1
|
Douglas T, Zhang J, Wu Z, Abdallah K, McReynolds M, Gilbert WV, Iwai K, Peng J, Young LH, Crews CM. An atypical E3 ligase safeguards the ribosome during nutrient stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617692. [PMID: 39416039 PMCID: PMC11482868 DOI: 10.1101/2024.10.10.617692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Metabolic stress must be effectively mitigated for the survival of cells and organisms. Ribosomes have emerged as signaling hubs that sense metabolic perturbations and coordinate responses that either restore homeostasis or trigger cell death. As yet, the mechanisms governing these cell fate decisions are not well understood. Here, we report an unexpected role for the atypical E3 ligase HOIL-1 in safeguarding the ribosome. We find HOIL-1 mutations associated with cardiomyopathy broadly sensitize cells to nutrient and translational stress. These signals converge on the ribotoxic stress sentinel ZAKα. Mechanistically, mutant HOIL-1 excludes a ribosome quality control E3 ligase from its functional complex and remodels the ribosome ubiquitin landscape. This quality control failure renders glucose starvation ribotoxic, precipitating a ZAKα-ATF4-xCT-driven noncanonical cell death. We further show HOIL-1 loss exacerbates cardiac dysfunction under pressure overload. These data reveal an unrecognized ribosome signaling axis and a molecular circuit controlling cell fate during nutrient stress.
Collapse
|
2
|
Wu KJ, Chen Q, Leung CH, Sun N, Gao F, Chen Z. Recent discoveries of the role of histone modifications and related inhibitors in pathological cardiac hypertrophy. Drug Discov Today 2024; 29:103878. [PMID: 38211819 DOI: 10.1016/j.drudis.2024.103878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Pathological cardiac hypertrophy is a common response of the heart to various pathological stimuli. In recent years, various histone modifications, including acetylation, methylation, phosphorylation and ubiquitination, have been identified to have crucial roles in regulating chromatin remodeling and cardiac hypertrophy. Novel drugs targeting these epigenetic changes have emerged as potential treatments for pathological cardiac hypertrophy. In this review, we provide a comprehensive summary of the roles of histone modifications in regulating the development of pathological cardiac hypertrophy, and discuss potential therapeutic targets that could be utilized for its treatment.
Collapse
Affiliation(s)
- Ke-Jia Wu
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China
| | - Qi Chen
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macau; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Taipa 999078, Macau; MoE Frontiers Science Centre for Precision Oncology, University of Macau, Taipa 999078, Macau.
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China.
| | - Fei Gao
- Department of Cardiology, Beijing An Zhen Hospital, Capital Medical University, Chaoyang District, Beijing 100029, PR China.
| | - Zhaoyang Chen
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, Fujian 350001, PR China.
| |
Collapse
|
3
|
Baena-Montes JM, Kraśny MJ, O’Halloran M, Dunne E, Quinlan LR. In Vitro Models for Improved Therapeutic Interventions in Atrial Fibrillation. J Pers Med 2023; 13:1237. [PMID: 37623487 PMCID: PMC10455620 DOI: 10.3390/jpm13081237] [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: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Atrial fibrillation is the most common type of cardiac arrhythmias in humans, mostly caused by hyper excitation of specific areas in the atrium resulting in dyssynchronous atrial contractions, leading to severe consequences such as heart failure and stroke. Current therapeutics aim to target this condition through both pharmacological and non-pharmacological approaches. To test and validate any of these treatments, an appropriate preclinical model must be carefully chosen to refine and optimise the therapy features to correctly reverse this condition. A broad range of preclinical models have been developed over the years, with specific features and advantages to closely mimic the pathophysiology of atrial fibrillation. In this review, currently available models are described, from traditional animal models and in vitro cell cultures to state-of-the-art organoids and organs-on-a-chip. The advantages, applications and limitations of each model are discussed, providing the information to select the appropriate model for each research application.
Collapse
Affiliation(s)
- Jara M. Baena-Montes
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
| | - Marcin J. Kraśny
- Smart Sensors Lab, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Martin O’Halloran
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Electrical & Electronic Engineering, School of Engineering, University of Galway, H91 TK33 Galway, Ireland
| | - Eoghan Dunne
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R. Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
- CÚRAM SFI Centre for Research in Medical Devices, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
4
|
Watanabe M, Yano T, Sato T, Umetsu A, Higashide M, Furuhashi M, Ohguro H. mTOR Inhibitors Modulate the Physical Properties of 3D Spheroids Derived from H9c2 Cells. Int J Mol Sci 2023; 24:11459. [PMID: 37511214 PMCID: PMC10380298 DOI: 10.3390/ijms241411459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
To establish an appropriate in vitro model for the local environment of cardiomyocytes, three-dimensional (3D) spheroids derived from H9c2 cardiomyoblasts were prepared, and their morphological, biophysical phase contrast and biochemical characteristics were evaluated. The 3D H9c2 spheroids were successfully obtained, the sizes of the spheroids decreased, and they became stiffer during 3-4 days. In contrast to the cell multiplication that occurs in conventional 2D planar cell cultures, the 3D H9c2 spheroids developed into a more mature form without any cell multiplication being detected. qPCR analyses of the 3D H9c2 spheroids indicated that the production of collagen4 (COL4) and fibronectin (FN), connexin43 (CX43), β-catenin, N-cadherin, STAT3, and HIF1 molecules had increased and that the production of COL6 and α-smooth muscle actin (α-SMA) molecules had decreased as compared to 2D cultured cells. In addition, treatment with rapamycin (Rapa), an mTOR complex (mTORC) 1 inhibitor, and Torin 1, an mTORC1/2 inhibitor, resulted in significantly decreased cell densities of the 2D cultured H9c2 cells, but the size and stiffness of the H9c2 cells within the 3D spheroids were reduced with the gene expressions of several of the above several factors being reduced. The metabolic responses to mTOR modulators were also different between the 2D and 3D cultures. These results suggest that as unique aspects of the local environments of the 3D spheroids, the spontaneous expression of GJ-related molecules and hypoxia within the core may be associated with their maturation, suggesting that this may become a useful in vitro model that replicates the local environment of cardiomyocytes.
