fibronectin but not collagen-1 production. This effect of β-MCD on fibronectin production could be abolished by cholesterol, which restored HG and TGF-β1 induced caveolin-1 tyrosine phosphorylation. In addition, HG and TGF-β1 induced fibronectin production was attenuated by a caveolin-1 scaffold domain peptide. These findings indicate that mesangial cell caveolae regulate fibronectin production at least partly through caveolin-1 phosphorylation.
Collapse
Affiliation(s)
- Yuantao Liu
- Department of Endocrinology, The Second Hospital of Shandong UniversityJinan, Shandong, China
| | - Shengxia Lu
- Department of Nephrology, The Second Hospital of Shandong UniversityJinan, Shandong, China
- Department of Cardiology, Shandong Electric Power Central HospitalJinan, China
| | - Yuchao Zhang
- Institute of Cell Biology, Shandong UniversityJinan, China
| | - Xiangdong Wang
- Institute of Cell Biology, Shandong UniversityJinan, China
| | - Feng Kong
- Central Laboratory, The Second Hospital of Shandong UniversityJinan, Shandong, China
| | - Ye Liu
- Department of Nephrology, The Second Hospital of Shandong UniversityJinan, Shandong, China
| | - Li Peng
- Department of Endocrinology, The Second Hospital of Shandong UniversityJinan, Shandong, China
| | - Yuqin Fu
- Department of Nephrology, The Second Hospital of Shandong UniversityJinan, Shandong, China
| |
Collapse
|
32
|
Choi HJ, Kim HG, Kim J, Park SH, Park J, Oh CG, Do KH, Lee SJ, Park YC, Ahn SC, Kim YS, Moon Y. Pro-apoptotic action of macrophage inhibitory cytokine 1 and counteraction of activating transcription factor 3 in carrageenan-exposed enterocytes. Toxicol Lett 2014; 231:1-8. [PMID: 25180886 DOI: 10.1016/j.toxlet.2014.08.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 08/28/2014] [Accepted: 08/29/2014] [Indexed: 12/15/2022]
Abstract
Carrageenan (CGN), a widely used food additive, has been shown to injure the epithelial barrier in animal models. This type of damage is a clinical feature of inflammatory bowel disease (IBD) in humans. In the present study, the effects of CGN on pro-apoptotic responses associated with macrophage inhibitory cytokine 1 (MIC-1) regulation in human enterocytes were evaluated. CGN up-regulated the expression of MIC-1 that promoted epithelial cell apoptosis. Although MIC-1 induction was dependent on pro-apoptotic p53 protein, the pro-survival protein activating transcription factor 3 (ATF3) was negatively regulated by p53 expression. However, MIC-1 enhanced the expression of the pro-survival protein ATF3 in enterocytes exposed to CGN. Functionally, MIC-1-mediated epithelial cell apoptosis was counteracted by the pro-survival action of ATF3 in response to CGN exposure. These findings demonstrated that the counterbalance between MIC-1 and ATF3 is critical for deciding the fate of enterocytes under the food chemical stress.
Collapse
Affiliation(s)
- Hye Jin Choi
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hwi-Gon Kim
- Department of Obstetrics and Gynecology, Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jiyeon Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Chang Gyu Oh
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Kee Hun Do
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Young Chul Park
- Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Soon Cheol Ahn
- Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Yong Sik Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea; Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea; Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Busan, South Korea.
| |
Collapse
|
33
|
Manickam N, Patel M, Griendling KK, Gorin Y, Barnes JL. RhoA/Rho kinase mediates TGF-β1-induced kidney myofibroblast activation through Poldip2/Nox4-derived reactive oxygen species. Am J Physiol Renal Physiol 2014; 307:F159-71. [PMID: 24872317 PMCID: PMC4101629 DOI: 10.1152/ajprenal.00546.2013] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 05/18/2014] [Indexed: 02/07/2023] Open
Abstract
The small G proteins Rac1 and RhoA regulate actin cytoskeleton, cell shape, adhesion, migration, and proliferation. Recent studies in our laboratory have shown that NADPH oxidase Nox4-derived ROS are involved in transforming growth factor (TGF)-β1-induced rat kidney myofibroblast differentiation assessed by the acquisition of an α-smooth muscle actin (α-SMA) phenotype and expression of an alternatively spliced fibronectin variant (Fn-EIIIA). Rac1 and RhoA are essential in signaling by some Nox homologs, but their role as effectors of Nox4 in kidney myofibroblast differentiation is not known. In the present study, we explored a link among Rac1 and RhoA and Nox4-dependent ROS generation in TGF-β1-induced kidney myofibroblast activation. TGF-β1 stimulated an increase in Nox4 protein expression, NADPH oxidase activity, and abundant α-SMA and Fn-EIIIA expression. RhoA but not Rac1 was involved in TGF-β1 induction of Nox4 signaling of kidney myofibroblast activation. TGF-β1 stimulated active RhoA-GTP and increased Rho kinase (ROCK). Inhibition of RhoA with small interfering RNA and ROCK using Y-27632 significantly reduced TGF-β1-induced stimulation of Nox4 protein, NADPH oxidase activity, and α-SMA and Fn-EIIIA expression. Treatment with diphenyleneiodonium, an inhibitor of NADPH oxidase, did not decrease RhoA activation but inhibited TGF-β1-induced α-SMA and Fn-EIIIA expression, indicating that RhoA is upstream of ROS generation. RhoA/ROCK also regulated polymerase (DNA-directed) δ-interacting protein 2 (Poldip2), a newly discovered Nox4 enhancer protein. Collectively, these data indicate that RhoA/ROCK is upstream of Poldip2-dependent Nox4 regulation and ROS production and induces redox signaling of kidney myofibroblast activation and may broader implications in the pathophysiology of renal fibrosis.
