1
|
Zhao R, Hu J, Wen H, Zhao J, Wang Y, Niu X, Zhang M, Wang T, Li Y. Inhibition of N-acetylglucosaminyltransferase V alleviates diabetic cardiomyopathy in mice by attenuating cardiac hypertrophy and fibrosis. Nutr Metab (Lond) 2024; 21:53. [PMID: 39080739 PMCID: PMC11290217 DOI: 10.1186/s12986-024-00797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The pathogenesis of diabetic cardiomyopathy is closely linked to abnormal glycosylation modifications. N-acetylglucosaminyltransferase V (GnT-V), which catalyzes the production of N-linked -1-6 branching of oligosaccharides, is involved in several pathophysiological mechanisms of many disorders, including cardiac hypertrophy and heart failure. However, the mechanism by which GnT-V regulates cardiac hypertrophy in diabetic cardiomyopathy is currently poorly understood. In this study, we investigated the role of GnT-V on myocardial hypertrophy in diabetic cardiomyopathy and elucidated the underlying mechanisms. MATERIAL AND METHODS Streptozotocin (STZ) was intraperitoneally injected into mice to induce diabetic cardiomyopathy. An adeno-associated virus (AAV) carrying negative control small hairpin RNA (shNC) or GnT-V-specifc small hairpin RNA (shGnT-V) was used to manipulate GnT-V expression. In our study, forty male C57BL/6J mice were randomly divided into four groups (10 mice per group): control mice with AAV-shNC, diabetic cardiomyopathy mice with AAV-shNC, control mice with AAV-shGnT-V, and diabetic cardiomyopathy mice with AAV-shGnT-V. In addition, H9C2 cells and primary neonatal cardiac fibroblasts treated with high glucose were used as a cell model of diabetes. Analysis of cardiac hypertrophy and fibrosis, as well as functional studies, were used to investigate the underlying molecular pathways. RESULTS AAV-mediated GnT-V silencing dramatically improved cardiac function and alleviated myocardial hypertrophy and fibrosis in diabetic mice. In vitro experiments demonstrated that GnT-V was elevated in cardiomyocytes and induced cardiomyocyte hypertrophy in response to high glucose stimulation. GnT-V knockdown significantly reduced the expression of the integrinβ1 signaling pathway, as evidenced by decreased downstream ERK1/2 activity, which inhibited cardiomyocyte hypertrophy accompanied by reduced ANP, BNP, and β-MHC expression. Furthermore, knocking down GnT-V expression lowered the TGF-β1-Smads signaling pathway, which reduced the expression of α-SMA, collagen I, and collagen III. CONCLUSIONS Overall, our research indicated that GnT-V may be a useful therapeutic target to treat diabetic cardiomyopathy, primarily in the inhibition of myocardial hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Jianqiang Hu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - He Wen
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Jieqiong Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Xiaona Niu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| | - Tingting Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| |
Collapse
|
2
|
Xiong X, Li W, Nam J, Qu M, Kay SA, Ma K. The actin cytoskeleton-MRTF/SRF cascade transduces cellular physical niche cues to entrain the circadian clock. J Cell Sci 2022; 135:jcs260094. [PMID: 36093830 PMCID: PMC10658898 DOI: 10.1242/jcs.260094] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
The circadian clock is entrained to daily environmental cues. Integrin-linked signaling via actin cytoskeleton dynamics transduces physical niche cues from the extracellular matrix to myocardin-related transcription factor (MRTF)/serum response factor (SRF)-mediated transcription. The actin cytoskeleton organization and SRF-MRTF activity display diurnal oscillations. By interrogating disparate upstream events in the actin cytoskeleton-MRTF-A/SRF signaling cascade, we show that this pathway transduces extracellular niche cues to modulate circadian clock function. Pharmacological inhibition of MRTF-A/SRF by disrupting actin polymerization or blocking the ROCK kinase induced period lengthening with augmented clock amplitude, and genetic loss of function of Srf or Mrtfa mimicked the effects of treatment with actin-depolymerizing agents. In contrast, actin polymerization shortened circadian clock period and attenuated clock amplitude. Moreover, interfering with the cell-matrix interaction through blockade of integrin, inhibition of focal adhesion kinase (FAK, encoded by Ptk2) or attenuating matrix rigidity reduced the period length while enhancing amplitude. Mechanistically, we identified that the core clock repressors Per2, Nr1d1 and Nfil3 are direct transcriptional targets of MRTF-A/SRF in mediating actin dynamics-induced clock response. Collectively, our findings defined an integrin-actin cytoskeleton-MRTF/SRF pathway in linking clock entrainment with extracellular cues that might facilitate cellular adaptation to the physical niche environment.
Collapse
Affiliation(s)
- Xuekai Xiong
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Weini Li
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jin Nam
- Department of Bioengineering, University of California at Riverside, Riverside, CA 92521, USA
| | - Meng Qu
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Steve A. Kay
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Deshpande A, Shetty PMV, Frey N, Rangrez AY. SRF: a seriously responsible factor in cardiac development and disease. J Biomed Sci 2022; 29:38. [PMID: 35681202 PMCID: PMC9185982 DOI: 10.1186/s12929-022-00820-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
The molecular mechanisms that regulate embryogenesis and cardiac development are calibrated by multiple signal transduction pathways within or between different cell lineages via autocrine or paracrine mechanisms of action. The heart is the first functional organ to form during development, which highlights the importance of this organ in later stages of growth. Knowledge of the regulatory mechanisms underlying cardiac development and adult cardiac homeostasis paves the way for discovering therapeutic possibilities for cardiac disease treatment. Serum response factor (SRF) is a major transcription factor that controls both embryonic and adult cardiac development. SRF expression is needed through the duration of development, from the first mesodermal cell in a developing embryo to the last cell damaged by infarction in the myocardium. Precise regulation of SRF expression is critical for mesoderm formation and cardiac crescent formation in the embryo, and altered SRF levels lead to cardiomyopathies in the adult heart, suggesting the vital role played by SRF in cardiac development and disease. This review provides a detailed overview of SRF and its partners in their various functions and discusses the future scope and possible therapeutic potential of SRF in the cardiovascular system.
Collapse
Affiliation(s)
- Anushka Deshpande
- Department of Internal Medicine III, Cardiology and Angiology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Prithviraj Manohar Vijaya Shetty
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
4
|
Bildyug N. Integrins in cardiac hypertrophy: lessons learned from culture systems. ESC Heart Fail 2021; 8:3634-3642. [PMID: 34232557 PMCID: PMC8497369 DOI: 10.1002/ehf2.13497] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/16/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Heart growth and pathological changes are accompanied by extracellular matrix‐dependent alterations in integrins and integrin‐associated proteins, suggesting their role in heart development and disease. Most of our knowledge on the involvement of integrins in heart pathology is provided by the in vivo experiments, including cardiac hypertrophy models. However, in vivo studies are limited by the complex organization of heart tissue and fail to discern cell types and particular integrins implicated in hypertrophic signalling. This problem is being addressed by isolated cardiomyocyte primary cultures, which have been successfully used in different in vitro disease models. This review aimed to analyse the general approaches to studying integrins and integrin‐associated signalling pathways in cardiac hypertrophy focusing on the in vitro systems. The lessons learned from culture experiments on the models of hypertrophy induced by stretch, stimulating factors, and/or extracellular matrix components are summarized, demonstrating the major involvement of integrin‐mediated signalling in cardiac hypertrophic response and its apparent crosstalk with signal pathways induced by stretch or hypertrophy stimulating factors. The benefits and perspectives of using cardiomyocyte primary culture as a hypertrophy model are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, 194064, Russia
| |
Collapse
|
5
|
Amaroli A, Sabbieti MG, Marchetti L, Zekiy AO, Utyuzh AS, Marchegiani A, Laus F, Cuteri V, Benedicenti S, Agas D. The effects of 808-nm near-infrared laser light irradiation on actin cytoskeleton reorganization in bone marrow mesenchymal stem cells. Cell Tissue Res 2020; 383:1003-1016. [PMID: 33159579 DOI: 10.1007/s00441-020-03306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 09/17/2020] [Indexed: 11/29/2022]
Abstract
Tailoring the cell organelles and thus changing cell homeostatic behavior has permitted the discovery of fascinating metabolic features enabling enhanced viability, differentiation, or quenching inflammation. Recently, photobiomodulation (PBM) has been accredited as an effective cell manipulation technique with promising therapeutic potential. In this prospective, in vitro results revealed that 808-nm laser light emitted by a hand-piece with a flat-top profile at an irradiation set up of 60 J/cm2 (1 W, 1 W/cm2; 60 s, continuous wave) regulates bone marrow stromal cell (BMSC) differentiation toward osteogenesis. Considering the importance of actin cytoskeleton reorganization, which controls a range of cell metabolic activities, comprising shape change, proliferation and differentiation, the aim of the current work is to assess whether PBM therapy, using a flat-top hand-piece at higher-fluence irradiation on BMSCs, is able to switch photon signals into the stimulation of biochemical/differentiating pathways involving key activators that regulate de novo actin polymerization. Namely, for the first time, we unearthed the role of the flat-top hand-piece at higher-fluence irradiation on cytoskeletal characteristics of BMSCs. These novel findings meet the needs of novel therapeutically protocols provided by laser treatment and the manipulation of BMSCs as anti-inflammatory, osteo-inductive platforms.
Collapse
Affiliation(s)
- Andrea Amaroli
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C.), University of Genova, Genova, Italy
- Department of Orthopaedic Dentistry, Sechenov First Moscow State Medical University, Trubetzkaya St., 8, Bd. 2, 119991, Moscow, Russian Federation
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Luigi Marchetti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Angelina O Zekiy
- Department of Orthopaedic Dentistry, Sechenov First Moscow State Medical University, Trubetzkaya St., 8, Bd. 2, 119991, Moscow, Russian Federation
| | - Anatoliy S Utyuzh
- Department of Orthopaedic Dentistry, Sechenov First Moscow State Medical University, Trubetzkaya St., 8, Bd. 2, 119991, Moscow, Russian Federation
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Vincenzo Cuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Stefano Benedicenti
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C.), University of Genova, Genova, Italy
| | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy.
| |
Collapse
|
6
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
7
|
Boppart MD, Mahmassani ZS. Integrin signaling: linking mechanical stimulation to skeletal muscle hypertrophy. Am J Physiol Cell Physiol 2019; 317:C629-C641. [PMID: 31314586 DOI: 10.1152/ajpcell.00009.2019] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The α7β1-integrin is a transmembrane adhesion protein that connects laminin in the extracellular matrix (ECM) with actin in skeletal muscle fibers. The α7β1-integrin is highly expressed in skeletal muscle and is concentrated at costameres and myotendious junctions, providing the opportunity to transmit longitudinal and lateral forces across the membrane. Studies have demonstrated that α7-integrin subunit mRNA and protein are upregulated following eccentric contractions as a mechanism to reinforce load-bearing structures and resist injury with repeated bouts of exercise. It has been hypothesized for many years that the integrin can also promote protein turnover in a manner that can promote beneficial adaptations with resistance exercise training, including hypertrophy. This review provides basic information about integrin structure and activation and then explores its potential to serve as a critical mechanosensor and activator of muscle protein synthesis and growth. Overall, the hypothesis is proposed that the α7β1-integrin can contribute to mechanical-load induced skeletal muscle growth via an mammalian target of rapamycin complex 1-independent mechanism.