Collapse
Affiliation(s)
- Megumi Watanabe
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Araya Umetsu
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Megumi Higashide
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
| | - Hiroshi Ohguro
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| |
Collapse
|
5
|
Zhou Y, Yu H, Vind AC, Kong L, Liu Y, Song X, Tu Z, Yun C, Smaill JB, Zhang QW, Ding K, Bekker-Jensen S, Lu X. Rational Design of Covalent Kinase Inhibitors by an Integrated Computational Workflow (Kin-Cov). J Med Chem 2023. [PMID: 37220641 DOI: 10.1021/acs.jmedchem.3c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Covalent kinase inhibitors (CKIs) hold great promise for drug development. However, examples of computationally guided design of CKIs are still scarce. Here, we present an integrated computational workflow (Kin-Cov) for rational design of CKIs. The design of the first covalent leucine-zipper and sterile-α motif kinase (ZAK) inhibitor was presented as an example to showcase the power of computational workflow for CKI design. The two representative compounds, 7 and 8, inhibited ZAK kinase with half-maximal inhibitory concentration (IC50) values of 9.1 and 11.5 nM, respectively. Compound 8 displayed an excellent ZAK target specificity in Kinome profiling against 378 wild-type kinases. Structural biology and cell-based Western blot washout assays validated the irreversible binding characteristics of the compounds. Our study presents a rational approach for the design of CKIs based on the reactivity and accessibility of nucleophilic amino acid residues in a kinase. The workflow is generalizable and can be applied to facilitate CKI-based drug design.
Collapse
Affiliation(s)
- Yang Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Hang Yu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Anna Constance Vind
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Lulu Kong
- Department of Biochemistry and Biophysics, Institute of Systems Biomedicine and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yiling Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaojuan Song
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Zhengchao Tu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Caihong Yun
- Department of Biochemistry and Biophysics, Institute of Systems Biomedicine and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jeff B Smaill
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 92019, New Zealand
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| |
Collapse
|
6
|
Zhuang J, Cheng G, Huang J, Guo H, Lai Y, Wang J, Shan Z, Zheng S. Rosuvastatin exerts cardioprotective effect in lipopolysaccharide-mediated injury of cardiomyocytes in an MG53-dependent manner. BMC Cardiovasc Disord 2022; 22:69. [PMID: 35196979 PMCID: PMC8865731 DOI: 10.1186/s12872-022-02458-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 01/04/2022] [Indexed: 12/03/2022] Open
Abstract
Background Myocarditis is a cardiomyopathy associated with the inflammatory response. Rosuvastatin (RS) demonstrates cardioprotective effect in the clinical setting, although its cellular and molecular mechanisms in ameliorating myocarditis are largely unknown. MG53 (muscle-specific E3 ligase Mitsugumin 53), a newly identified striated muscle-specific protein, is involved in skeletal muscle membrane repair. We aimed to explore whether RS mediated the repair of cardiomyocytes in an MG53-dependent manner. Methods The RS-induced upregulation of MG53 was determined using RT-qPCR and western blotting. A lipopolysaccharide (LPS)-induced cell inflammatory model was constructed using rat cardiac muscle cell H9C2. Inflammatory injury was evaluated according to the alterations of cell viability, mitochondrial membrane potential, cell apoptosis, and expression of pro-inflammatory cytokines (interleukin-1β, interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1). Small interfering RNAs (siRNAs) were used to silence MG53. The cardioprotective effect of RS and the inhibition of this protection by MG53 silence were evaluated in the forementioned in vitro model. The underlying mechanism was finally investigated using western blotting to detected the expressions of apoptotic markers (Bcl-2, Bax, Cleaved caspase-9, Cleaved caspase-3), cell cycle regulatory factors (Cyclin A, Cyclin E1, Cyclin D1, CDK2), and components involved in NF-κB signaling pathway (p-IκBa, Iκba, p-p65, p65). Results RS ameliorated LPS-induced inflammatory injury. RS upregulated the expression of MG53. MG53 was crucial for the RS-mediated repair response in vitro. Ablation of MG53 inhibited the RS-mediated protective effect. Furthermore, RS and MG53 interact in multiple signaling pathways to modulate recovery. Conclusion RS exerts cardioprotective effect in an MG53-dependent manner. MG53 may serve as a novel drug target for myocarditis treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02458-3.
Collapse
Affiliation(s)
- Jiawei Zhuang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Gangyi Cheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jian Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongwei Guo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yiquan Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiamao Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhonggui Shan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China.