Collapse
Affiliation(s)
- Nagaraj Manickam
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Mandakini Patel
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Kathy K Griendling
- The Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Yves Gorin
- The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| | - Jeffrey L Barnes
- The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Department of Medicine, Division of Nephrology, The University of Texas Health Science Center, San Antonio, Texas; and
| |
Collapse
|
34
|
Wu SZ, Peng FF, Li JL, Ye F, Lei SQ, Zhang BF. Akt and RhoA activation in response to high glucose require caveolin-1 phosphorylation in mesangial cells. Am J Physiol Renal Physiol 2014; 306:F1308-17. [PMID: 24694591 DOI: 10.1152/ajprenal.00447.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glomerular matrix accumulation is a hallmark of diabetic renal disease. Serine/threonine kinase PKC-β1 mediates glucose-induced Akt S473 phosphorylation, RhoA activation, and transforming growth factor (TGF)-β1 upregulation and finally leads to matrix upregulation in mesangial cells (MCs). It has been reported that glucose-induced PKC-β1 activation is dependent on caveolin-1 and the presence of intact caveolae in MCs; however, whether activated PKC-β1 regulates caveolin-1 expression and phosphorylation are unknown. Here, we showed that, although the caveolin-1 protein level had no significant change, the PKC-β-specific inhibitor LY-333531 blocked caveolin-1 Y14 phosphorylation in high glucose (HG)-treated MCs and in the renal cortex of diabetic rats. The Src-specific inhibitor SU-6656 prevented the HG-induced association between PKC-β1 and caveolin-1 and PKC-β1 membrane translocation, whereas PKC-β1 small interfering RNA failed to block Src activation, indicating that Src kinase is upstream of PKC-β1 activation. Although LY-333531 blocked PKC-β1 membrane translocation, it had no effect on the PKC-β1/caveolin-1 association, suggesting that PKC-β1 activation requires the interaction of caveolin-1 and PKC-β1. PKC-β1-mediated Akt S473 phosphorylation, RhoA activation, and fibronectin upregulation in response to HG were prevented by SU-6656 and nonphosphorylatable mutant caveolin-1 Y14A. In conclusion, Src activation by HG mediates the PKC-β1/caveolin-1 association and PKC-β1 activation, which assists in caveolin-1 Y14 phosphorylation by Src kinase. The downstream effects, including Akt S473 phosphorylation, RhoA activation, and fibronectin upregulation, require caveolin-1 Y14 phosphorylation. Caveolin-1 is thus an important mediator of the profibrogenic process in diabetic renal disease.
Collapse
Affiliation(s)
- Su-Zhen Wu
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, People's Republic of China; and
| | - Fang-Fang Peng
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, People's Republic of China; and
| | - Jia-Lin Li
- Gannan Medical University, Ganzhou, People's Republic of China; and
| | - Feng Ye
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, People's Republic of China; and
| | - Shao-Qing Lei
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, People's Republic of China; and
| | - Bai-Fang Zhang
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, People's Republic of China; and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, People's Republic of China
| |
Collapse
|
35
|
Lipid rafts are required for signal transduction by angiotensin II receptor type 1 in neonatal glomerular mesangial cells. Exp Cell Res 2014; 324:92-104. [PMID: 24662198 DOI: 10.1016/j.yexcr.2014.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 11/24/2022]
Abstract
Angiotensin II (ANG-II) receptors (AGTRs) contribute to renal physiology and pathophysiology, but the underlying mechanisms that regulate AGTR function in glomerular mesangium are poorly understood. Here, we show that AGTR1 is the functional AGTR subtype expressed in neonatal pig glomerular mesangial cells (GMCs). Cyclodextrin (CDX)-mediated cholesterol depletion attenuated cell surface AGTR1 protein expression and ANG-II-induced intracellular Ca(2+) ([Ca(2+)]i) elevation in the cells. The COOH-terminus of porcine AGTR1 contains a caveolin (CAV)-binding motif. However, neonatal GMCs express CAV-1, but not CAV-2 and CAV-3. Colocalization and in situ proximity ligation assay detected an association between endogenous AGTR1 and CAV-1 in the cells. A synthetic peptide corresponding to the CAV-1 scaffolding domain (CSD) sequence also reduced ANG-II-induced [Ca(2+)]i elevation in the cells. Real-time imaging of cell growth revealed that ANG-II stimulates neonatal GMC proliferation. ANG-II-induced GMC growth was attenuated by EMD 66684, an AGTR1 antagonist; BAPTA, a [Ca(2+)]i chelator; KN-93, a Ca(2+)/calmodulin-dependent protein kinase II inhibitor; CDX; and a CSD peptide, but not PD 123319, a selective AGTR2 antagonist. Collectively, our data demonstrate [Ca(2+)]i-dependent proliferative effect of ANG-II and highlight a critical role for lipid raft microdomains in AGTR1-mediated signal transduction in neonatal GMCs.