Collapse
Affiliation(s)
- Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah
| |
Collapse
|
8
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
9
|
Exercise and the control of muscle mass in human. Pflugers Arch 2018; 471:397-411. [DOI: 10.1007/s00424-018-2217-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
|
10
|
Civitarese RA, Kapus A, McCulloch CA, Connelly KA. Role of integrins in mediating cardiac fibroblast–cardiomyocyte cross talk: a dynamic relationship in cardiac biology and pathophysiology. Basic Res Cardiol 2016; 112:6. [DOI: 10.1007/s00395-016-0598-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/14/2016] [Indexed: 12/16/2022]
|
11
|
Shen L, Qin K, Wang D, Zhang Y, Bai N, Yang S, Luo Y, Xiang R, Tan X. Overexpression of Oct4 suppresses the metastatic potential of breast cancer cells via Rnd1 downregulation. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2087-95. [PMID: 25068817 DOI: 10.1016/j.bbadis.2014.07.015] [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: 03/24/2014] [Revised: 07/07/2014] [Accepted: 07/21/2014] [Indexed: 02/05/2023]
Abstract
Although Oct4 is known as a critical transcription factor involved in maintaining "stemness", its role in tumor metastasis is still controversial. Herein, we overexpressed and silenced Oct4 expression in two breast cancer cell lines, MDA-MB-231 and 4T1, separately. Our data showed that ectopic overexpression of Oct4 suppressed cell migration and invasion in vitro and the formation of metastatic lung nodules in vivo. Conversely, Oct4 downregulation increased the metastatic potential of breast cancer cells both in vitro and in vivo. Furthermore, we identified Rnd1 as the downstream target of Oct4 by ribonucleic acid sequencing (RNA-seq) analysis, which was significantly downregulated upon Oct4 overexpression. Chromatin immunoprecipitation assays revealed the binding of Oct4 to the promoter region of Rnd1 by ectopic overexpression of Oct4. Dual luciferase assays indicated that Oct4 overexpression suppressed transcriptional activity of the Rnd1 promoter. Moreover, overexpression of Rnd1 partially rescued the inhibitory effects of Oct4 on the migration and invasion of breast cancer cells. Overexpression of Rnd1 counteracted the influence of Oct4 on the formation of cell adhesion and lamellipodia, which implied a potential underlying mechanism involving Rnd1. In addition, we also found that overexpression of Oct4 led to an elevation of E-cadherin expression, even in 4T1 cells that possess a relatively high basal level of E-cadherin. Rnd1 overexpression impaired the promoting effects of Oct4 on E-cadherin expression in MDA-MB-231 cells. These results suggest that Oct4 affects the metastatic potential of breast cancer cells through Rnd1-mediated effects that influence cell motility and E-cadherin expression.
Collapse
Affiliation(s)
- Long Shen
- Department of Pathology, Medical School of Nankai University, Tianjin 300071, China
| | - Kunhua Qin
- Department of Immunology, Medical School of Nankai University, Tianjin 300071, China
| | - Dekun Wang
- Department of Pathology, Medical School of Nankai University, Tianjin 300071, China
| | - Yan Zhang
- Department of Immunology, Medical School of Nankai University, Tianjin 300071, China
| | - Nan Bai
- Department of Immunology, Medical School of Nankai University, Tianjin 300071, China
| | - Shengyong Yang
- West China Hospital, Molecular Medicine Research Centre, State Key Lab Biotherapy, Sichuan University, Chengdu 610064, China
| | - Yunping Luo
- Department of Immunology, Beijing Union Medical School, Beijing 100010, China
| | - Rong Xiang
- Department of Immunology, Medical School of Nankai University, Tianjin 300071, China
| | - Xiaoyue Tan
- Department of Pathology, Medical School of Nankai University, Tianjin 300071, China.
| |
Collapse
|
12
|
Martin TM, Plautz SA, Pannier AK. Network analysis of endogenous gene expression profiles after polyethyleneimine-mediated DNA delivery. J Gene Med 2013; 15:142-54. [PMID: 23526566 DOI: 10.1002/jgm.2704] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/08/2013] [Accepted: 03/15/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND DNA delivery systems, which transport exogenous DNA to cells, have applications that include gene therapy, tissue engineering and medical devices. Although the cationic nonviral DNA carrier polyethyleneimine (PEI) has been widely studied, the molecular factors and pathways underlying PEI-mediated DNA transfer remain largely unknown, preventing the design of more efficient delivery systems. METHODS HEK 293 T cells were treated with polyplexes formed with PEI and pEGFPLuc encoding for green fluorescent protein (GFP). Transfected cells expressing GFP were flow-separated from treated, untransfected cells. Gene expression profiles were obtained using Affymetrix HG-U133 2.0 microarrays and differentially expressed genes were identified using R/Bioconductor. Gene network analysis using EGAN (exploratory gene association network) bioinformatics tools was then used to find interaction among genes and enriched gene ontology (GO) terms related to transfection. Genes identified by this method were perturbed using pharmacologic activators or inhibitors to assess their effect on DNA transfer. RESULTS Microarray analysis comparing transfected cells to untransfected cells revealed 215 genes to be differentially expressed, with the majority enriched to GO processes including metabolism, response to stimulus, cell cycle, biological regulation and cellular component organization or biogenesis pathways. Gene network analysis revealed a coordinated induction of RAP1A, SCG5, PGAP1, ATF3 and NEB genes implicated in cell stress, cell cycle and cytoskeletal processes. Altering pathways with pharmacologic agents confirmed the potential role of RAP1A, SCG5 and ATF3 in transfection. CONCLUSIONS Microarray and gene network analyses of the sorted, transfected cell population can identify potential mediators of transfection, providing a basis for the design of improved delivery systems.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | |
Collapse
|
13
|
Zhang X, Li G, Zhang L, Yang D, Zhang Z, Yan A, Linghu H. C3G overexpression promotes the survival of rat-derived H9C2 cardiomyocytes by p-ERK1/2. Cell Biol Int 2013; 37:1106-13. [PMID: 23686869 DOI: 10.1002/cbin.10136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 04/26/2013] [Indexed: 11/08/2022]
Affiliation(s)
| | - Gang Li
- Division of Cardiology, Department of Geriatrics; The First Affiliated Hospital of Chongqing Medical University; No. 1 Yixueyuan Road; Yuzhong District, Chongqing; 400016; China
| | | | | | | | | | - Hua Linghu
- Department of Gynecology and Obstetrics; The First Affiliated Hospital of Chongqing Medical University; Chongqing; 400016; China
| |
Collapse
|
14
|
Rajagopalan V, Zhao M, Reddy S, Fajardo G, Wang X, Dewey S, Gomes AV, Bernstein D. Altered ubiquitin-proteasome signaling in right ventricular hypertrophy and failure. Am J Physiol Heart Circ Physiol 2013; 305:H551-62. [PMID: 23729213 DOI: 10.1152/ajpheart.00771.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alterations in the ubiquitin-proteasome system (UPS) have been described in left ventricular hypertrophy and failure, although results have been inconsistent. The role of the UPS in right ventricular (RV) hypertrophy (RVH) and RV failure (RVF) is unknown. Given the greater percent increase in RV mass associated with RV afterload stress, as present in many congenital heart lesions, we hypothesized that alterations in the UPS could play an important role in RVH/RVF. UPS expression and activity were measured in the RV from mice with RVH/RVF secondary to pulmonary artery constriction (PAC). Epoxomicin and MG132 were used to inhibit the proteasome, and overexpression of the 11S PA28α subunit was used to activate the proteasome. PAC mice developed RVH (109.3% increase in RV weight to body weight), RV dilation with septal shift, RV dysfunction, and clinical RVF. Proteasomal function (26S β₅ chymotrypsin-like activity) was decreased 26% (P < 0.05). Protein expression of 19S subunit Rpt5 (P < 0.05), UCHL1 deubiquitinase (P < 0.0001), and Smurf1 E3 ubiquitin ligase (P < 0.01) were increased, as were polyubiquitinated proteins (P < 0.05) and free-ubiquitins (P = 0.05). Pro-apoptotic Bax was increased (P < 0.0001), whereas anti-apoptotic Bcl-2 decreased (P < 0.05), resulting in a sixfold increase in the Bax/Bcl-2 ratio. Proteasomal inhibition did not accelerate RVF. However, proteasome enhancement by cardiac-specific proteasome overexpression partially improved survival. Proteasome activity is decreased in RVH/RVF, associated with upregulation of key UPS regulators and pro-apoptotic signaling. Enhancement of proteasome function partially attenuates RVF, suggesting that UPS dysfunction contributes to RVF.
Collapse
Affiliation(s)
- Viswanathan Rajagopalan
- Department of Pediatrics (Cardiology Stanford University School of Medicine, Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Yan A, Li G, Zhang X, Zhu B, Linghu H. Pro-survival effect of Dock180 overexpression on rat-derived H9C2 cardiomyocytes. Med Sci Monit Basic Res 2013; 19:12-9. [PMID: 23314417 PMCID: PMC3638688 DOI: 10.12659/msmbr.883738] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Integrin β1 subunit and its downstream molecule, focal adhesion kinase (FAK), have been demonstrated to be indispensible to the promotion of cell proliferation and survival and anti-apoptosis in cardiomyocytes via activation of their downstream pro-survival signaling molecule, AKT. As a component of the integrin pathway, Dock180 (dedicator of cytokinesis 1) protein is also thought to be involved in the promotion of cell proliferation and survival and anti-apoptosis in the H9C2 cardiomyocytes. Material/Methods Rat-derived H9C2 cardiomyocytes were transfected with pCXN2-flag-hDock180, a human Dock180 overexpression eukaryotic recombinant plasmid. The rat and human Dock180 mRNA and protein expression, apoptosis and cell proliferation and survival were analyzed in the H9C2 cardiomyocytes treated with either hypoxia/reoxygenation (H/R) or not, respectively. Results Human Dock180 mRNA overexpression could significantly increase the Dock180 protein expression in the H9C2 cardiomyocytes, no matter whether treated with H/R or not. Dock180 overexpression could promote the cell proliferation and survival and anti-apoptosis, and relieve the cell proliferative and survival inhibition and apoptosis induced by H/R in the H9C2 cardiomyocytes via activation of its downstream pro-survival signaling molecule AKT. Conclusions Dock180 could act as a pro-survival molecule in H9C2 cardiomyocytes via activation of its downstream pro-survival signaling molecule, AKT.