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Bourque K, Hawey C, Jones-Tabah J, Pétrin D, Martin RD, Ling Sun Y, Hébert TE. Measuring hypertrophy in neonatal rat primary cardiomyocytes and human iPSC-derived cardiomyocytes. Methods 2021; 203:447-464. [PMID: 34933120 DOI: 10.1016/j.ymeth.2021.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In the heart, left ventricular hypertrophy is initially an adaptive mechanism that increases wall thickness to preserve normal cardiac output and function in the face of coronary artery disease or hypertension. Cardiac hypertrophy develops in response to pressure and volume overload but can also be seen in inherited cardiomyopathies. As the wall thickens, it becomes stiffer impairing the distribution of oxygenated blood to the rest of the body. With complex cellular signalling and transcriptional networks involved in the establishment of the hypertrophic state, several model systems have been developed to better understand the molecular drivers of disease. Immortalized cardiomyocyte cell lines, primary rodent and larger animal models have all helped understand the pathological mechanisms underlying cardiac hypertrophy. Induced pluripotent stem cell-derived cardiomyocytes are also used and have the additional benefit of providing access to human samples with direct disease relevance as when generated from patients suffering from hypertrophic cardiomyopathies. Here, we briefly review in vitro and in vivo model systems that have been used to model hypertrophy and provide detailed methods to isolate primary neonatal rat cardiomyocytes as well as to generate cardiomyocytes from human iPSCs. We also describe how to model hypertrophy in a "dish" using gene expression analysis and immunofluorescence combined with automated high-content imaging.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Yi Ling Sun
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
8
|
Yang J, Shibu MA, Kong L, Luo J, BadrealamKhan F, Huang Y, Tu ZC, Yun CH, Huang CY, Ding K, Lu X. Design, Synthesis, and Structure-Activity Relationships of 1,2,3-Triazole Benzenesulfonamides as New Selective Leucine-Zipper and Sterile-α Motif Kinase (ZAK) Inhibitors. J Med Chem 2020; 63:2114-2130. [PMID: 31244114 DOI: 10.1021/acs.jmedchem.9b00664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
ZAK is a new promising target for discovery of drugs with activity against antihypertrophic cardiomyopathy (HCM). A series of 1,2,3-triazole benzenesulfonamides were designed and synthesized as selective ZAK inhibitors. One of these compounds, 6p binds tightly to ZAK protein (Kd = 8.0 nM) and potently suppresses the kinase function of ZAK with single-digit nM (IC50 = 4.0 nM) and exhibits excellent selectivity in a KINOMEscan screening platform against a panel of 403 wild-type kinases. This compound dose dependently blocks p38/GATA-4 and JNK/c-Jun signaling and demonstrates promising in vivo anti-HCM efficacy upon oral administration in a spontaneous hypertensive rat (SHR) model. Compound 6p may serve as a lead compound for new anti-HCM drug discovery.
Collapse
Affiliation(s)
- Jianzhang Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | | | - Lulu Kong
- Department of Biochemistry and Biophysics, Institute of Systems Biomedicine and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jinfeng Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Farheen BadrealamKhan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Yanhui Huang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Zheng-Chao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Cai-Hong Yun
- Department of Biochemistry and Biophysics, Institute of Systems Biomedicine and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
- College of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
9
|
Gao J, Shao K, Chen X, Li Z, Liu Z, Yu Z, Aung LHH, Wang Y, Li P. The involvement of post-translational modifications in cardiovascular pathologies: Focus on SUMOylation, neddylation, succinylation, and prenylation. J Mol Cell Cardiol 2020; 138:49-58. [DOI: 10.1016/j.yjmcc.2019.11.146] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
|
10
|
Jiang W, Ding L, Dai T, Guo J, Dai R, Chang Y. Studies of pharmacokinetics in beagle dogs and drug-drug interaction potential of a novel selective ZAK inhibitor 3h for hypertrophic cardiomyopathy treatment. J Pharm Biomed Anal 2019; 172:206-213. [PMID: 31060033 DOI: 10.1016/j.jpba.2019.04.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
Overexpression of leucine-zipper and sterile-α motif kinase (ZAK) in heart has been closely associated with the development of hypertrophic cardiomyopathy (HCM). N-(3-(1H-pyrazolo[3,4-b]pyridin-5-yl)ethynyl) benzene-sulfonamides, novel highly selective ZAK inhibitors, had exhibited reasonable orally therapeutic effects on HCM in spontaneous hypertensive rat models. In the present study, a rapid and sensitive HPLC-MS/MS method for determining ZAK inhibitor 3h in beagle dog plasma was developed and validated. Meanwhile, the pharmacokinetics in beagle dog and drug-drug interaction potential of 3h had been conducted. The pharmacokinetic results showed that the absolute oral bioavailability for 3h in beagle dogs was determined to be 61.9%, which was significantly higher than that in the previous determination in Spragur-Dawley rats (F = 20%). The Cytochrome P450 enzymes and P-glycoprotein mediated drug-drug interactions by 3h were also investigated using dog and human liver microsomes and Caco-2 cells. The results demonstrated that only CYP2C9 was obviously inhibited (IC50 = 1.66 μM). Besides, 3h could significantly decrease digoxin efflux ratio in Caco-2 experiments in a dose-dependent manner (IC50 = 13.3 μM). Considering 3h strongly suppressed the ZAK kinase activity with an IC50 of 3.3 nM, there are significantly differences between this IC50 value for ZAK inhibition and the present determinations of IC50 values. In general, the clinical drug-drug interaction potential for 3h could be well monitored during the treatment of HCM.