Collapse
|
36
|
Gobe GC, Bennett NC, West M, Colditz P, Brown L, Vesey DA, Johnson DW. Increased progression to kidney fibrosis after erythropoietin is used as a treatment for acute kidney injury. Am J Physiol Renal Physiol 2014; 306:F681-92. [PMID: 24402097 DOI: 10.1152/ajprenal.00241.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Treatment of renal ischemia-reperfusion (IR) injury with recombinant human erythropoietin (rhEPO) reduces acute kidney injury and improves function. We aimed to investigate whether progression to chronic kidney disease associated with acute injury was also reduced by rhEPO treatment, using in vivo and in vitro models. Rats were subjected to bilateral 40-min renal ischemia, and kidneys were studied at 4, 7, and 28 days postreperfusion for renal function, tubular injury and repair, inflammation, and fibrosis. Acute injury was modulated using rhEPO (1,000 or 5,000 IU/kg, intraperitoneally) at the time of reperfusion. Renal tubular epithelial cells or fibroblasts in culture were subjected to hypoxia or oxidative stress, with or without rhEPO (200 IU/ml), and fibrogenesis was studied. The results of the in vivo model confirmed functional and structural improvement with rhEPO at 4 days post-IR (P < 0.05). At 7 days post-IR, fibrosis and myofibroblast stimulation were increased with IR with and without rhEPO (P < 0.01). However, at 28 days post-IR, renal fibrosis and myofibroblast numbers were significantly greater with IR plus rhEPO (P < 0.01) compared with IR only. Mechanistically, rhEPO stimulated profibrotic transforming growth factor-β, oxidative stress (marker 8-hydroxy-deoxyguanosine), and phosphorylation of the signal transduction protein extracellular signal-regulated kinase. In vitro, rhEPO protected tubular epithelium from apoptosis but stimulated epithelial-to-mesenchymal transition and also protected and activated fibroblasts, particularly with oxidative stress. In summary, although rhEPO was protective of renal function and structure in acute kidney injury, the supraphysiological dose needed for renoprotection contributed to fibrogenesis and stimulated chronic kidney disease in the long term.
Collapse
Affiliation(s)
- Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Univ. of Queensland, Translational Research Institute, Kent St., Woolloongabba, Brisbane, Australia 4102..
| | | | | | | | | | | | | |
Collapse
|
37
|
Kirihara T, Shimazaki A, Nakamura M, Miyawaki N. Ocular hypotensive efficacy of Src-family tyrosine kinase inhibitors via different cellular actions from Rock inhibitors. Exp Eye Res 2013; 119:97-105. [PMID: 24321889 DOI: 10.1016/j.exer.2013.11.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 11/24/2013] [Accepted: 11/27/2013] [Indexed: 11/16/2022]
Abstract
We investigated the effects of Src-family tyrosine kinase (SFK) inhibitors on intraocular pressure (IOP) and trabecular meshwork (TM) cells. The SFK inhibitors, PP2, PP1, and damnacanthal, significantly lowered IOP from baseline following intracameral injection in ocular normotensive rabbits, and PP2 decreased trans-epithelial electrical resistance (TEER) of TM cell layers in a dose-dependent manner ranging from 0.1 μM to 100 μM. The maximal efficacy of PP2 on TEER was a reduction to 71.7% relative to the vehicle-treated group at 100 μM. PP2 decreased the adhesion of TM cells to culture surfaces either uncoated with specific ECM proteins dose-dependently or coated with extracellular matrix proteins such as laminin I, fibronectin, collagen type I and basement membrane extraction. Tyrosine phosphorylation of focal adhesion kinase and p130(cas) was decreased by PP2. On the other hand, major changes in actin staining of TM cells were not able to be detected after PP2 treatment, although quantitative analysis showed that PP2 induced some morphological changes which were in the different direction to those caused by Y-27632, a Rock inhibitor. Y-27632 at 10 μM increased the permeability of TM cell layers, but did not induce changes in the adhesion of TM cells. These results suggest that SFK inhibitors lower IOP, at least partly, by acting on TM cells in a manner that is distinct from Rock inhibitors.