Collapse
Affiliation(s)
- An Yan
- Division of Cardiology, Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | | | | | | | | |
Collapse
|
16
|
Malik M, Segars J, Catherino WH. Integrin β1 regulates leiomyoma cytoskeletal integrity and growth. Matrix Biol 2012; 31:389-97. [PMID: 23023061 DOI: 10.1016/j.matbio.2012.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 09/14/2012] [Accepted: 09/18/2012] [Indexed: 11/17/2022]
Abstract
Uterine leiomyomas are characterized by an excessive extracellular matrix, increased mechanical stress, and increased active RhoA. Previously, we observed that mechanical signaling was attenuated in leiomyoma, but the mechanisms responsible remain unclear. Integrins, especially integrin β1, are transmembrane adhesion receptors that couple extracellular matrix stresses to the intracellular cytoskeleton to influence cell proliferation and differentiation. Here we characterized integrin and laminin to signaling in leiomyoma cells. We observed a 2.25±0.32 fold increased expression of integrin β1 in leiomyoma cells, compared to myometrial cells. Antibody-mediated inhibition of integrin β1 led to significant growth inhibition in leiomyoma cells and a loss of cytoskeletal integrity. Specifically, polymerization of actin filaments and formation of focal adhesions were reduced by inhibition of integrin β1. Inhibition of integrin β1 in leiomyoma cells led to 0.81±0.02 fold decrease in active RhoA, and resembled levels found in serum-starved cells. Likewise, inhibition of integrin β1 was accompanied by a decrease in phospho-ERK. Compared to myometrial cells, leiomyoma cells demonstrated increased expression of integrin α6 subunit to laminin receptor (1.91±0.11 fold), and increased expression of laminin 5α (1.52±0.02), laminin 5β (3.06±0.92), and laminin 5γ (1.66±0.06). Of note, leiomyoma cells grown on laminin matrix appear to realign themselves. Taken together, the findings reveal that the attenuated mechanical signaling in leiomyoma cells is accompanied by an increased expression and a dependence on integrin β1 signaling in leiomyoma cells, compared to myometrial cells.
Collapse
Affiliation(s)
- Minnie Malik
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | | | | |
Collapse
|
17
|
Kim JH, Lee MC, Seong SC, Park KH, Lee S. Enhanced Proliferation and Chondrogenic Differentiation of Human Synovium-Derived Stem Cells Expanded with Basic Fibroblast Growth Factor. Tissue Eng Part A 2011; 17:991-1002. [DOI: 10.1089/ten.tea.2010.0277] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ji Hyun Kim
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Myung Chul Lee
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Cheol Seong
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Ho Park
- Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Sahnghoon Lee
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Tsai FC, Pai MH, Chiu CC, Chou CM, Hsieh MS. Denervation dynamically regulates integrin alpha7 signaling pathways and microscopic structures in rats. THE JOURNAL OF TRAUMA 2011; 70:220-227. [PMID: 21268308 DOI: 10.1097/ta.0b013e3181e4d558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Peripheral nerve injury causes serious problems in orthopedic and plastic surgeries. Cell adhesion molecules such as integrin alpha7 provoke cell binding and signaling pathways within myofibers. Expression profiles of integrin alpha7 signaling pathways and the molecule's microscopic structure were assessed to investigate the long-term dynamic changes in denervated rat skeletal muscle. METHODS A denervated rat skeletal muscle model was established by severing the sciatic nerve for 1 week, 2 weeks, 4 weeks, 8 weeks, 12 weeks, 20 weeks, and 26 weeks. Molecular expressions were investigated by mRNA and Western blot. The structural alterations were detected by immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. RESULTS The denervated muscle atrophy presented the following dynamic molecular alterations: an initial increase around postdenervation in week (PIW) 8 and then a subsequent decay of integrin alpha7, integrin downstream signaling pathway (Ras or Raf or, ERK1/2), Akt, cleaved caspase-3, fast myosin heavy chain (MHC), beta actin, and RhoA. We demonstrated that the expressions of multiple signaling molecules were highly upregulated at PIW 8 (p<0.01). Scanning electron microscopy findings of the surface textures of myofibers showed more severe damage at PIW 8 and subsequently became smoother. Inner structures of myofibers separated with discontinuity on transmission electron microscopy examinations. CONCLUSION Our novel finding showed that time-series alterations of integrin alpha7 signaling molecules and surface microstructures in the long-term denervated rat skeletal muscle are biphasic and coherently dynamic. Persisted p-Akt elevation suggested that denervated muscle may regenerate if reinnervation or other treatment was performed.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/physiology
- Blotting, Western
- Female
- Integrin alpha Chains/biosynthesis
- Integrin alpha Chains/physiology
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Muscle Denervation
- Muscle Fibers, Skeletal/diagnostic imaging
- Muscle Fibers, Skeletal/physiology
- Muscle, Skeletal/innervation
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Muscle, Skeletal/ultrastructure
- Polymerase Chain Reaction
- Rats
- Signal Transduction/physiology
- Ultrasonography
Collapse
Affiliation(s)
- Feng-Chou Tsai
- Center for Mathematical Biology, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, and Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
19
|
Li Q, Song XW, Zou J, Wang GK, Kremneva E, Li XQ, Zhu N, Sun T, Lappalainen P, Yuan WJ, Qin YW, Jing Q. Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. J Cell Sci 2010; 123:2444-2452. [PMID: 20571053 DOI: 10.1242/jcs.067165] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs are involved in several aspects of cardiac hypertrophy, including cardiac growth, conduction, and fibrosis. However, their effects on the regulation of the cardiomyocyte cytoskeleton in this pathological process are not known. Here, with microRNA microarray and small RNA library sequencing, we show that microRNA-1 (miR-1) is the most abundant microRNA in the human heart. By applying bioinformatic target prediction, a cytoskeleton regulatory protein twinfilin-1 was identified as a potential target of miR-1. Overexpression of miR-1 not only reduced the luciferase activity of the reporter containing the 3' untranslated region of twinfilin-1 mRNA, but also suppressed the endogenous protein expression of twinfilin-1, indicating that twinfilin-1 is a direct target of miR-1. miR-1 was substantially downregulated in the rat hypertrophic left ventricle and phenylephrine-induced hypertrophic cardiomyocytes, and accordingly, the protein level of twinfilin-1 was increased. Furthermore, overexpression of miR-1 in hypertrophic cardiomyocytes reduced the cell size and attenuated the expression of hypertrophic markers, whereas silencing of miR-1 in cardiomyocytes resulted in the hypertrophic phenotype. In accordance, twinfilin-1 overexpression promoted cardiomyocyte hypertrophy. Taken together, our results demonstrate that the cytoskeleton regulatory protein twinfilin-1 is a novel target of miR-1, and that reduction of miR-1 by hypertrophic stimuli induces the upregulation of twinfilin-1, which in turn evokes hypertrophy through the regulation of cardiac cytoskeleton.
Collapse
Affiliation(s)
- Qing Li
- Key Laboratory of Stem Cell Biology and Laboratory of Nucleic Acid and Molecular Medicine, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Mayers CM, Wadell J, McLean K, Venere M, Malik M, Shibata T, Driggers PH, Kino T, Guo XC, Koide H, Gorivodsky M, Grinberg A, Mukhopadhyay M, Abu-Asab M, Westphal H, Segars JH. The Rho guanine nucleotide exchange factor AKAP13 (BRX) is essential for cardiac development in mice. J Biol Chem 2010; 285:12344-54. [PMID: 20139090 PMCID: PMC2852973 DOI: 10.1074/jbc.m110.106856] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Indexed: 11/06/2022] Open
Abstract
A fundamental biologic principle is that diverse biologic signals are channeled through shared signaling cascades to regulate development. Large scaffold proteins that bind multiple proteins are capable of coordinating shared signaling pathways to provide specificity to activation of key developmental genes. Although much is known about transcription factors and target genes that regulate cardiomyocyte differentiation, less is known about scaffold proteins that couple signals at the cell surface to differentiation factors in developing heart cells. Here we show that AKAP13 (also known as Brx-1, AKAP-Lbc, and proto-Lbc), a unique protein kinase A-anchoring protein (AKAP) guanine nucleotide exchange region belonging to the Dbl family of oncogenes, is essential for cardiac development. Cardiomyocytes of Akap13-null mice had deficient sarcomere formation, and developing hearts were thin-walled and mice died at embryonic day 10.5-11.0. Disruption of Akap13 was accompanied by reduced expression of Mef2C. Consistent with a role of AKAP13 upstream of MEF2C, Akap13 siRNA led to a reduction in Mef2C mRNA, and overexpression of AKAP13 augmented MEF2C-dependent reporter activity. The results suggest that AKAP13 coordinates Galpha(12) and Rho signaling to an essential transcription program in developing cardiomyocytes.
Collapse
Affiliation(s)
| | - Jennifer Wadell
- From the Program in Reproductive and Adult Endocrinology and
| | - Kate McLean
- From the Program in Reproductive and Adult Endocrinology and
| | - Monica Venere
- From the Program in Reproductive and Adult Endocrinology and
| | - Minnie Malik
- the Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | | | | | - Tomoshige Kino
- From the Program in Reproductive and Adult Endocrinology and
| | | | - Hisashi Koide
- From the Program in Reproductive and Adult Endocrinology and
| | | | - Alex Grinberg
- the Laboratory of Mammalian Genes and Development, NICHD, and
| | | | - Mones Abu-Asab
- the Laboratory of Pathology, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Heiner Westphal
- the Laboratory of Mammalian Genes and Development, NICHD, and
| | - James H. Segars
- From the Program in Reproductive and Adult Endocrinology and
| |
Collapse
|
21
|
SHP2 mediates gp130-dependent cardiomyocyte hypertrophy via negative regulation of skeletal alpha-actin gene. J Mol Cell Cardiol 2010; 49:157-64. [PMID: 20226789 DOI: 10.1016/j.yjmcc.2010.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/27/2010] [Accepted: 03/02/2010] [Indexed: 11/21/2022]
Abstract
Morphological and biochemical phenotypes of cardiomyocyte hypertrophy are determined by neurohumoral factors. Stimulation of G protein-coupled receptor (GPCR) results in uniform cell enlargement in all directions with an increase in skeletal alpha-actin (alpha-SKA) gene expression, while stimulation of gp130 receptor by interleukin-6 (IL-6)-related cytokines induces longitudinal elongation with no increase in alpha-SKA gene expression. Thus, alpha-SKA is a discriminating marker for hypertrophic phenotypes; however, regulatory mechanisms of alpha-SKA gene expression remain unknown. Here, we clarified the role of SH2-containing protein tyrosine phosphatase 2 (SHP2) in alpha-SKA gene expression. In neonatal rat cardiomyocytes, endothelin-1 (ET-1), a GPCR agonist, but not leukemia inhibitory factor (LIF), an IL-6-related cytokine, induced RhoA activation and promotes alpha-SKA gene expression via RhoA. In contrast, LIF, but not ET-1, induced activation of SHP2 in cardiomyocytes, suggesting that SHP2 might negatively regulate alpha-SKA gene expression downstream of gp130. Therefore, we examined the effect of adenovirus-mediated overexpression of wild-type SHP2 (SHP2(WT)), dominant-negative SHP2 (SHP2(C/S)), or beta-galactosidase (beta-gal), on alpha-SKA gene expression. LIF did not upregulate alpha-SKA mRNA in cardiomyocytes overexpressing either beta-gal or SHP2(WT). In cardiomyocytes overexpressing SHP2(C/S), LIF induced upregulation of alpha-SKA mRNA, which was abrogated by concomitant overexpression of either C3-toxin or dominant-negative RhoA. RhoA was activated after LIF stimulation in the cardiomyocytes overexpressing SHP2(C/S), but not in myocytes overexpressing beta-gal. Furthermore, SHP2 mediates LIF-induced longitudinal elongation of cardiomyocytes via ERK5 activation. Collectively, these findings indicate that SHP2 negatively regulates alpha-SKA expression via RhoA inactivation and suggest that SHP2 implicates ERK5 in cardiomyocyte elongation downstream of gp130.