Collapse
Affiliation(s)
- Weifan Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lan Ding
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Tianming Dai
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiayin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Renke Dai
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China.
| | - Yu Chang
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
11
|
Ninjurin1 regulates striated muscle growth and differentiation. PLoS One 2019; 14:e0216987. [PMID: 31091274 PMCID: PMC6519837 DOI: 10.1371/journal.pone.0216987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pressure overload due to aortic valve stenosis leads to pathological cardiac hypertrophy and heart failure. Hypertrophy is accompanied by an increase in myocyte surface area, which requires a proportional increase in the number of cell-cell and cell-matrix contacts to withstand enhanced workload. In a proteomic analysis we identified nerve injury-induced protein 1 (Ninjurin1), a 16kDa transmembrane cell-surface protein involved in cell adhesion and nerve repair, to be increased in hypertrophic hearts from patients with aortic stenosis. We hypothesised that Ninjurin1 is involved in myocyte hypertrophy. We analyzed cardiac biopsies from aortic-stenosis patients and control patients undergoing elective heart surgery. We studied cardiac hypertrophy in mice after transverse aortic constriction and angiotensin II infusions, and performed mechanistic analyses in cultured myocytes. We assessed the physiological role of ninjurin1 in zebrafish during heart and skeletal muscle development. Ninjurin1 was increased in hearts of aortic stenosis patients, compared to controls, as well as in hearts from mice with cardiac hypertrophy. Besides the 16kDa Ninjurin1 (Ninjurin1-16) we detected a 24kDa variant of Ninjurin1 (Ninjurin1-24), which was predominantly expressed during myocyte hypertrophy. We disclosed that the higher molecular weight of Ninjurin1-24 was caused by N-glycosylation. Ninjurin1-16 was contained in the cytoplasm of myocytes where it colocalized with stress-fibers. In contrast, Ninjurin1-24 was localized at myocyte membranes. Gain and loss-of-function experiments showed that Ninjurin1-24 plays a role in myocyte hypertrophy and myogenic differentiation in vitro. Reduced levels of ninjurin1 impaired cardiac and skeletal muscle development in zebrafish. We conclude that Ninjurin1 contributes to myocyte growth and differentiation, and that these effects are mainly mediated by N-glycosylated Ninjurin1-24.
Collapse
|
12
|
Pai P, Shibu MA, Chang RL, Yang JJ, Su CC, Lai CH, Liao HE, Viswanadha VP, Kuo WW, Huang CY. ERβ targets ZAK and attenuates cellular hypertrophy via SUMO-1 modification in H9c2 cells. J Cell Biochem 2018; 119:7855-7864. [PMID: 29932238 DOI: 10.1002/jcb.27199] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 05/24/2018] [Indexed: 11/11/2022]
Abstract
Aberrant expression of leucine zipper- and sterile ɑ motif-containing kinase (ZAK) observed in pathological human myocardial tissue is associated with the progression and elevation of hypertrophy. Our previous reports have correlated high levels of estrogen (E2) and abundant estrogen receptor (ER) α with a low incidence of pathological cardiac-hypertrophy and heart failure in the premenopause female population. However, the effect of elevated ERβ expression is not well known yet. Therefore, in this study, we have analyzed the cardioprotective effects and mechanisms of E2 and/or ERβ against ZAK overexpression-induced cellular hypertrophy. We have used transient transfection to overexpress ERβ into the ZAK tet-on H9c2 cells that harbor the doxycycline-inducible ZAK plasmid. The results show that ZAK overexpression in H9c2 cells resulted in hypertrophic effects, which was correlated with the upregulation of p-JNK and p-p38 MAPKs and their downstream transcription factors c-Jun and GATA-4. However, ERβ and E2 with ERβ overexpressions totally suppressed the effects of ZAK overexpression and inhibited the levels of p-JNK, p-p38, c-Jun, and GATA-4 effectively. Our results further reveal that ERβ directly binds with ZAK under normal conditions; however, ZAK overexpression reduced the association of ZAK-ERβ. Interestingly, increase in ERβ and E2 along with ERβ overexpression both enhanced the binding strengths of ERβ and ZAK and reduced the ZAK protein level. ERβ overexpression also suppressed the E3 ligase-casitas B-lineage lymphoma (CBL) and attenuated CBL-phosphoinositide 3-kinase (PI3K) protein association to prevent PI3K protein degradation. Moreover, ERβ and/or E2 blocked ZAK nuclear translocation via the inhibition of small ubiquitin-like modifier (SUMO)-1 modification. Taken together, our results further suggest that ERβ overexpression strongly suppresses ZAK-induced cellular hypertrophy and myocardial damage.
Collapse
Affiliation(s)
- Peiying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | | | - Ruey-Lin Chang
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jaw-Ji Yang
- Institute of Medicine, School of Dentistry, Chung-Shan Medical University, Taichung, Taiwan
| | - Chia-Chi Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chao-Hung Lai
- Division of Cardiology, Department of Internal Medicine, Taichung Armed Force General Hospital, Taichung, Taiwan
| | - Hung-En Liao
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
13
|
Fu CY, Tseng YS, Chen MC, Hsu HH, Yang JJ, Tu CC, Lin YM, Viswanadha VP, Ding K, Kuo WW, Huang CY. Overexpression of ZAKβ in human osteosarcoma cells enhances ZAKα expression, resulting in a synergistic apoptotic effect. Cell Biochem Funct 2018; 36:176-182. [PMID: 29654619 DOI: 10.1002/cbf.3329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/21/2018] [Accepted: 02/13/2018] [Indexed: 11/07/2022]
Abstract
ZAK is a novel mixed lineage kinase-like protein that contains a leucine-zipper and a sterile-alpha motif as a protein-protein interaction domain, and it is located in the cytoplasm. There are 2 alternatively spliced forms of ZAK: ZAKα and ZAKβ. Previous studies showed that ZAKα is involved in various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy, but the molecular mechanism of ZAKβ is not yet known. In a recent study in our laboratory, we found that ZAKβ can ameliorate the apoptotic effect induced by ZAKα in H9c2 cells. We further hypothesized that ZAKβ could also improve the apoptotic effect induced by ZAKα in human osteosarcoma cells. The results of this study show that ZAKβ can induce apoptosis and decrease cell viability similar to the effects of ZAKα. Interestingly, our ZAKα-specific inhibitor assay shows that the expression of ZAKβ is highly dependent on ZAKα expression. However, ZAKβ expression effectively induces ZAKα expression and results in synergistic enhancement of apoptosis in human osteosarcoma cells. Furthermore, co-immunoprecipitation results revealed that ZAKα can directly interact with ZAKβ, and this interaction may contribute to the enhanced apoptotic effects. SIGNIFICANCE OF THE STUDY ZAK is a mixed lineage kinase involved in cell differentiation, proliferation, and hypertrophic growth. ZAKα isoform of ZAK is associated with tumorigenesis, but the function of ZAKβ is not yet known. In H9c2 cells, ZAKβ was found to ameliorate the apoptotic effect induced by ZAKα. However, in osteosarcoma cells, ZAKβ elevates the apoptotic effect induced by ZAKα. In this study, we show that similar to ZAKα, the ZAKβ induces apoptosis and decreases cell viability. Interestingly, the expression of ZAKβ is dependent on ZAKα expression, and ZAKβ further enhances ZAKα expression and results in synergistic enhancement of apoptosis in osteosarcoma cells.