Collapse
Affiliation(s)
- Tomoko Kirihara
- Ophthalmic Research and Development Center, Santen Pharmaceutical Co., Ltd., 8916-16 Takayama-cho, Ikoma-shi, Nara 630-0101, Japan; Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma-shi, Nara 630-0192, Japan.
| | - Atsushi Shimazaki
- Ophthalmic Research and Development Center, Santen Pharmaceutical Co., Ltd., 8916-16 Takayama-cho, Ikoma-shi, Nara 630-0101, Japan
| | - Masatsugu Nakamura
- Ophthalmic Research and Development Center, Santen Pharmaceutical Co., Ltd., 8916-16 Takayama-cho, Ikoma-shi, Nara 630-0101, Japan
| | - Nobuaki Miyawaki
- Ophthalmic Research and Development Center, Santen Pharmaceutical Co., Ltd., 8916-16 Takayama-cho, Ikoma-shi, Nara 630-0101, Japan
| |
Collapse
|
38
|
Guan T, Gao B, Chen G, Chen X, Janssen M, Uttarwar L, Ingram AJ, Krepinsky JC. Colchicine attenuates renal injury in a model of hypertensive chronic kidney disease. Am J Physiol Renal Physiol 2013; 305:F1466-76. [DOI: 10.1152/ajprenal.00057.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hypertension is a risk factor for chronic kidney disease, particularly when associated with impaired renal autoregulation and thereby increased intraglomerular pressure (Pgc). Elevated Pgc can be modeled in vitro by exposing glomerular mesangial cells to mechanical strain. We previously showed that RhoA mediates strain-induced matrix production. Here, we show that RhoA activation is dependent on an intact microtubule network. Upregulation of the profibrotic cytokine connective tissue growth factor (CTGF) by mechanical strain is dependent on RhoA activation and inhibited by microtubule disruption. We tested the effects of the microtubule depolymerizing agent colchicine in 5/6 nephrectomized rats, a model of chronic kidney disease driven by elevated Pgc. Colchicine inhibited glomerular RhoA activation and attenuated both glomerular sclerosis and interstitial fibrosis without affecting systemic blood pressure. Upregulation of the matrix proteins collagen I and fibronectin, as well as CTGF, was attenuated by colchicine. Activity of the profibrotic cytokine TGF-β, as assessed by Smad3 phosphorylation, was also inhibited by colchicine. Microtubule disruption significantly decreased renal infiltration of lymphocytes and macrophages. Our studies thus indicate that colchicine modifies hypertensive renal fibrosis. Its protective effects are likely mediated by inhibition of RhoA signaling and renal infiltration of inflammatory cells. Already well-established in clinical practice for other indications, prevention of hypertension-associated renal fibrosis may represent a new potential use for colchicine.
Collapse
Affiliation(s)
- Tianxiu Guan
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Bo Gao
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Guang Chen
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Xing Chen
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Melissa Janssen
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Lalita Uttarwar
- Division of Nephrology, McMaster University, Hamilton, Canada
| | | | | |
Collapse
|
39
|
Low-dose endothelial monocyte-activating polypeptide-ii increases permeability of blood-tumor barrier by caveolae-mediated transcellular pathway. J Mol Neurosci 2013; 52:313-22. [PMID: 24526454 DOI: 10.1007/s12031-013-0148-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 10/10/2013] [Indexed: 10/26/2022]
Abstract
Low-dose endothelial monocyte-activating polypeptide-II (EMAP-II) can selectively increase blood-tumor barrier (BTB) permeability via the paracellular pathway. The role of the transcellular pathway in this process is unclear. This study was conducted to evaluate the potential involvement of the transcellular pathway in EMAP-II-induced opening of the BTB and to identify the associated mechanisms. Evans blue extravasation test was used to measure changes in BTB permeability after EMAP-II (80 ng/kg) administration in a rat model of C6 glioma. Changes in the quantity of pinocytotic vesicles in rat brain microvascular endothelial cells (BMECs) were observed using transmission electron microscopy. Reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemistry assays were performed to detect the expression of the caveolar structural proteins, caveolin-1 and caveolin-2, in BMECs. Alterations in the expression of phospho (p)-Src, p-caveolin-1, and p-caveolin-2 and the activity of RhoA also were measured. Effects of tyrosine kinase inhibition on EMAP-II-induced RhoA/Rho kinase activations and tyrosine kinase, RhoA, or Rho kinase inhibition on EMAP-II-induced caveolin-1 and caveolin-2 phosphorylation were determined by inhibition studies. One hour after EMAP-II administration, the quantity of pinocytotic vesicles in BMECs increased markedly, consistent with changes in BTB permeability. The expression levels of caveolin-1, caveolin-2, p-caveolin-1, and p-caveolin-2 in BMECs also were significantly increased at 1 h. The peak expression level of p-Src and the peak activity of RhoA occurred at 0.25 and 0.5 h, respectively. Inhibition of tyrosine kinase significantly diminished the activities of RhoA and Rho kinase induced by EMAP-II. In addition, EMAP-II-induced phosphorylation of caveolin-1 and caveolin-2 was completely blocked by inhibition of tyrosine kinase, RhoA, or Rho kinase. We suggest that low-dose EMAP-II can induce BTB hyperpermeability via the transcellular pathway, which is associated with phosphorylation and upregulation of caveolin-1 and caveolin-2 and involves the tyrosine kinase/RhoA/Rho kinase signaling pathway.