Collapse
|
22
|
Hoe HS, Lee KJ, Carney RSE, Lee J, Markova A, Lee JY, Howell BW, Hyman BT, Pak DTS, Bu G, Rebeck GW. Interaction of reelin with amyloid precursor protein promotes neurite outgrowth. J Neurosci 2009; 29:7459-73. [PMID: 19515914 PMCID: PMC2759694 DOI: 10.1523/jneurosci.4872-08.2009] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 03/19/2009] [Accepted: 04/15/2009] [Indexed: 01/12/2023] Open
Abstract
The processing of amyloid precursor protein (APP) to Abeta is an important event in the pathogenesis of Alzheimer's disease, but the physiological function of APP is not well understood. Our previous work has shown that APP processing and Abeta production are regulated by the extracellular matrix protein Reelin. In the present study, we examined whether Reelin interacts with APP, and the functional consequences of that interaction in vitro. Using coimmunoprecipitation, we found that Reelin interacted with APP through the central domain of Reelin (repeats 3-6) and the E1 extracellular domain of APP. Reelin increased cell surface levels of APP and decreased endocytosis of APP in hippocampal neurons in vitro. In vivo, Reelin levels were increased in brains of APP knock-out mice and decreased in APP-overexpressing mice. RNA interference knockdown of APP decreased neurite outgrowth in vitro and prevented Reelin from increasing neurite outgrowth. Knock-out of APP or Reelin decreased dendritic arborization in cortical neurons in vivo, and APP overexpression increased dendritic arborization. APP and Reelin have previously been shown to promote neurite outgrowth through interactions with integrins. We confirmed that APP interacted with alpha3beta1 integrin, and alpha3beta1 integrin altered APP trafficking and processing. Addition of an alpha3beta1 integrin antibody prevented APP and Reelin-induced neurite outgrowth. These findings demonstrate that Reelin interacts with APP, potentially having important effects on neurite development.
Collapse
MESH Headings
- Amyloid beta-Protein Precursor/genetics
- Amyloid beta-Protein Precursor/metabolism
- Animals
- Brain/physiology
- COS Cells
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Chlorocebus aethiops
- Dendrites/physiology
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Humans
- Integrin alpha3beta1/metabolism
- Integrin beta1/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Neurologic Mutants
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neurites/physiology
- Protease Nexins
- Rats
- Rats, Sprague-Dawley
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reelin Protein
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
Collapse
Affiliation(s)
| | - Kea Joo Lee
- Pharmacology, Georgetown University Medical Center, Washington, DC 20057-1464
| | - Rosalind S. E. Carney
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010-2970
| | - Jiyeon Lee
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Alexandra Markova
- Department of Neurology, Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disorders, Charlestown, Massachusetts 02129, and
| | - Ji-Yun Lee
- Pharmacology, Georgetown University Medical Center, Washington, DC 20057-1464
| | - Brian W. Howell
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke Porter Neuroscience Research Center, National Institutes of Health, Bethesda, Maryland 20892-3705
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disorders, Charlestown, Massachusetts 02129, and
| | - Daniel T. S. Pak
- Pharmacology, Georgetown University Medical Center, Washington, DC 20057-1464
| | - Guojun Bu
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110
| | | |
Collapse
|
23
|
Higuchi C, Nakamura N, Yoshikawa H, Itoh K. Transient dynamic actin cytoskeletal change stimulates the osteoblastic differentiation. J Bone Miner Metab 2009; 27:158-67. [PMID: 19183835 DOI: 10.1007/s00774-009-0037-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/11/2008] [Indexed: 11/26/2022]
Abstract
Dynamic cytoskeletal changes appear to be one of intracellular signals that control cell differentiation. To test this hypothesis, we examined the effects of short-term actin cytoskeletal changes on osteoblastic differentiation. We found an actin polymerization interfering reagent, cytochalasin D, promoted osteoblastic differentiation in mouse preosteoblastic MC3T3-E1 cells. We also found that these effects were mediated by the protein kinase D (PKD) pathway. Short-term cytochalasin D treatment increased alkaline phosphatase (ALP) activity, osteocalcin (OCN) secretion, and mineralization of the extracellular matrix in MC3T3-E1 cells, with temporary changes in actin cytoskeleton. Furthermore, the disruption of actin cytoskeleton induced phosphorylation of 744/748 serine within the activation loop of PKD in a dose-dependent manner. The protein kinase C (PKC)/PKD inhibitor Go6976 suppressed cytochalasin D-induced acceleration of osteoblastic differentiation, whereas Go6983, a specific inhibitor of conventional PKCs, did not. Involvement of PKD signaling was confirmed by using small interfering RNA to knock down PKD. In addition, another actin polymerization interfering reagent, latrunculin B, also stimulated ALP activity and OCN secretion with PKD activation. On the other hand, the present data suggested that transient dynamic actin cytoskeletal reorganization could be a novel cellular signal that directly stimulated osteoblastic differentiation.
Collapse
Affiliation(s)
- Chikahisa Higuchi
- Department of Orthopedic Surgery, Osaka University Medical School, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
24
|
Zhang SJ, Truskey GA, Kraus WE. Effect of cyclic stretch on β1D-integrin expression and activation of FAK and RhoA. Am J Physiol Cell Physiol 2007; 292:C2057-69. [PMID: 17267546 DOI: 10.1152/ajpcell.00493.2006] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Integrins play a pivotal role in proliferation, differentiation, and survival in skeletal and cardiac myocytes. The β1D-isoform of the β1-integrin is specifically expressed in striated skeletal muscle. However, little is known about the role and the mechanisms by which the splice variant β1D-integrin regulates myogenesis and mechanotransduction. We observed that cyclic mechanical stretch increases β1D-integrin protein levels and activates the downstream cytoskeletal signaling proteins focal adhesion kinase (FAK) and RhoA. Elimination of native β1D-integrin expression by RNA interference in immature developing myoblasts abolished stretch-induced increases in FAK phosphorylation and further downregulated RhoA activity. Blocking of β1D-integrin expression prevented myocellular fusion to form multinucleated mature myotubes. Restoration of human β1D-integrin expression in β1D-integrin-deficient cells partially restored myotube formation. The onset of myofusion also requires the generation of nitric oxide (NO). The release of NO affects cytoskeletal proteins by mediating RhoA activity and protein degradation. Our previous study demonstrated that stretch-induced NO positively modulates mechanical properties of differentiating skeletal myocytes. We found a significant decrease in NO production and apparent elastic modulus in β1D-integrin-deficient cells, suggesting signaling interactions between β1D-integrin and neuronal NO synthase to mediate mechanotransduction and myogenesis in skeletal myocytes. These results suggest that, in addition to regulating differentiation, the β1D-integrin isoform plays a critical role in the response of skeletal myoblasts to cyclic stretch by activating the downstream components of FAK and RhoA activity and affecting NO release.
Collapse
|
25
|
Shen X, Hong MS, Moss J, Vaughan M. BIG1, a brefeldin A-inhibited guanine nucleotide-exchange protein, is required for correct glycosylation and function of integrin beta1. Proc Natl Acad Sci U S A 2007; 104:1230-5. [PMID: 17227842 PMCID: PMC1783117 DOI: 10.1073/pnas.0610535104] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glycosylation of beta1 integrin (beta1) in the Golgi complex has been related to its function in multiple cell processes, e.g., invasiveness, matrix adhesion, and migration. Brefeldin A-inhibited guanine nucleotide-exchange proteins (BIG) 1 and BIG2 activate human ADP-ribosylation factors (ARF) 1 and ARF3 by catalyzing the replacement of ARF-bound GDP with GTP to regulate Golgi vesicular transport. We show here a requirement for BIG1 (but not BIG2) in glycosylation and function of beta1. In HepG2 cells treated for 48 or 72 h with BIG1, but not BIG2, siRNA, both the amount and electrophoretic mobility of the initially 130-kDa beta1 were increased. BIG1 content had risen by 48 h after removal of BIG1 siRNA, and the faster-migrating, aberrant 130-kDa beta1 was not seen. Peptide N-glycosidase F, but not endoglycosidase H, digestion converted all beta1 to an approximately 85-kDa (core protein) form. By electron microscopy, Golgi membranes in BIG1-depleted cells were less sharply defined than those in mock or BIG2 siRNA-treated cells, with more vesicle-like structures at the transface. Amounts of active RhoA-GTP also were decreased in such cells and restored by overexpression of HA-BIG1. Aberrant beta1 was present on the cell surface, but its function in cell spreading, adhesion, and migration was impaired. By immunofluorescence microscopy, BIG1 siRNA-treated cells showed less spreading and concentration of beta1 at the cell surface. These results indicate a previously unrecognized role for BIG1 in the glycosylation of beta1 by Golgi enzymes, which is critical for its function in developmental and other vital cell processes.
Collapse
Affiliation(s)
- Xiaoyan Shen
- *Pulmonary-Critical Care Medicine Branch
- To whom correspondence may be addressed at:
National Institutes of Health, Building 10, Room 5N307, MSC 1434, Bethesda, MD 20892-1434. E-mail:
or
| | - Myoung-Soon Hong
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Joel Moss
- *Pulmonary-Critical Care Medicine Branch
| | - Martha Vaughan
- *Pulmonary-Critical Care Medicine Branch
- To whom correspondence may be addressed at:
National Institutes of Health, Building 10, Room 5N307, MSC 1434, Bethesda, MD 20892-1434. E-mail:
or
| |
Collapse
|
26
|
Missan S, Linsdell P, McDonald TF. Role of kinases and G-proteins in the hyposmotic stimulation of cardiac IKs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1758:1641-52. [PMID: 16836976 DOI: 10.1016/j.bbamem.2006.05.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 05/19/2006] [Accepted: 05/30/2006] [Indexed: 12/16/2022]
Abstract
Exposure of cardiac myocytes to hyposmotic solution stimulates slowly-activating delayed-rectifying K(+) current (I(Ks)) via unknown mechanisms. In the present study, I(Ks) was measured in guinea-pig ventricular myocytes that were pretreated with modulators of cell signaling processes, and then exposed to hyposmotic solution. Pretreatment with compounds that (i) inhibit serine/threonine kinase activity (10-100 microM H89; 200 microM H8; 50 microM H7; 1 microM bisindolylmaleimide I; 10 microM LY294002; 50 microM PD98059), (ii) stimulate serine/threonine kinase activity (1-5 microM forskolin; 0.1 microM phorbol-12-myristate-13-acetate; 10 microM acetylcholine; 0.1 microM angiotensin II; 20 microM ATP), (iii) suppress G-protein activation (10 mM GDPbetaS), or (iv) disrupt the cytoskeleton (10 microM cytochalasin D), had little effect on the stimulation of I(Ks) by hyposmotic solution. In marked contrast, pretreatment with tyrosine kinase inhibitor tyrphostin A25 (20 microM) strongly attenuated both the hyposmotic stimulation of I(Ks) in myocytes and the hyposmotic stimulation of current in BHK cells co-expressing Ks channel subunits KCNQ1 and KCNE1. Since attenuation of hyposmotic stimulation was not observed in myocytes and cells pretreated with inactive tyrphostin A1, we conclude that TK has an important role in the response of cardiac Ks channels to hyposmotic solution.