Collapse
Affiliation(s)
- Chien-Yao Fu
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan.,Orthopaedic Department, Armed Forces General Hospital, Taichung, Taiwan.,Department of Orthopaedic, National Defense Medical Center, Taipei, Taiwan
| | - Yan-Shen Tseng
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan.,Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | - Jaw-Ji Yang
- School of Dentistry, Chung-Shan Medical University, Taichung, Taiwan
| | - Chuan-Chou Tu
- Division of Chest Medicine, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | | | - Ke Ding
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
14
|
Fu CY, Tseng YS, Chen MC, Hsu HH, Yang JJ, Tu CC, Lin YM, Viswanadha VP, Kuo WW, Huang CY. Doxorubicin induces ZAKα overexpression with a subsequent enhancement of apoptosis and attenuation of survivability in human osteosarcoma cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:191-197. [PMID: 29105997 DOI: 10.1002/tox.22507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/10/2017] [Accepted: 10/22/2017] [Indexed: 06/07/2023]
Abstract
Human osteosarcoma (OS) is a malignant cancer of the bone. It exhibits a characteristic malignant osteoblastic transformation and produces a diseased osteoid. A previous study demonstrated that doxorubicin (DOX) chemotherapy decreases human OS cell proliferation and might enhance the relative RNA expression of ZAK. However, the impact of ZAKα overexpression on the OS cell proliferation that is inhibited by DOX and the molecular mechanism underlying this effect are not yet known. ZAK is a protein kinase of the MAPKKK family and functions to promote apoptosis. In our study, we found that ZAKα overexpression induced an apoptotic effect in human OS cells. Treatment of human OS cells with DOX enhanced ZAKα expression and decreased cancer cell viability while increasing apoptosis of human OS cells. In the meantime, suppression of ZAKα expression using shRNA and inhibitor D1771 both suppressed the DOX therapeutic effect. These findings reveal a novel molecular mechanism underlying the DOX effect on human OS cells. Taken together, our findings demonstrate that ZAKα enhances the apoptotic effect and decreases cell viability in DOX-treated human OS cells.
Collapse
Affiliation(s)
- Chien-Yao Fu
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan
- Orthopaedic Department, Armed Forces General Hospital, Taichung, Taiwan
- Department of Orthopaedic, National Defense Medical Center, Taipei, Taiwan
| | - Yan-Shen Tseng
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, 10449, Taiwan
- Nursing and Management College, Mackay Medicine, Taipei, 11260, Taiwan
| | - Jaw-Ji Yang
- School of Dentistry, Chung-Shan Medical University, Taichung, 402, Taiwan
| | - Chuan-Chou Tu
- Division of Chest Medicine, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung, 41152, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, 500, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
15
|
Chang Y, Lu X, Shibu MA, Dai YB, Luo J, Zhang Y, Li Y, Zhao P, Zhang Z, Xu Y, Tu ZC, Zhang QW, Yun CH, Huang CY, Ding K. Structure Based Design of N-(3-((1H-Pyrazolo[3,4-b]pyridin-5-yl)ethynyl)benzenesulfonamides as Selective Leucine-Zipper and Sterile-α Motif Kinase (ZAK) Inhibitors. J Med Chem 2017; 60:5927-5932. [PMID: 28586211 DOI: 10.1021/acs.jmedchem.7b00572] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A series of N-(3-((1H-pyrazolo[3,4-b]pyridin-5-yl)ethynyl)benzenesulfonamides were designed as the first class of highly selective ZAK inhibitors. The representative compound 3h strongly inhibits the kinase activity of ZAK with an IC50 of 3.3 nM and dose-dependently suppresses the activation of ZAK downstream signals in vitro and in vivo, while it is significantly less potent for the majority of 403 nonmutated kinases evaluated. Compound 3h also exhibits orally therapeutic effects on cardiac hypertrophy in a spontaneous hypertensive rat model.