Collapse
|
40
|
Guan TH, Chen G, Gao B, Janssen MR, Uttarwar L, Ingram AJ, Krepinsky JC. Caveolin-1 deficiency protects against mesangial matrix expansion in a mouse model of type 1 diabetic nephropathy. Diabetologia 2013; 56:2068-77. [PMID: 23793581 DOI: 10.1007/s00125-013-2968-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 05/24/2013] [Indexed: 01/10/2023]
Abstract
AIMS/HYPOTHESIS Glomerular matrix protein accumulation, mediated largely by resident mesangial cells (MCs), is central to the pathogenesis of diabetic nephropathy. We previously showed that caveolin (CAV)-1/caveolae mediate matrix upregulation by MCs in response to high glucose and TGFβ, two important pathogenic mediators of diabetic glomerular sclerosis. Here, we evaluated the in vivo role of CAV-1/caveolae in the development of diabetic nephropathy. METHODS Diabetes was induced in Cav1-knockout (KO) mice and their wild-type (WT) counterparts by streptozotocin injection. After 10 months, kidneys were evaluated for the development of nephropathy, including glomerular sclerosis and upregulation of matrix proteins. Parallel experiments assessing glucose-induced matrix upregulation were carried out in MCs isolated from KO mice. RESULTS KO diabetic mice developed hyperglycaemia and renal hypertrophy, but were protected from developing albuminuria and glomerular sclerosis compared with WT mice. KO mice were significantly protected from the upregulation of glomerular collagen I, fibronectin, connective tissue growth factor (CTGF) and TGFβ. In vitro, glucose induced collagen I A1 promoter activation and collagen I, fibronectin and CTGF protein upregulation in WT but not KO MCs. Re-expression of Cav1 in KO cells restored this response. CONCLUSIONS/INTERPRETATION Cav1 deletion rendered significant protection from glomerular matrix accumulation and albuminuria in a mouse model of type 1 diabetes. These studies provide a foundation for the development of renal-targeted interference with CAV-1/caveolae as a novel approach to the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- T H Guan
- Division of Nephrology, McMaster University, St Joseph's Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON, Canada L8N 4A6
| | | | | | | | | | | | | |
Collapse
|
41
|
Ni J, Dong Z, Han W, Kondrikov D, Su Y. The role of RhoA and cytoskeleton in myofibroblast transformation in hyperoxic lung fibrosis. Free Radic Biol Med 2013; 61:26-39. [PMID: 23517783 PMCID: PMC3849210 DOI: 10.1016/j.freeradbiomed.2013.03.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 02/05/2013] [Accepted: 03/12/2013] [Indexed: 01/08/2023]
Abstract
Myofibroblast transformation is a key process in the pathogenesis of lung fibrosis. We have previously reported that hyperoxia induces RhoA activation in HFL-1 lung fibroblasts and RhoA mediates collagen synthesis in hyperoxic lung fibrosis. In this study, we investigated the role of RhoA and actin cytoskeleton in hyperoxia-induced myofibroblast transformation. Exposure of HFL-1 lung fibroblasts to hyperoxia stimulated actin filament formation, shift of G-actin to F-actin, nuclear colocalization of myocardin-related transcription factor-A (MRTF-A), recruitment of MRTF-A to the α-smooth muscle actin (α-SMA) gene promoter, myofibroblast transformation, and collagen-I synthesis. Inhibition of RhoA by C3 transferase CT-04 or dominant-negative RhoA mutant T19N, and inhibition of ROCK by Y27632, prevented myofibroblast transformation and collagen-I synthesis. Moreover, inhibition of RhoA by CT-04 prevented hyperoxia-induced actin filament formation, shift of G-actin to F-actin, and nuclear colocalization of MRTF-A. In addition, disrupting actin filaments with cytochalasin D or scavenging reactive oxygen species (ROS) with tiron attenuated actin filament formation, nuclear colocalization of MRTF-A, myofibroblast transformation, and collagen-I synthesis. Furthermore, overexpression of constitutively active RhoA mutant Q63L or stabilization of actin filaments recapitulated the effects of hyperoxia on the actin cytoskeleton and nuclear colocalization of MRTF-A, myofibroblast transformation, and collagen-I synthesis. Interestingly, knocking down MRTF-A prevented hyperoxia-induced increase in the recruitment of MRTF-A to the serum response factor transcriptional complex on the α-SMA gene promoter, myofibroblast transformation, and collagen-I synthesis. Finally, Y27632 and tiron attenuated hyperoxia-induced increases in α-SMA and collagen-I in mouse lungs. Together, these results indicate that the actin cytoskeletal reorganization due to the ROS/RhoA-ROCK pathway mediates myofibroblast transformation and collagen synthesis in lung fibrosis of oxygen toxicity. MRTF-A contributes to the regulatory effect of the actin cytoskeleton on myofibroblast transformation during hyperoxia.