Collapse
Affiliation(s)
- Sergey Missan
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4H7
| | | | | |
Collapse
|
27
|
Miano JM, Long X, Fujiwara K. Serum response factor: master regulator of the actin cytoskeleton and contractile apparatus. Am J Physiol Cell Physiol 2006; 292:C70-81. [PMID: 16928770 DOI: 10.1152/ajpcell.00386.2006] [Citation(s) in RCA: 374] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serum response factor (SRF) is a highly conserved and widely expressed, single copy transcription factor that theoretically binds up to 1,216 permutations of a 10-base pair cis element known as the CArG box. SRF-binding sites were defined initially in growth-related genes. Gene inactivation or knockdown studies in species ranging from unicellular eukaryotes to mice have consistently shown loss of SRF to be incompatible with life. However, rather than being critical for proliferation and growth, these genetic studies point to a crucial role for SRF in cellular migration and normal actin cytoskeleton and contractile biology. In fact, recent genomic studies reveal nearly half of the >200 SRF target genes encoding proteins with functions related to actin dynamics, lamellipodial/filopodial formation, integrin-cytoskeletal coupling, myofibrillogenesis, and muscle contraction. SRF has therefore emerged as a dispensable transcription factor for cellular growth but an absolutely essential orchestrator of actin cytoskeleton and contractile homeostasis. This review summarizes the recent genomic and genetic analyses of CArG-SRF that support its role as an ancient, master regulator of the actin cytoskeleton and contractile machinery.
Collapse
Affiliation(s)
- Joseph M Miano
- Cardiovascular Research Institute, University of Rochester School of Medicine, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
28
|
Lee JH, Sun D, Cho KJ, Kim MS, Hong MH, Kim IK, Lee JS, Lee JH. Overexpression of human 27 kDa heat shock protein in laryngeal cancer cells confers chemoresistance associated with cell growth delay. J Cancer Res Clin Oncol 2006; 133:37-46. [PMID: 16906418 DOI: 10.1007/s00432-006-0143-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Accepted: 07/05/2006] [Indexed: 10/25/2022]
Abstract
PURPOSE Among the family of heat shock proteins (HSPs), HSP70 and HSP27 have been implicated in tumorigenesis and chemoresistance, probably via the prevention of apoptosis. HSP27 levels are frequently increased in large populations of tumors of the head and neck, but the mechanism of its chemoresistance is not yet fully understood. In the present study, the role of HSP27 in the resistance to cytotoxic stress was studied in Hep-2 human laryngeal cancer cells. METHOD We established a Hep-2 cell line overexpressing HSP27 and examined whether the expression of HSP27 provides resistance to heat shock and several cytotoxic agents using a MTT colorimetic assay. Cell cycle progression was assessed by flow cytometry and fluorescence staining was performed for F-actin filaments. RESULTS HSP27 overexpression induced cellular resistance to heat shock at 45 degrees C for 1 h as well as against several cytotoxic agents, including cisplatin, staurosporin and H(2)O(2). However, no difference in sensitivity to irradiation or serum starvation was found. Moreover, HSP27 overexpressing Hep-2 cells showed a delayed cell growth, compared to control cells. To determine if the decreased cell proliferation in HSP27 overexpressing cells contributed to chemoresistance, control Hep-2 cells were synchronized at the late G1 phase by treatment with mimosine. The synchronized Hep-2 cells were resistant to cisplatin and H(2)O(2), but not to irradiation or serum starvation, correlating the protection effect shown in HSP27 overexpressing cells. These results suggest that the overexpression of HSP27 in Hep-2 cells confers chemoresistance which is associated with the delay in cell growth. We also propose that the stabilization of F-actin observed in Hep-2/hsp27 cells is partly related to the delay in cell cycle progression, by showing that the induction of actin polymerization in Hep-2/neo cells results in the retardation of cell growth as well as a cytoprotective effect as observed in Hep-2/hsp27.
Collapse
Affiliation(s)
- Jung-Hee Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Chen H, Huang XN, Yan W, Chen K, Guo L, Tummalapali L, Dedhar S, St-Arnaud R, Wu C, Sepulveda JL. Role of the integrin-linked kinase/PINCH1/alpha-parvin complex in cardiac myocyte hypertrophy. J Transl Med 2005; 85:1342-56. [PMID: 16170337 DOI: 10.1038/labinvest.3700345] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Outside-in signaling from fibronectin (FN) through integrin receptors has been shown to play an important role in promoting cardiac myocyte hypertrophy and synergizes with other hypertrophic stimuli such as the alpha-adrenergic agonist phenylephrine (PE) and mechanical strain. The integrin-linked kinase (ILK) is a critical molecule involved in cell adhesion, motility and survival in nonmyocytes such as fibroblasts and epithelial cells. Its role in cardiac myocytes is unclear. In this study, we demonstrate that (1) ILK forms a complex with PINCH1 and alpha-parvin proteins (IPAP1 complex) in neonatal rat ventricular myocytes; (2) localization of IPAP1 complex proteins to costameres in cardiac myocytes is stimulated by FN, PE and synergistically by the combination of FN and PE in an integrin beta1-dependent manner; (3) a dominant-negative mutant lacking the PINCH-binding N-terminus of ILK (ILK-C) prevents costamere association of ILK and alpha-parvin, but not PINCH1; (4) FN- and PE-induced hypertrophy, measured by increased protein/DNA ratio, beating frequency and atrial natriuretic peptide expression, is stimulated by low levels of ILK-C but repressed by high ILK-C expression; and (5) overexpression of ILK-C, as well as deletion of the ILK gene in mouse neonatal ventricular myocytes, induces marked apoptosis of cardiac myocytes. These results suggest that the IPAP1 complex plays an important role in mediating integrin-signaling pathways that regulate cardiac myocyte hypertrophy and resistance to apoptosis.
Collapse
Affiliation(s)
- Hua Chen
- Department of Pathology, New York University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kong JY, Rabkin SW. The association between RhoB and caspase-2: changes with lovastatin-induced apoptosis. Biochem Cell Biol 2005; 83:608-19. [PMID: 16234849 DOI: 10.1139/o05-066] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Because cytoskeletal actin is regulated, in part, by Rho, and because Rho and caspases are involved in apoptosis, we sought to determine whether there was an association between RhoB and caspase-2. A RhoB–caspase-2 association was consistently demonstrated in neonatal mouse cardiomyocytes with Western Blotting, either after im mun o precipitation with RhoB followed by immunoblotting with caspase-2, or in reciprocal experiments after immuno precipitation with caspase-2 and immunoblotting with RhoB (n = 14). Although the RhoB–caspase-2 complex was constitutively present, the link between RhoB and caspase-2 may be operative in apoptosis because the HMG-CoA reductase inhibitor lovastatin increased the RhoB–caspase complex, especially in the nuclear fraction of the cell, with a peak occurrence 2 h after treatment. This association was unaffected by the caspase-2 inhibitor zVDVAD. Lovastatin produced apoptosis that was accompanied by an activation of caspase-2, as demonstrated by its immunohistochemistry and by the fact that the caspase-2 inhibitor zVDVAD reduced lovastatin-induced apoptosis. Lovastatin induced dramatic changes in cell morphology and a reduction in F-actin. Immunoblotting for actin suggests that lovastatin does not induce a degradation of the actin molecule, but rather affects filamentous F-actin. Caspase-2 inhibition with zVDVAD reduced lovastatin-induced alteration in cytoskeletal F-actin. The Rho inhibitor, Clostridium difficile toxin B, blunted the ability of lovastatin to induce apoptosis. In summary, these data show a previously unrecognized association between RhoB and caspase-2 in the cytosolic and nuclear fractions, which has ramifications for processes regulated by RhoB and caspase-2, including apoptosis.Key words: actin, apoptosis, caspase-2, cardiomyocyte, heart, lovastatin.
Collapse
Affiliation(s)
- Jennifer Y Kong
- University of BritishColumbia, Room D410, 2733 Heather Street, Vancouver, BCV5Z 3J5, Canada
| | | |
Collapse
|
31
|
Torsoni AS, Marin TM, Velloso LA, Franchini KG. RhoA/ROCK signaling is critical to FAK activation by cyclic stretch in cardiac myocytes. Am J Physiol Heart Circ Physiol 2005; 289:H1488-96. [PMID: 15923313 DOI: 10.1152/ajpheart.00692.2004] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Focal adhesion kinase (FAK) has been shown to be activated in cardiac myocytes exposed to mechanical stress. However, details of how mechanical stimuli induce FAK activation are unknown. We investigated whether signaling events mediated by the RhoA/Rho-associated coiled coil-containing kinase (ROCK) pathway are involved in regulation of stretch-induced FAK phosphorylation at Tyr(397) in neonatal rat ventricular myocytes (NRVMs). Immunostaining showed that RhoA localized to regions of myofilaments alternated with phalloidin (actin) staining. The results of coimmunoprecipitation assays indicated that FAK and RhoA are associated in nonstretched NRVMs, but cyclic stretch significantly reduced the amount of RhoA recovered from anti-FAK immunoprecipitates. Cyclic stretch induced rapid and sustained (up to 2 h) increases in phosphorylation of FAK at Tyr(397) and ERK1/2 at Thr(202)/Tyr(204). Blockade of RhoA/ROCK signaling by pharmacological inhibitors of RhoA (Clostridium botulinum C3 exoenzyme) or ROCK (Y-27632, 10 micromol/l, 1 h) markedly attenuated stretch-induced FAK and ERK1/2 phosphorylation. Similar effects were observed in cells treated with the inhibitor of actin polymerization cytochalasin D. Transfection of NRVMs with RhoA antisense oligonucleotide attenuated stretch-induced FAK and ERK1/2 phosphorylation and expression of beta-myosin heavy chain mRNA. Similar results were seen in cells transfected with FAK antisense oligonucleotide. These findings demonstrate that RhoA/ROCK signaling plays a crucial role in stretch-induced FAK phosphorylation, presumably by coordinating upstream events operationally linked to the actin cytoskeleton.