Collapse
Affiliation(s)
- Yu Chang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao, China
- School of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaoyun Lu
- School of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Marthandam Asokan Shibu
- Graduate Institute of Basic Medical Science, China Medical University , Taichung 404, Taiwan, China
- Department of Health and Nutrition Biotechnology, Asia University , Taichung 433, Taiwan, China
| | - Yi-Bo Dai
- Institute of Systems Biomedicine, Department of Biophysics and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Jinfeng Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yan Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yingjun Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Peng Zhao
- Institute of Systems Biomedicine, Department of Biophysics and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Zhang Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yong Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Zheng-Chao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao, China
| | - Cai-Hong Yun
- Institute of Systems Biomedicine, Department of Biophysics and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University , Taichung 404, Taiwan, China
- Department of Health and Nutrition Biotechnology, Asia University , Taichung 433, Taiwan, China
| | - Ke Ding
- School of Pharmacy, Jinan University , 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
16
|
Vasli N, Harris E, Karamchandani J, Bareke E, Majewski J, Romero NB, Stojkovic T, Barresi R, Tasfaout H, Charlton R, Malfatti E, Bohm J, Marini-Bettolo C, Choquet K, Dicaire MJ, Shao YH, Topf A, O'Ferrall E, Eymard B, Straub V, Blanco G, Lochmüller H, Brais B, Laporte J, Tétreault M. Recessive mutations in the kinase ZAK cause a congenital myopathy with fibre type disproportion. Brain 2016; 140:37-48. [PMID: 27816943 DOI: 10.1093/brain/aww257] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/03/2016] [Accepted: 08/31/2016] [Indexed: 01/09/2023] Open
Abstract
Congenital myopathies define a heterogeneous group of neuromuscular diseases with neonatal or childhood hypotonia and muscle weakness. The genetic cause is still unknown in many patients, precluding genetic counselling and better understanding of the physiopathology. To identify novel genetic causes of congenital myopathies, exome sequencing was performed in three consanguineous families. We identified two homozygous frameshift mutations and a homozygous nonsense mutation in the mitogen-activated protein triple kinase ZAK. In total, six affected patients carry these mutations. Reverse transcription polymerase chain reaction and transcriptome analyses suggested nonsense mRNA decay as a main impact of mutations. The patients demonstrated a generalized slowly progressive muscle weakness accompanied by decreased vital capacities. A combination of proximal contractures with distal joint hyperlaxity is a distinct feature in one family. The low endurance and compound muscle action potential amplitude were strongly ameliorated on treatment with anticholinesterase inhibitor in another patient. Common histopathological features encompassed fibre size variation, predominance of type 1 fibre and centralized nuclei. A peculiar subsarcolemmal accumulation of mitochondria pointing towards the centre of the fibre was a novel histological hallmark in one family. These findings will improve the molecular diagnosis of congenital myopathies and implicate the mitogen-activated protein kinase (MAPK) signalling as a novel pathway altered in these rare myopathies.
Collapse
Affiliation(s)
- Nasim Vasli
- 1 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404 Illkirch, France.,2 INSERM U974, 67404 Illkirch, France.,3 CNRS, UMR7104, 67404 Illkirch, France.,4 Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67404 Illkirch, France
| | - Elizabeth Harris
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Jason Karamchandani
- 6 Department of Pathology, McGill University Health Centre, Montreal Neurological Institute Hospital, Montreal, QC H3A 2B4, Canada
| | - Eric Bareke
- 7 Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada.,8 McGill University and Genome Quebec Innovation Center, Montreal, QC H3A 1A4, Canada
| | - Jacek Majewski
- 7 Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada.,8 McGill University and Genome Quebec Innovation Center, Montreal, QC H3A 1A4, Canada
| | - Norma B Romero
- 9 Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013 Paris, France.,10 Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Tanya Stojkovic
- 10 Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Rita Barresi
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Hichem Tasfaout
- 1 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404 Illkirch, France.,2 INSERM U974, 67404 Illkirch, France.,3 CNRS, UMR7104, 67404 Illkirch, France.,4 Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67404 Illkirch, France
| | - Richard Charlton
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Edoardo Malfatti
- 9 Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013 Paris, France.,10 Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Johann Bohm
- 1 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404 Illkirch, France.,2 INSERM U974, 67404 Illkirch, France.,3 CNRS, UMR7104, 67404 Illkirch, France.,4 Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67404 Illkirch, France
| | - Chiara Marini-Bettolo
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Karine Choquet
- 7 Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada.,11 Rare Neurological Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Marie-Josée Dicaire
- 11 Rare Neurological Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Yi-Hong Shao
- 11 Rare Neurological Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ana Topf
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Erin O'Ferrall
- 6 Department of Pathology, McGill University Health Centre, Montreal Neurological Institute Hospital, Montreal, QC H3A 2B4, Canada
| | - Bruno Eymard
- 10 Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Volker Straub
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Gonzalo Blanco
- 12 Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Hanns Lochmüller
- 5 John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Bernard Brais
- 7 Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada.,11 Rare Neurological Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jocelyn Laporte
- 1 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1, rue Laurent Fries, BP 10142, 67404 Illkirch, France .,2 INSERM U974, 67404 Illkirch, France.,3 CNRS, UMR7104, 67404 Illkirch, France.,4 Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67404 Illkirch, France
| | - Martine Tétreault
- 7 Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada .,8 McGill University and Genome Quebec Innovation Center, Montreal, QC H3A 1A4, Canada
| |
Collapse
|
17
|
Nyati S, Chator A, Schinske K, Gregg BS, Ross BD, Rehemtulla A. A Requirement for ZAK Kinase Activity in Canonical TGF-β Signaling. Transl Oncol 2016; 9:473-481. [PMID: 27783979 PMCID: PMC5080675 DOI: 10.1016/j.tranon.2016.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 11/15/2022] Open
Abstract
The sterile alpha motif and leucine zipper containing kinase ZAK (AZK, MLT, MLK7), is a MAPK-kinase kinase (MKKK). Like most MAPKKKs which are known to activate the c-Jun. amino-terminal kinase (JNK) pathway, ZAK has been shown to participate in the transduction of Transforming growth factor-β (TGF-β)-mediated non-canonical signaling. A role for ZAK in SMAD-dependent, canonical TGF-β signaling has not been previously appreciated. Using a combination of functional genomics and biochemical techniques, we demonstrate that ZAK regulates canonical TGFβRI/II signaling in lung and breast cancer cell lines and may serve as a key node in the regulation of TGFBR kinase activity. Remarkably, we demonstrate that siRNA mediated depletion of ZAK strongly inhibited TGF-β dependent SMAD2/3 activation and subsequent promoter activation (SMAD binding element driven luciferase expression; SBE4-Luc). A ZAK specific inhibitor (DHP-2), dose-dependently activated the bioluminescent TGFBR-kinase activity reporter (BTR), blocked TGF-β induced SMAD2/3 phosphorylation and SBE4-Luc activation and cancer cell-invasion. In aggregate, these findings identify a novel role for the ZAK kinase in canonical TGF-β signaling and an invasive cancer cell phenotype thus providing a novel target for TGF-β inhibition.