Collapse
Affiliation(s)
- Jixiang Ni
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Department of Respiratory Medicine, The First People's Hospital of Yichang, Yichang, China; The People's Hospital, China Three Gorges University, Yichang, Hubei Province, China
| | - Zheng Dong
- Department of Cell Biology & Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Weihong Han
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Dmitry Kondrikov
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA; Center for Biotechnology & Genomic Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
42
|
The less-often-traveled surface of stem cells: caveolin-1 and caveolae in stem cells, tissue repair and regeneration. Stem Cell Res Ther 2013; 4:90. [PMID: 23899671 PMCID: PMC3854699 DOI: 10.1186/scrt276] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells are an important resource for tissue repair and regeneration. While a great deal of attention has focused on derivation and molecular regulation of stem cells, relatively little research has focused on how the subcellular structure and composition of the cell membrane influences stem cell activities such as proliferation, differentiation and homing. Caveolae are specialized membrane lipid rafts coated with caveolin scaffolding proteins, which can regulate cholesterol transport and the activity of cell signaling receptors and their downstream effectors. Caveolin-1 is involved in the regulation of many cellular processes, including growth, control of mitochondrial antioxidant levels, migration and senescence. These activities are of relevance to stem cell biology, and in this review evidence for caveolin-1 involvement in stem cell biology is summarized. Altered stem and progenitor cell populations in caveolin-1 null mice suggest that caveolin-1 can regulate stem cell proliferation, and in vitro studies with isolated stem cells suggest that caveolin-1 regulates stem cell differentiation. The available evidence leads us to hypothesize that caveolin-1 expression may stabilize the differentiated and undifferentiated stem cell phenotype, and transient downregulation of caveolin-1 expression may be required for transition between the two. Such regulation would probably be critical in regenerative applications of adult stem cells and during tissue regeneration. We also review here the temporal changes in caveolin-1 expression reported during tissue repair. Delayed muscle regeneration in transgenic mice overexpressing caveolin-1 as well as compromised cardiac, brain and liver tissue repair and delayed wound healing in caveolin-1 null mice suggest that caveolin-1 plays an important role in tissue repair, but that this role may be negative or positive depending on the tissue type and the nature of the repair process. Finally, we also discuss how caveolin-1 quiescence-inducing activities and effects on mitochondrial antioxidant levels may influence stem cell aging.
Collapse
|
43
|
Chen G, Chen X, Sukumar A, Gao B, Curley J, Schnaper HW, Ingram AJ, Krepinsky JC. TGFβ receptor I transactivation mediates stretch-induced Pak1 activation and CTGF upregulation in mesangial cells. J Cell Sci 2013; 126:3697-712. [PMID: 23781022 DOI: 10.1242/jcs.126714] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Increased intraglomerular pressure is an important pathogenic determinant of kidney fibrosis in the progression of chronic kidney disease, and can be modeled by exposing glomerular mesangial cells (MC) to mechanical stretch. MC produce extracellular matrix and profibrotic cytokines, including connective tissue growth factor (CTGF) when stretched. We show that p21-activated kinase 1 (Pak1) is activated by stretch in MC in culture and in vivo in a process marked by elevated intraglomerular pressures. Its activation is essential for CTGF upregulation. Rac1 is an upstream regulator of Pak1 activation. Stretch induces transactivation of the type I transforming growth factor β1 receptor (TβRI) independently of ligand binding. TβRI transactivation is required not only for Rac1/Pak1 activation, but also for activation of the canonical TGFβ signaling intermediate Smad3. We show that Smad3 activation is an essential requirement for CTGF upregulation in MC under mechanical stress. Pak1 regulates Smad3 C-terminal phosphorylation and transcriptional activation. However, a second signaling pathway, that of RhoA/Rho-kinase and downstream Erk activation, is also required for stretch-induced CTGF upregulation in MC. Importantly, this is also regulated by Pak1. Thus, Pak1 serves as a novel central mediator in the stretch-induced upregulation of CTGF in MC.
Collapse
Affiliation(s)
- Guang Chen
- Division of Nephrology, St. Joseph's Hospital, McMaster University, Hamilton, ON L8N 4A6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Schinner E, Schramm A, Kees F, Hofmann F, Schlossmann J. The cyclic GMP-dependent protein kinase Iα suppresses kidney fibrosis. Kidney Int 2013; 84:1198-206. [PMID: 23760283 DOI: 10.1038/ki.2013.219] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 03/20/2013] [Accepted: 04/18/2013] [Indexed: 11/09/2022]
Abstract
Cyclic guanosine monophosphate (cGMP) is synthesized by nitric oxide or natriuretic peptide-stimulated guanylyl cyclases and exhibits pleiotropic regulatory functions in the kidney. Hence, integration of cGMP signaling by cGMP-dependent protein kinases (cGKs) might play a critical role in renal physiology; however, detailed renal localization of cGKs is still lacking. Here, we performed an immunohistochemical analysis of cGKIα and cGKIβ isozymes in the mouse kidney and found both in arterioles, the mesangium, and within the cortical interstitium. In contrast to cGKIα, the β-isoform was not detected in the juxtaglomerular apparatus or medullary fibroblasts. Since interstitial fibroblasts play a prominent role in interstitial fibrosis, we focused our study on cGKI function in the interstitium, emphasizing a functional differentiation of both isoforms, and determined whether cGKIs influence renal fibrosis induced by unilateral ureter obstruction. Treatment with the guanylyl cyclase activators YC1 or isosorbide dinitrate showed stronger antifibrotic effects in wild-type than in cGKI-knockout or in smooth muscle-cGKIα-rescue mice, which are cGKI deficient in the kidney except in the renal vasculature. Moreover, fibrosis influenced the mRNA and protein expression levels of cGKIα more strongly than cGKIβ. Thus, our results indicate that cGMP, acting primarily through cGKIα, is an important suppressor of kidney fibrosis.