Collapse
Affiliation(s)
- Adriana S Torsoni
- Department of Internal Medicine, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | | | | | | |
Collapse
|
32
|
Kuwahara K, Barrientos T, Pipes GCT, Li S, Olson EN. Muscle-specific signaling mechanism that links actin dynamics to serum response factor. Mol Cell Biol 2005; 25:3173-81. [PMID: 15798203 PMCID: PMC1069631 DOI: 10.1128/mcb.25.8.3173-3181.2005] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myocardin and the myocardin-related transcription factors (MRTFs) MRTF-A and MRTF-B are coactivators for serum response factor (SRF), which regulates genes involved in cell proliferation, migration, cytoskeletal dynamics, and myogenesis. MRTF-A has been shown to translocate to the nucleus and activate SRF in response to Rho signaling and actin polymerization. Previously, we described a muscle-specific actin-binding protein named striated muscle activator of Rho signaling (STARS) that also activates SRF through a Rho-dependent mechanism. Here we show that STARS activates SRF by inducing the nuclear translocation of MRTFs. The STARS-dependent nuclear import of MRTFs requires RhoA and actin polymerization, and the actin-binding domain of STARS is necessary and sufficient for this activity. A knockdown of endogenous STARS expression by using small interfering RNA significantly reduced SRF activity in differentiated C2C12 skeletal muscle cells and cardiac myocytes. The ability of STARS to promote the nuclear localization of MRTFs and SRF-mediated transcription provides a potential muscle-specific mechanism for linking changes in actin dynamics and sarcomere structure with striated muscle gene expression.
Collapse
MESH Headings
- Actins/metabolism
- Active Transport, Cell Nucleus/physiology
- Animals
- Cell Line
- Cell Nucleus/chemistry
- Cell Nucleus/metabolism
- DNA-Binding Proteins/analysis
- DNA-Binding Proteins/metabolism
- Gene Expression Regulation, Developmental
- Humans
- Mice
- Microfilament Proteins/antagonists & inhibitors
- Microfilament Proteins/metabolism
- Microfilament Proteins/physiology
- Muscle Development/genetics
- Muscle Development/physiology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Nuclear Proteins/metabolism
- Oncogene Proteins, Fusion/analysis
- Oncogene Proteins, Fusion/metabolism
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
- Serum Response Factor/metabolism
- Signal Transduction
- Trans-Activators/metabolism
- Transcription Factors/analysis
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/metabolism
- Transcription Factors/physiology
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Koichiro Kuwahara
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
33
|
Abstract
RhoA and Rho-kinase (ROCK) participate in a wide variety of cell signal functions such as cell growth, smooth and cardiac muscle contraction, cytoskeleton rearrangement, cell migration and proliferation. In vascular smooth muscle cells, RhoA and ROCK play an important role in Ca2+ sensitization and regulate vascular smooth muscle tone. In the heart, RhoA and ROCK mediate hypertrophic response leading to cardiac hypertrophy. Recent cellular and molecular biology studies using ROCK inhibitors such as Y-27632 and fasudil have indicated a pivotal role of the RhoA-ROCK cascade in many aspects of cardiovascular function such as cardiac hypertrophy and ventricular remodeling following myocardial infarction. Inhibition of the RhoA-ROCK signaling pathway may be a suitable target for a number of cardiovascular diseases including hypertension, atherosclerosis, diabetes and hypertrophic heart failure. This review focuses on the current understanding of the RhoA-ROCK signal pathway in heart diseases and discusses the use of ROCK inhibitors as therapeutic agents for heart diseases ranging from hypertensive cardiomyopathy to heart failure.
Collapse
Affiliation(s)
- Jun Ren
- Center for Cardiovascular Research and Alternative Medicine and Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY 82071, USA.
| | | |
Collapse
|
34
|
Komati H, Minasi A, Naro F, Lagarde M, Prigent AF, Adamo S, Némoz G. Phorbol ester-induced differentiation of L6 myogenic cells involves phospholipase D activation. FEBS Lett 2005; 577:409-14. [PMID: 15556619 DOI: 10.1016/j.febslet.2004.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2004] [Revised: 09/24/2004] [Accepted: 10/05/2004] [Indexed: 10/26/2022]
Abstract
TPA, a potent PKC activator, inhibits myogenic differentiation and activates phospholipase D (PLD). We evaluated the involvement of PLD in the TPA effects on L6 myoblasts differentiation. TPA, at concentrations inhibiting differentiation of L6 cells, induced a strong, though transient, PLD activation. Surprisingly, at nanomolar concentration, TPA induced both myogenic differentiation and sustained activation of PLD. Differential effect of TPA can be ascribed to PKC downregulation induced by highest TPA concentrations. TPA-induced differentiation was inhibited by 1-butanol, confirming the involvement of PLD in this effect. These data suggest that prolonged elevation of PLD activity is required for myogenic differentiation.
Collapse
Affiliation(s)
- Hiba Komati
- Laboratoire de Physiopathologie des Lipides et Membranes, Institut National de la Santé et de la Recherche Médicale Unité 585, Institut National des Sciences Appliquées de Lyon, 69621 Villeurbanne, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Li S, Czubryt MP, McAnally J, Bassel-Duby R, Richardson JA, Wiebel FF, Nordheim A, Olson EN. Requirement for serum response factor for skeletal muscle growth and maturation revealed by tissue-specific gene deletion in mice. Proc Natl Acad Sci U S A 2005; 102:1082-7. [PMID: 15647354 PMCID: PMC545866 DOI: 10.1073/pnas.0409103102] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Serum response factor (SRF) controls the transcription of muscle genes by recruiting a variety of partner proteins, including members of the myocardin family of transcriptional coactivators. Mice lacking SRF fail to form mesoderm and die before gastrulation, precluding an analysis of the roles of SRF in muscle tissues. To investigate the functions of SRF in skeletal muscle development, we conditionally deleted the Srf gene in mice by skeletal muscle-specific expression of Cre recombinase. In mice lacking skeletal muscle SRF expression, muscle fibers formed, but failed to undergo hypertrophic growth after birth. Consequently, mutant mice died during the perinatal period from severe skeletal muscle hypoplasia. The myopathic phenotype of these mutant mice resembled that of mice expressing a dominant negative mutant of a myocardin family member in skeletal muscle. These findings reveal an essential role for the partnership of SRF and myocardin-related transcription factors in the control of skeletal muscle growth and maturation in vivo.
Collapse
Affiliation(s)
- Shijie Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9148, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Cell-extracellular matrix (ECM) adhesion is crucial for control of cell behavior. It connects the ECM to the intracellular cytoskeleton and transduces bidirectional signals between the extracellular and intracellular compartments. The subcellular machinery that mediates cell-ECM adhesion and signaling is complex. It consists of transmembrane proteins (e.g., integrins) and at least several dozens of membrane-proximal proteins that assemble into a network through multiple protein interactions. Furthermore, despite sharing certain common components, cell-ECM adhesions exhibit considerable heterogeneity in different types of cells (e.g., the cell-ECM adhesions in cardiac myocytes are considerably different from those in fibroblasts). Here, we will first briefly describe the general properties of the integrin-mediated cell-ECM adhesion and signal transduction. Next, we will focus on one of the recently discovered cell-ECM adhesion protein complexes consisting of PINCH, integrin-linked kinase (ILK), and Parvin and use it as an example to illustrate the molecular basis underlying the assembly and functions of cell-ECM adhesions. Finally, we will discuss in detail the structure and regulation of cell-ECM adhesion complexes in cardiac myocytes, which illustrate the importance and complexity of the cell-ECM adhesion structures in organogenesis and diseases.
Collapse
Affiliation(s)
- Jorge L Sepulveda
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | |
Collapse
|
37
|
Yatani A, Irie K, Otani T, Abdellatif M, Wei L. RhoA GTPase regulates L-type Ca2+ currents in cardiac myocytes. Am J Physiol Heart Circ Physiol 2004; 288:H650-9. [PMID: 15471984 DOI: 10.1152/ajpheart.00268.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Regulation of ionic channels plays a pivotal role in controlling cardiac function. Here we show that the Rho family of small G proteins regulates L-type Ca2+ currents in ventricular cardiomyocytes. Ventricular myocytes isolated from transgenic (TG) mice that overexpress the specific GDP dissociation inhibitor Rho GDI-alpha exhibited significantly decreased basal L-type Ca2+ current density (approximately 40%) compared with myocytes from nontransgenic (NTG) mice. The Ca2+ channel agonist BAY K 8644 and the beta-adrenergic agonist isoproterenol increased Ca2+ currents in both NTG and TG myocytes to a similar maximal level, and no changes in mRNA or protein levels were observed in the Ca2+ channel alpha1-subunits. These results suggest that the channel activity but not the expression level was altered in TG myocytes. In addition, the densities of inward rectifier and transient outward K+ currents were unchanged in TG myocytes. The amplitudes and rates of basal twitches and Ca2+ transients were also similar between the two groups. When the protein was delivered directly into adult ventricular myocytes via TAT-mediated protein transduction, Rho GDI-alpha significantly decreased Ca2+ current density, which supports the idea that the defective Ca2+ channel activity in TG myocytes was a primary effect of the transgene. In addition, expression of a dominant-negative RhoA but not a dominant-negative Rac-1 or Cdc42 also significantly decreased Ca2+ current density, which indicates that inhibition of Ca2+ channel activity by overexpression of Rho GDI-alpha is mediated by inhibition of RhoA. This study points to the L-type Ca2+ channel activity as a novel downstream target of the RhoA signaling pathway.
Collapse
Affiliation(s)
- Atsuko Yatani
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, USA
| | | | | | | | | |
Collapse
|
38
|
Chen H, Huang XN, Stewart AFR, Sepulveda JL. Gene expression changes associated with fibronectin-induced cardiac myocyte hypertrophy. Physiol Genomics 2004; 18:273-83. [PMID: 15306692 DOI: 10.1152/physiolgenomics.00104.2004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Fibronectin (FN) is an extracellular matrix protein that binds to integrin receptors and couples cardiac myocytes to the basal lamina. Cardiac FN expression is elevated in models of pressure overload, and FN causes cultured cardiac myocytes to hypertrophy by a mechanism that has not been characterized in detail. In this study, we analyzed the gene expression changes induced by FN in purified rat neonatal ventricular myocytes using the Affymetrix RAE230A microarray, to understand how FN affects gene expression in cardiac myocytes and to separate the effects contributed by cardiac nonmyocytes in vivo. Pathway analysis using z-score statistics and comparison with a mouse model of cardiac hypertrophy revealed several pathways stimulated by FN in cardiac myocytes. In addition to the known cardiac myocyte hypertrophy markers, FN significantly induced metabolic pathways including virtually all of the enzymes of cholesterol biosynthesis, fatty acid biosynthesis, and the mitochondrial electron transport chain. FN also increased the expression of genes coding for ribosomal proteins, translation factors, and the ubiquitin-proteasome pathway. Interestingly, cardiac myocytes plated on FN showed elevated expression of the fibrosis-promoting peptides connective tissue growth factor (CTGF), WNT1 inducible signaling pathway protein 2 (WISP2), and secreted acidic cysteine-rich glycoprotein (SPARC). Our data complement in vivo studies and reveal several novel genes and pathways stimulated by FN, pointing to cardiac myocyte-specific mechanisms that lead to development of the hypertrophic phenotype.