Collapse
Affiliation(s)
- Shyam Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48105, USA; Center for Molecular Imaging, University of Michigan, Ann Arbor, MI 48105, USA.
| | - Areeb Chator
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Katerina Schinske
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brandon S Gregg
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brian Dale Ross
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48105, USA; Center for Molecular Imaging, University of Michigan, Ann Arbor, MI 48105, USA; Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
18
|
Impaired ALDH2 activity decreases the mitochondrial respiration in H9C2 cardiomyocytes. Cell Signal 2015; 28:1-6. [PMID: 26577527 DOI: 10.1016/j.cellsig.2015.11.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/02/2015] [Accepted: 11/09/2015] [Indexed: 12/26/2022]
Abstract
Reactive oxygen species (ROS)-mediated reactive aldehydes induce cellular stress. In cardiovascular diseases such as ischemia-reperfusion injury, lipid-peroxidation derived reactive aldehydes such as 4-hydroxy-2-nonenal (4HNE) are known to contribute to the pathogenesis. 4HNE is involved in ROS formation, abnormal calcium handling and more importantly defective mitochondrial respiration. Aldehyde dehydrogenase (ALDH) superfamily contains NAD(P)(+)-dependent isozymes which can detoxify endogenous and exogenous aldehydes into non-toxic carboxylic acids. Therefore we hypothesize that 4HNE afflicts mitochondrial respiration and leads to cell death by impairing ALDH2 activity in cultured H9C2 cardiomyocyte cell lines. H9C2 cardiomyocytes were treated with 25, 50 and 75 μM 4HNE and its vehicle, ethanol as well as 25, 50 and 75 μM disulfiram (DSF), an inhibitor of ALDH2 and its vehicle (DMSO) for 4 h. 4HNE significantly decreased ALDH2 activity, ALDH2 protein levels, mitochondrial respiration and mitochondrial respiratory reserve capacity, and increased 4HNE adduct formation and cell death in cultured H9C2 cardiomyocytes. ALDH2 inhibition by DSF and ALDH2 siRNA attenuated ALDH2 activity besides reducing ALDH2 levels, mitochondrial respiration and mitochondrial respiratory reserve capacity and increased cell death. Our results indicate that ALDH2 impairment can lead to poor mitochondrial respiration and increased cell death in cultured H9C2 cardiomyocytes.
Collapse
|
19
|
The MAP3K ZAK, a novel modulator of ERK-dependent migration, is upregulated in colorectal cancer. Oncogene 2015; 35:3190-200. [PMID: 26522728 DOI: 10.1038/onc.2015.379] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 08/30/2015] [Accepted: 09/04/2015] [Indexed: 12/22/2022]
Abstract
Often described as a mediator of cell cycle arrest or as a pro-apoptotic factor in stressful conditions, the MAP3K ZAK (Sterile alpha motif and leucine zipper-containing kinase) has also been proven to positively regulate epidermal growth factor receptor (EGFR) and WNT signaling pathways, cancer cell proliferation and cellular neoplastic transformation. Here, we show that both isoforms of ZAK, ZAK-α and ZAK-β are key factors in cancer cell migration. While ZAK depletion reduced cell motility of HeLa and HCT116 cells, its overexpression triggered the activation of all three mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK) and p38, as well as an increase in cell motion. On the contrary, the kinase-dead mutants, ZAK-α K45M and ZAK-β K45M, were not able to provoke such events, and instead exerted a dominant-negative effect on MAPK activation and cell migration. Pharmacological inhibition of ZAK by nilotinib, preventing ZAK-autophosphorylation and thereby auto-activation, led to the same results. Activated by epidermal growth factor (EGF), we further showed that ZAK constitutes an essential element of the EGF/ERK-dependent cell migration pathway. Using public transcriptomic databases and tissue microarrays, we finally established that, as strong factors of the EGFR signaling pathway, ZAK-α and/or ZAK-β transcripts and protein(s) are frequently upregulated in colorectal adenoma and carcinoma patients. Notably, gene set enrichment analysis disclosed a significant correlation between ZAK+ colorectal premalignant lesions and gene sets belonging to the MAPK/ERK and motility-related signaling pathways of the reactome database, strongly suggesting that ZAK induces such pro-tumoral reaction cascades in human cancers.