Collapse
Affiliation(s)
- Elisabeth Schinner
- Lehrstuhl für Pharmakologie und Toxikologie, Institut für Pharmazie, Universität Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
45
|
Yang H, Li G, Wu JJ, Wang L, Uhler M, Simeone DM. Protein kinase A modulates transforming growth factor-β signaling through a direct interaction with Smad4 protein. J Biol Chem 2013; 288:8737-8749. [PMID: 23362281 PMCID: PMC3605691 DOI: 10.1074/jbc.m113.455675] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor β (TGFβ) signaling normally functions to regulate embryonic development and cellular homeostasis. It is increasingly recognized that TGFβ signaling is regulated by cross-talk with other signaling pathways. We previously reported that TGFβ activates protein kinase A (PKA) independent of cAMP through an interaction of an activated Smad3-Smad4 complex and the regulatory subunit of the PKA holoenzyme (PKA-R). Here we define the interaction domains of Smad4 and PKA-R and the functional consequences of this interaction. Using a series of Smad4 and PKA-R truncation mutants, we identified amino acids 290-300 of the Smad4 linker region as critical for the specific interaction of Smad4 and PKA-R. Co-immunoprecipitation assays showed that the B cAMP binding domain of PKA-R was sufficient for interaction with Smad4. Targeting of B domain regions conserved among all PKA-R isoforms and exposed on the molecular surface demonstrated that amino acids 281-285 and 320-329 were required for complex formation with Smad4. Interactions of these specific regions of Smad4 and PKA-R were necessary for TGFβ-mediated increases in PKA activity, CREB (cAMP-response element-binding protein) phosphorylation, induction of p21, and growth inhibition. Moreover, this Smad4-PKA interaction was required for TGFβ-induced epithelial mesenchymal transition, invasion of pancreatic tumor cells, and regulation of tumor growth in vivo.
Collapse
Affiliation(s)
- Huibin Yang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Gangyong Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Jing-Jiang Wu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Lidong Wang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Michael Uhler
- Department of Biochemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Diane M Simeone
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109; Translational Oncology Program, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
46
|
Wu T, Zhang B, Ye F, Xiao Z. A potential role for caveolin-1 in VEGF-induced fibronectin upregulation in mesangial cells: involvement of VEGFR2 and Src. Am J Physiol Renal Physiol 2013; 304:F820-30. [DOI: 10.1152/ajprenal.00294.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
VEGF is known to be an endothelial cell mitogen that stimulates angiogenesis by promoting endothelial cell survival, proliferation, migration, and differentiation. Recent studies have suggested that VEGF may play a pivotal role in glomerular sclerosis through extracellular matrix protein (ECM) accumulation, although the signaling mechanism is still unclear. The GTPase RhoA has been implicated in VEGF-induced type IV collagen accumulation in some settings. Here we study the role of different VEGF receptors and membrane microdomain caveolae in VEGF-induced RhoA activation and fibronectin upregulation in mesangial cells (MCs). In primary rat MC, VEGF time and dose dependently increased fibronectin production. Rho pathway inhibition blocked VEGF-induced fibronectin upregulation. VEGF-induced RhoA activation was prevented by disrupting caveolae with cholesterol depletion and rescued by cholesterol repletion. VEGF stimulation led to a markedly increased VEGFR2/caveolin-1 but failed to increase VEGFR1/caveolin-1 association. VEGF also increased caveolin-1/Src association and activated Src, and Src inhibitor blocked RhoA activation and fibronectin upregulation. Src-mediated phosphorylation of caveolin-1 on Y14 has also been implicated in signaling responses. Overexpression of nonphosphorylatable caveolin-1 Y14A prevented VEGF-induced RhoA activation and fibronectin upregulation. In vivo, although VEGFR1 and VEGFR2 protein levels were both increased in the kidney cortices of diabetic rats, VEGFR2/caveolin-1 association but not VEGFR1/caveolin-1 association was significantly increased. In conclusion, VEGF-induced RhoA activation and fibronectin upregulation require caveolae and caveolin-1 interaction with VEGFR2 and Src. Interference with caveolin/-ae signaling may provide new avenues for the treatment of fibrotic renal disease.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, People's Republic of China
| | - Baifang Zhang
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, People's Republic of China
| | - Feng Ye
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, People's Republic of China
| | - Zeling Xiao
- Department of Biochemistry, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
47
|
Zhu L, Qi XY, Aoudjit L, Mouawad F, Baldwin C, Nattel S, Takano T. Nuclear factor of activated T cells mediates RhoA-induced fibronectin upregulation in glomerular podocytes. Am J Physiol Renal Physiol 2013; 304:F849-62. [PMID: 23389455 DOI: 10.1152/ajprenal.00495.