Collapse
Affiliation(s)
- Hua Chen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
39
|
Curtis C, Hemmeryckx B, Haataja L, Senadheera D, Groffen J, Heisterkamp N. Scambio, a novel guanine nucleotide exchange factor for Rho. Mol Cancer 2004; 3:10. [PMID: 15107133 PMCID: PMC420252 DOI: 10.1186/1476-4598-3-10] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2003] [Accepted: 04/23/2004] [Indexed: 12/17/2022] Open
Abstract
Background Small GTPases of the Rho family are critical regulators of various cellular functions including actin cytoskeleton organization, activation of kinase cascades and mitogenesis. For this reason, a major objective has been to understand the mechanisms of Rho GTPase regulation. Here, we examine the function of a novel protein, Scambio, which shares homology with the DH-PH domains of several known guanine nucleotide exchange factors for Rho family members. Results Scambio is located on human chromosome 14q11.1, encodes a protein of around 181 kDa, and is highly expressed in both heart and skeletal muscle. In contrast to most DH-PH-domain containing proteins, it binds the activated, GTP-bound forms of Rac and Cdc42. However, it fails to associate with V14RhoA. Immunofluorescence studies indicate that Scambio and activated Rac3 colocalize in membrane ruffles at the cell periphery. In accordance with these findings, Scambio does not activate either Rac or Cdc42 but rather, stimulates guanine nucleotide exchange on RhoA and its close relative, RhoC. Conclusion Scambio associates with Rac in its activated conformation and functions as a guanine nucleotide exchange factor for Rho.
Collapse
Affiliation(s)
- Christina Curtis
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
- Current address: Molecular and Computational Biology Department, University of Southern California, Los Angeles, CA 90089, USA
| | - Bianca Hemmeryckx
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
| | - Leena Haataja
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
- Current address: Larry Hillblom Islet Research Center, UCLA Division of Endocrinology, Los Angeles, CA 90095-7073, USA
| | - Dinithi Senadheera
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
| | - John Groffen
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
| | - Nora Heisterkamp
- Division of Hematology/Oncology, Section of Molecular Carcinogenesis, Childrens Hospital Los Angeles Research Institute and the Keck School of Medicine of the University of Southern California, Los Angeles, California 90027, USA
| |
Collapse
|
40
|
McClung JM, Thompson RW, Lowe LL, Carson JA. RhoA expression during recovery from skeletal muscle disuse. J Appl Physiol (1985) 2004; 96:1341-8. [PMID: 15016791 DOI: 10.1152/japplphysiol.01015.2003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Functional overload and anabolic steroid administration induce signaling pathways that regulate skeletal muscle RhoA expression. The purpose of this study was to determine RhoA and associated protein expression at the onset of disuse and after a brief period of reloading. Male Sprague-Dawley rats were randomly assigned to cage control (Con), 3 days of hindlimb suspension (Sus), or 3 days of hindlimb suspension with 12 h of reloading (12-h Reload). The reloading stimuli consisted of 12 h of resumed normal locomotion after 3 days of hindlimb suspension. Plantaris muscle-to-body weight (mg/g) ratio decreased 17% from Con with Sus but returned to Con with 12-h Reload, increasing 13% from Sus. Sus decreased RhoA protein concentration 46%, whereas 12-h Reload induced a 24% increase compared with Sus. The ratio of cytosolic- to membrane-associated RhoA protein was not changed with either Sus or 12-h Reload. RhoA mRNA concentration was decreased 48% by Sus, and 12-h Reload induced a 170% increase from Sus. β1-Integrin protein, a transmembrane protein associated with RhoA activation, was not altered by Sus but increased 155% with 12-h Reload. Although β1-integrin mRNA was not altered by Sus, it increased 70% from Con with 12-h Reload. Rho family member Cdc42 protein associated with the muscle membrane was decreased 60% with Sus, and 12-h Reload induced a 172% increase compared with Sus. In conclusion, decreased RhoA protein expression and mRNA abundance are early adaptations to disuse but recover rapidly after normal locomotion is resumed.
Collapse
Affiliation(s)
- J M McClung
- Department of Exercise Science, School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | | | | | | |
Collapse
|
41
|
Abstract
This review is divided into two parts, the first dealing with the cell and molecular biology of muscle in terms of growth and wasting and the second being an account of current knowledge of physiological mechanisms involved in the alteration of size of the human muscle mass. Wherever possible, attempts have been made to interrelate the information in each part and to provide the most likely explanation for phenomena that are currently only partially understood. The review should be of interest to cell and molecular biologists who know little of human muscle physiology and to physicians, physiotherapists, and kinesiologists who may be familiar with the gross behavior of human muscle but wish to understand more about the underlying mechanisms of change.
Collapse
Affiliation(s)
- Michael J Rennie
- Division of Molecular Physiology, School of Life Sciences, University of Dundee, Dundee, DD1 4HN, Scotland, United Kingdom.
| | | | | | | |
Collapse
|
42
|
Wittwer M, Billeter R, Hoppeler H, Flück M. Regulatory gene expression in skeletal muscle of highly endurance-trained humans. ACTA ACUST UNITED AC 2004; 180:217-27. [PMID: 14738480 DOI: 10.1046/j.0001-6772.2003.01242.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIM AND BACKGROUND Changes in regulatory and structural gene expression provide the molecular basis for the adaptation of human skeletal muscle to endurance exercise. HYPOTHESIS The steady-state levels of multiple mRNAs mainly involved in regulatory functions differ between highly endurance-trained and untrained subjects in a muscle heavily recruited during the exercise. METHODS Biopsies from musculus vastus lateralis of seven untrained (UT) subjects [maximal oxygen consumption (VO2max) = 39 mL kg-1 min-1] and seven trained (T) professional cyclists (VO2max = 72 mL kg-1 min-1) were analysed for the contents of 597 different mRNAs using commercially available cDNA arrays (Clontech no. 7740-1). Intra-individual expression profiles were compared by least-square linear regression analysis. Differences in gene expression between the two groups were tested for statistical significance using L1 regression analysis combined with the sign test on all permutations of scatter plots of log raw values from UT vs. T subjects. RESULTS Transcripts for 144 of 597 genes were sufficiently abundant to be analysed quantitatively. The expression profiles of the T group had a better intragroup correlation (R2) than those of the UT group (0.78 vs. 0.65, P < 0.05). An intergroup (T vs. UT) correlation of expression profiles gave an R2 of 0.71. Statistical analysis at a false discovery rate of 5% identified differential expression of nine cell-regulatory genes between T and UT. The mRNA levels of eight genes, including two DNA repair enzymes, transcription factors, signal transducers, a glycolytic enzyme and a factor involved in steroid hormone metabolism were increased in T vs. UT. Conversely, the mRNA of the tumour suppressor APC was downregulated with endurance training. Selective reverse-transcriptase polymerase chain reaction experiments confirmed the signal estimates from the array analysis. CONCLUSIONS The repetitive impact of the complex exercise stimuli in professional cyclists attenuated the interindividual differences in regulatory gene expression in skeletal muscle. Long-term nuclear reprogramming of regulatory gene expression seems to be characteristic of human musculus vastus lateralis in a highly endurance-trained steady state.
Collapse
Affiliation(s)
- M Wittwer
- Department of Anatomy, University of Bern, Bern, Switzerland
| | | | | | | |
Collapse
|
43
|
Ott C, Iwanciw D, Graness A, Giehl K, Goppelt-Struebe M. Modulation of the expression of connective tissue growth factor by alterations of the cytoskeleton. J Biol Chem 2003; 278:44305-11. [PMID: 12951326 DOI: 10.1074/jbc.m309140200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Modulation of the cytoskeletal architecture was shown to regulate the expression of CTGF (connective tissue growth factor, CCN2). The microtubule disrupting agents nocodazole and colchicine strongly up-regulated CTGF expression, which was prevented upon stabilization of the microtubules by paclitaxel. As a consequence of microtubule disruption, RhoA was activated and the actin stress fibers were stabilized. Both effects were related to CTGF induction. Overexpression of constitutively active RhoA induced CTGF synthesis. Interference with RhoA signaling by simvastatin, toxinB, C3 toxin, and Y27632 prevented up-regulation of CTGF. Likewise, direct disintegration of the actin cytoskeleton by latrunculin B interfered with nocodazole-mediated up-regulation of CTGF expression. Disassembly of actin fibers by cytochalasin D, however, unexpectedly increased CTGF expression indicating that the content of F-actin per se was not the major determinant for CTGF gene expression. Given the fact that cytochalasin D sequesters G-actin, a decrease in G-actin increased CTGF, while increased levels of G-actin corresponded to reduced CTGF expression. These data link alterations in the microtubule and actin cytoskeleton to the expression of CTGF and provide a molecular basis for the observation that CTGF is up-regulated in cells exposed to mechanical stress.
Collapse
Affiliation(s)
- Christian Ott
- Medizinische Klinik IV, Universität Erlangen-Nürnberg, Loschgestrasse 8, D-91054 Erlangen, Germany
| | | | | | | | | |
Collapse
|
44
|
Chang J, Wei L, Otani T, Youker KA, Entman ML, Schwartz RJ. Inhibitory cardiac transcription factor, SRF-N, is generated by caspase 3 cleavage in human heart failure and attenuated by ventricular unloading. Circulation 2003; 108:407-13. [PMID: 12874181 DOI: 10.1161/01.cir.0000084502.02147.83] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Knowledge about molecular mechanisms leading to heart failure is still limited, but reduced gene activities and modest activation of caspase 3 are hallmarks of end-stage heart failure. We postulated that serum response factor (SRF), a central cardiac transcription factor, might be a cleavage target for modest activated caspase 3, and this cleavage of SRF may play a dominant inhibitory role in propelling hearts toward failure. METHODS AND RESULTS We examined SRF protein levels from cardiac samples taken at the time of transplantation in 13 patients with end-stage heart failure and 7 normal hearts. Full-length SRF was markedly reduced and processed into 55- and 32-kDa subfragments in all failing hearts. SRF was intact in normal samples. In contrast, the hearts of 10 patients with left ventricular assist devices showed minimal SRF fragmentation. Specific antibodies to N- and C-terminal SRF sequences and site-directed mutagenesis revealed 2 alternative caspase 3 cleavage sites, so that 2 fragments were detected of each containing either the N- or C-terminal SRF. Expression of SRF-N, the 32-kDa fragment, in myogenic cells inhibited the transcriptional activity of alpha-actin gene promoters by 50% to 60%, which suggests that truncated SRF functioned as a dominant-negative transcription factor. CONCLUSIONS Caspase 3 activation in heart failure sequentially cleaved SRF and generated a dominant-negative transcription factor, which may explain the depression of cardiac-specific genes. Moreover, caspase 3 activation may be reversible in the failing heart with ventricular unloading.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Molecular and Cellular Biology, Center for Cardiovascular Development, Methodist Hospital, Baylor College of Medicine, One Baylor Plaza, Houston, Tex 77030, USA
| | | | | | | | | | | |
Collapse
|
45
|
Carson JA, Culberson DE, Thompson RW, Fillmore RA, Zimmer W. Smooth muscle gamma-actin promoter regulation by RhoA and serum response factor signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1628:133-9. [PMID: 12890560 DOI: 10.1016/s0167-4781(03)00122-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Smooth muscle gamma-actin (SMGA) is both an early marker of smooth muscle cell differentiation, which demonstrates an expression pattern restricted to smooth muscle and the post meiotic spermatocyte. Serum response factor (SRF) DNA-binding is an important regulator of muscle differentiation, including SMGA expression during smooth muscle cell differentiation. RhoA, a low molecular weight GTPase protein, can regulate cardiac, skeletal, and smooth muscle differentiation through SRF-dependent mechanisms. This study's purpose was to examine RhoA expression during smooth muscle cell development, and determine if the SMGA promoter activity is sensitive to RhoA-mediated signaling through SRF. Additionally, the study identified the promoter regulation modifying SMGA expression by RhoA signaling. Western blot analysis of embryonic chick gizzard whole protein extracts during 5 to 14 days of development demonstrated a large induction of RhoA (10-fold) and beta1 integrin expression at day 8, which corresponds to the time SMGA expression and differentiation are occurring. Transient transfections in CV-1 fibroblast cells demonstrated that co-overexpression of SRF and RhoA could induce a 40-fold induction of -176 bp SMGA promoter activity. Mutational analysis demonstrated that serum response element (SRE)-1, but not SRE2, was necessary for RhoA/SRF activation of the SMGA promoter. Deletion analysis revealed that although SRE1 was necessary for SMGA promoter activation by RhoA and SRF, it was not sufficient, implicating a possible obligatory role of additional promoter sequences in the response. Overexpression of a mutated SRF protein that was unable to bind DNA demonstrated that the 40-fold RhoA/SRF activation was largely dependent on SRF binding to the SMGA promoter. Thus, as the SMGA promoter appears to be a target of RhoA-mediated transcriptional regulation, the uncovering of these signaling mechanisms effecting SMGA promoter activity should provide a regulatory paradigm that can then be examined during the regulation of other smooth muscle genes.
Collapse
Affiliation(s)
- James A Carson
- Integrative Muscle Biology Laboratory, Exercise Science Department, University of South Carolina, 1300 Wheat St., Columbia, SC 29208, USA.
| | | | | | | | | |
Collapse
|
46
|
Witteck A, Yao Y, Fechir M, Förstermann U, Kleinert H. Rho protein-mediated changes in the structure of the actin cytoskeleton regulate human inducible NO synthase gene expression. Exp Cell Res 2003; 287:106-15. [PMID: 12799187 DOI: 10.1016/s0014-4827(03)00129-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Rho proteins (Rho, Rac, Cdc 42) are known to control the organization of the actin cytoskeleton as well as gene expression. Inhibition of Rho proteins by Clostridium difficile toxin B disrupted the F-actin cytoskeleton and enhanced cytokine-induced inducible nitric oxide synthase (iNOS) expression in human epithelial cells. Also specific inhibition by Y-27632 of p160ROCK, which mediates Rho effects on actin fibers, caused a disruption of the actin cytoskeleton and a superinduction of cytokine-induced iNOS expression. Accordingly, direct disruption of the actin cytoskeleton by cytochalasin D, latrunculin B, or jasplakinolide enhanced cytokine-induced iNOS expression. The transcription factor serum response factor (SRF) has been described as mediating actin cytoskeleton-dependent regulation of gene expression. Direct targets of SRF are activating protein 1 (AP1)-dependent genes. All compounds used inhibited SRF- and AP1-dependent reporter gene expression in DLD-1 cells. However, the enhancing effect of the actin cytoskeleton-disrupting compounds on human iNOS promoter activity was much less pronounced than the effect on iNOS mRNA expression. Therefore, besides transcriptional mechanisms, posttranscriptional effects seem to be involved in the regulation of iNOS expression by the above compounds. In conclusion, our data suggest that Rho protein-mediated changes of the actin cytoskeleton negatively modulate the expression of human iNOS.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/metabolism
- Bacterial Proteins
- Bacterial Toxins/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cytochalasin D/pharmacology
- Cytokines/metabolism
- Cytokines/pharmacology
- Cytoskeleton/drug effects
- Cytoskeleton/metabolism
- Depsipeptides
- Enzyme Inhibitors/pharmacology
- Eukaryotic Cells/enzymology
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Humans
- Intracellular Signaling Peptides and Proteins
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/drug effects
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- Peptides, Cyclic/pharmacology
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Serum Response Factor/drug effects
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Thiazoles/pharmacology
- Thiazolidines
- Transcription Factor AP-1/drug effects
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/metabolism
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
- Tumor Cells, Cultured
- rho GTP-Binding Proteins/drug effects
- rho GTP-Binding Proteins/metabolism
- rho-Associated Kinases
Collapse
Affiliation(s)
- Andrea Witteck
- Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, D-55101, Mainz, Germany
| | | | | | | | | |
Collapse
|
47
|
Torsoni AS, Fonseca PM, Crosara-Alberto DP, Franchini KG. Early activation of p160ROCK by pressure overload in rat heart. Am J Physiol Cell Physiol 2003; 284:C1411-9. [PMID: 12570982 DOI: 10.1152/ajpcell.00098.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We investigated the effects of acute pressure overload on activation of p160(ROCK) in rat myocardium. Constriction of transverse aorta, controlled to increase peak systolic pressure of ascending aorta by approximately 40 mmHg, induced a rapid association of RhoA with Dbl-3 and p160(ROCK). The binding of p160(ROCK) to RhoA was rapidly increased, peaking at 30 min (approximately 3.5-fold), but reduced to lower levels (approximately 1.9-fold) by 60 min of pressure overload. The activity of immunoprecipitated p160(ROCK) toward myosin light chain increased approximately 2.5-fold within 10 min but decreased to lower levels (approximately 1.6-fold) after 60 min of pressure overload. Confocal microscopic analysis indicated that pressure overload induced the formation of aggregates of p160(ROCK) and RhoA along the longitudinal axis of cardiac myocytes. Immunoelectron microscopic analysis showed that pressure overload induced the association of p160(ROCK) and RhoA to Z-line, T-tubule, and subsarcolemmal areas. The rapid activation of p160(ROCK) by pressure overload and its aggregation in subcellular structures involved in transmission of mechanical force suggest a role for this enzyme in the mechanobiochemical transduction in the myocardium.
Collapse
Affiliation(s)
- Adriana S Torsoni
- Department of Internal Medicine, School of Medicine, State University of Campinas, 13081-970 Campinas, SP, Brazil
| | | | | | | |
Collapse
|
48
|
Karnik SK, Brooke BS, Bayes-Genis A, Sorensen L, Wythe JD, Schwartz RS, Keating MT, Li DY. A critical role for elastin signaling in vascular morphogenesis and disease. Development 2003; 130:411-23. [PMID: 12466207 DOI: 10.1242/dev.00223] [Citation(s) in RCA: 327] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Vascular proliferative diseases such as atherosclerosis and coronary restenosis are leading causes of morbidity and mortality in developed nations. Common features associated with these heterogeneous disorders involve phenotypic modulation and subsequent abnormal proliferation and migration of vascular smooth muscle cells into the arterial lumen, leading to neointimal formation and vascular stenosis. This fibrocellular response has largely been attributed to the release of multiple cytokines and growth factors by inflammatory cells. Previously, we demonstrated that the disruption of the elastin matrix leads to defective arterial morphogenesis. Here, we propose that elastin is a potent autocrine regulator of vascular smooth muscle cell activity and that this regulation is important for preventing fibrocellular pathology. Using vascular smooth muscle cells from mice lacking elastin (Eln(-/-)), we show that elastin induces actin stress fiber organization, inhibits proliferation, regulates migration and signals via a non-integrin, heterotrimeric G-protein-coupled pathway. In a porcine coronary model of restenosis, the therapeutic delivery of exogenous elastin to injured vessels in vivo significantly reduces neointimal formation. These findings indicate that elastin stabilizes the arterial structure by inducing a quiescent contractile state in vascular smooth muscle cells. Together, this work demonstrates that signaling pathways crucial for arterial morphogenesis can play an important role in the pathogenesis and treatment of vascular disease.
Collapse
Affiliation(s)
- Satyajit K Karnik
- Program in Human Molecular Biology and Genetics, University of Utah, Salt Lake City, UT, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Arai A, Spencer JA, Olson EN. STARS, a striated muscle activator of Rho signaling and serum response factor-dependent transcription. J Biol Chem 2002; 277:24453-9. [PMID: 11983702 DOI: 10.1074/jbc.m202216200] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Changes in actin dynamics influence diverse cellular processes and couple the actin-based cytoskeleton to changes in gene transcription. Members of the Rho GTPase family regulate cytoskeletal organization by stimulating actin polymerization and stress fiber formation when activated by extracellular signaling. The transcriptional activity of serum response factor (SRF) is stimulated in response to changes in actin dynamics and Rho signaling, but the proteins that mediate this phenomenon have not been fully identified. We describe a novel, evolutionarily conserved actin-binding protein, called STARS (striated muscle activator of Rho signaling), that is expressed specifically in cardiac and skeletal muscle cells. STARS binds to the I-band of the sarcomere and to actin filaments in transfected cells, where it activates Rho-signaling events. STARS stimulates the transcriptional activity of SRF through a mechanism that requires actin binding and involves Rho GTPase activation. STARS provides a potential mechanism for specifically enhancing Rho-dependent transcription in muscle cells and for linking changes in actin dynamics to gene transcription.
Collapse
Affiliation(s)
- Akiko Arai
- Department of Molecular Biology, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas 75390-9148, USA
| | | | | |
Collapse
|
50
|
Spangenburg EE, Booth FW. Multiple signaling pathways mediate LIF-induced skeletal muscle satellite cell proliferation. Am J Physiol Cell Physiol 2002; 283:C204-11. [PMID: 12055089 DOI: 10.1152/ajpcell.00574.2001] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There are many known growth factors/cytokines that induce skeletal muscle satellite cell proliferation. Currently, the signaling mechanisms in which these growth factors/cytokines activate satellite cell proliferation are not completely understood. Here, we sought to determine signaling mechanisms by which leukemia inhibitory factor (LIF) induces satellite cell proliferation in culture. First, we confirmed that LIF induces proliferation of C2C12 immortalized myoblasts and cultured primary rat satellite cells. In addition, we also found that this increase in proliferation can be inhibited by incubation of the cells in tyrphostin AG 490, a specific inhibitor of Janus-activated kinase (JAK) 2 activity. Furthermore, we also found that incubation of the cells at various time points with LIF (10 ng/ml) induces a significant, transient increase in JAK2 phosphorylation, signal transducers and activators of transcription (STAT3) phosphorylation, and STAT3 transcriptional activity. Increases in the STAT3-sensitive endogenous SOC3 protein followed these transient increases in STAT3 activation. In addition, AG 490 inhibited the increase in STAT3 phosphorylation. Finally, LIF did not change the phosphorylation status of extracellular signal-regulated protein kinase (ERK)1/2 or affect the phosphorylation status of Akt/protein kinase B. However, LY-294002, an inhibitor of phosphoinositide 3-kinase, blocked LIF-induced proliferation of satellite cells. These data suggest that LIF induces satellite cell proliferation by activation of the JAK2-STAT3 signaling pathway, suggesting that this may be an important pathway in muscle growth and/or hypertrophy.
Collapse
Affiliation(s)
- Espen E Spangenburg
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri-Columbia, Columbia, Missouri 65211, USA
| | | |
Collapse
|