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glomerulosclerosis is featured by accumulation of the extracellular matrixes in the glomerulus. We showed previously that activation of the small GTPase RhoA in podocytes induces heavy proteinuria and glomerulosclerosis in the mouse. In the current study, we investigated the mechanism by which RhoA stimulates the production of one of the extracellular matrixes, fibronectin, by podocytes, specifically testing the role of nuclear factor of activated T cells (NFAT). Expression of constitutively active RhoA in cultured podocytes activated the fibronectin promoter, upregulated fibronectin protein, and activated NFAT. Expression of constitutively active NFAT in podocytes also activated the fibronectin promoter and upregulated fibronectin protein. RhoA-induced NFAT activation and fibronectin upregulation were both dependent on the calcium/calmodulin pathway and Rho kinase. NFAT activation was also observed in vivo in the rat and mouse models of podocyte injury and proteinuria, and NFAT inhibition ameliorated fibronectin upregulation in the latter. RhoA activation induced a rise of intracellular calcium ion concentration ([Ca(2+)]i), which was at least in part dependent on the transient receptor potential canonical 6 (TRPC6) cation channel. The results indicate that RhoA activates NFAT by inducing a rise of [Ca(2+)]i in podocytes, which in turn contributes to fibronectin upregulation. This pathway may be responsible for the pathogenesis of certain glomerular diseases such as hypertension-mediated glomerulosclerosis.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
48
|
Fang JS, Dai C, Kurjiaka DT, Burt JM, Hirschi KK. Connexin45 regulates endothelial-induced mesenchymal cell differentiation toward a mural cell phenotype. Arterioscler Thromb Vasc Biol 2012; 33:362-8. [PMID: 23220276 DOI: 10.1161/atvbaha.112.255950] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The focus of this study was to investigate the role of connexin (Cx) 45 in endothelial-induced mural cell differentiation. METHODS AND RESULTS We created mural cell precursors that stably express only Cx45 in Cx43-deficient mesenchymal cells (ReCx45), and used our in vitro model of blood vessel assembly to assess the capacity of this Cx to support endothelial-induced mural cell differentiation. Lucifer Yellow dye injection and dual whole-cell patch clamping revealed that functional gap junctions exhibiting properties of Cx45-containing channels formed among ReCx45 transfectants, and between ReCx45 and endothelial cells. Heterocellular Cx45-containing gap junction channels enabled transforming growth factor-β activation and promoted the upregulation of mural cell-specific proteins in the mesenchymal precursors. CONCLUSIONS These studies reveal a critical role for Cx45 in the regulation of endothelial-induced mural cell differentiation, which is consistent with the phenotype of Cx45-deficient embryos that exhibit dysregulated transforming growth factor-β and lack mural cell development.
Collapse
Affiliation(s)
- Jennifer S Fang
- Yale Cardiovascular Research Center, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
49
|
Jin Q, Pulipati NR, Zhou W, Staub CM, Liotta LA, Mulder KM. Role of km23-1 in RhoA/actin-based cell migration. Biochem Biophys Res Commun 2012; 428:333-8. [PMID: 23079622 DOI: 10.1016/j.bbrc.2012.10.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
Abstract
km23-1 was originally identified as a TGFß receptor-interacting protein that plays an important role in TGFß signaling. Moreover, km23-1 is actually part of an ancient superfamily of NTPase-regulatory proteins, widely represented in archaea and bacteria. To further elucidate the function of km23-1, we identified novel protein interacting partners for km23-1 by using tandem affinity purification (TAP) and tandem mass spectrometry (MS). Here we show that km23-1 interacted with a class of proteins involved in actin-based cell motility and modulation of the actin cytoskeleton. We further showed that km23-1 modulates the formation of a highly organized stress fiber network. More significantly, we demonstrated that knockdown (KD) of km23-1 decreased RhoA activation in Mv1Lu epithelial cells. Finally, our results demonstrated for the first time that depletion of km23-1 inhibited cell migration of human colon carcinoma cells (HCCCs) in wound-healing assays. Overall, our findings demonstrate that km23-1 regulates RhoA and motility-associated actin modulating proteins, suggesting that km23-1 may represent a novel target for anti-metastatic therapy.
Collapse
Affiliation(s)
- Qunyan Jin
- Department of Biochemistry and Molecular Biology, Penn State Hershey College of Medicine, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
50
|
Maniatis NA, Chernaya O, Shinin V, Minshall RD. Caveolins and lung function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012. [PMID: 22411320 DOI: 10.1007/978-1-4614-1222-911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The primary function of the mammalian lung is to facilitate diffusion of oxygen to venous blood and to ventilate carbon dioxide produced by catabolic reactions within cells. However, it is also responsible for a variety of other important functions, including host defense and production of vasoactive agents to regulate not only systemic blood pressure, but also water, electrolyte and acid-base balance. Caveolin-1 is highly expressed in the majority of cell types in the lung, including epithelial, endothelial, smooth muscle, connective tissue cells, and alveolar macrophages. Deletion of caveolin-1 in these cells results in major functional aberrations, suggesting that caveolin-1 may be crucial to lung homeostasis and development. Furthermore, generation of mutant mice that under-express caveolin-1 results in severe functional distortion with phenotypes covering practically the entire spectrum of known lung diseases, including pulmonary hypertension, fibrosis, increased endothelial permeability, and immune defects. In this Chapter, we outline the current state of knowledge regarding caveolin-1-dependent regulation of pulmonary cell functions and discuss recent research findings on the role of caveolin-1 in various pulmonary disease states, including obstructive and fibrotic pulmonary vascular and inflammatory diseases.
Collapse
Affiliation(s)
- Nikolaos A Maniatis
- 2nd Department of Critical Care, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | | | | |
Collapse
|