Collapse
|
20
|
Hsieh YL, Tsai YL, Shibu MA, Su CC, Chung LC, Pai P, Kuo CH, Yeh YL, Viswanadha VP, Huang CY. ZAK induces cardiomyocyte hypertrophy and brain natriuretic peptide expression via p38/JNK signaling and GATA4/c-Jun transcriptional factor activation. Mol Cell Biochem 2015; 405:1-9. [PMID: 25869677 DOI: 10.1007/s11010-015-2389-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 03/26/2015] [Indexed: 01/19/2023]
Abstract
Cardiomyocyte hypertrophy is an adaptive response of heart to various stress conditions. During the period of stress accumulation, transition from physiological hypertrophy to pathological hypertrophy results in the promotion of heart failure. Our previous studies found that ZAK, a sterile alpha motif and leucine zipper containing kinase, was highly expressed in infarcted human hearts and demonstrated that overexpression of ZAK induced cardiac hypertrophy. This study evaluates, cellular events associated with the expression of two doxycycline (Dox) inducible Tet-on ZAK expression systems, a Tet-on ZAK WT (wild-type), and a Tet-on ZAK DN (mutant, Dominant-negative form) in H9c2 myoblast cells; Tet-on ZAK WT was found to increase cell size and hypertrophic marker BNP in a dose-dependent manner. To ascertain the mechanism of ZAK-mediated hypertrophy, expression analysis with various inhibitors of the related upstream and downstream proteins was performed. Tet-on ZAK WT expression triggered the p38 and JNK pathway and also activated the expression and nuclear translocation of p-GATA4 and p-c-Jun transcription factors, without the involvement of p-ERK or NFATc3. However, Tet-on ZAK DN showed no effect on the p38 and JNK signaling cascade. The results showed that the inhibitors of JNK1/2 and p38 significantly suppressed ZAK-induced BNP expression. The results show the role of ZAK and/or the ZAK downstream events such as JNK and p38 phosphorylation, c-Jun, and GATA-4 nuclear translocation in cardiac hypertrophy. ZAK and/or the ZAK downstream p38, and JNK pathway could therefore be potential targets to ameliorate cardiac hypertrophy symptoms in ZAK-overexpressed patients.
Collapse
Affiliation(s)
- You-Liang Hsieh
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Vin H, Ojeda SS, Ching G, Leung ML, Chitsazzadeh V, Dwyer DW, Adelmann CH, Restrepo M, Richards KN, Stewart LR, Du L, Ferguson SB, Chakravarti D, Ehrenreiter K, Baccarini M, Ruggieri R, Curry JL, Kim KB, Ciurea AM, Duvic M, Prieto VG, Ullrich SE, Dalby KN, Flores ER, Tsai KY. BRAF inhibitors suppress apoptosis through off-target inhibition of JNK signaling. eLife 2013; 2:e00969. [PMID: 24192036 PMCID: PMC3814616 DOI: 10.7554/elife.00969] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vemurafenib and dabrafenib selectively inhibit the v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) kinase, resulting in high response rates and increased survival in melanoma. Approximately 22% of individuals treated with vemurafenib develop cutaneous squamous cell carcinoma (cSCC) during therapy. The prevailing explanation for this is drug-induced paradoxical ERK activation, resulting in hyperproliferation. Here we show an unexpected and novel effect of vemurafenib/PLX4720 in suppressing apoptosis through the inhibition of multiple off-target kinases upstream of c-Jun N-terminal kinase (JNK), principally ZAK. JNK signaling is suppressed in multiple contexts, including in cSCC of vemurafenib-treated patients, as well as in mice. Expression of a mutant ZAK that cannot be inhibited reverses the suppression of JNK activation and apoptosis. Our results implicate suppression of JNK-dependent apoptosis as a significant, independent mechanism that cooperates with paradoxical ERK activation to induce cSCC, suggesting broad implications for understanding toxicities associated with BRAF inhibitors and for their use in combination therapies. DOI: http://dx.doi.org/10.7554/eLife.00969.001.
Collapse
Affiliation(s)
- Harina Vin
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
The H9C2 cell line and primary neonatal cardiomyocyte cells show similar hypertrophic responses in vitro. In Vitro Cell Dev Biol Anim 2010; 47:125-31. [DOI: 10.1007/s11626-010-9368-1] [Citation(s) in RCA: 258] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Accepted: 10/20/2010] [Indexed: 11/27/2022]
|
23
|
Garlena RA, Gonda RL, Green AB, Pileggi RM, Stronach B. Regulation of mixed-lineage kinase activation in JNK-dependent morphogenesis. J Cell Sci 2010; 123:3177-88. [PMID: 20736302 DOI: 10.1242/jcs.063313] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Normal cells respond appropriately to various signals, while sustaining proper developmental programs and tissue homeostasis. Inappropriate signal reception, response or attenuation, can upset the normal balance of signaling within cells, leading to dysfunction or tissue malformation. To understand the molecular mechanisms that regulate protein-kinase-based signaling in the context of tissue morphogenesis, we analyzed the domain requirements of Drosophila Slpr, a mixed-lineage kinase (MLK), for Jun N-terminal kinase (JNK) signaling. The N-terminal half of Slpr is involved in regulated signaling whereas the C-terminal half promotes cortical protein localization. The SH3 domain negatively regulates Slpr activity consistent with autoinhibition via a conserved proline motif. Also, like many kinases, conserved residues in the activation segment of the catalytic domain regulate Slpr. Threonine 295, in particular, is essential for function. Slpr activation requires dual input from the MAP4K Misshapen (Msn), through its C-terminal regulatory domain, and the GTPase Rac, which both bind to the LZ-CRIB region of Slpr in vitro. Although Rac is sufficient to activate JNK signaling, our results indicate that there are Slpr-independent functions for Rac in dorsal closure. Finally, expression of various Slpr constructs alone or with upstream activators reveals a wide-ranging response at the cell and tissue level.
Collapse
Affiliation(s)
- Rebecca A Garlena
- University of Pittsburgh, Department of Biological Sciences, Pittsburgh, PA 15260, USA
| | | | | | | | | |
Collapse
|
24
|
Rix U, Remsing Rix LL, Terker AS, Fernbach NV, Hantschel O, Planyavsky M, Breitwieser FP, Herrmann H, Colinge J, Bennett KL, Augustin M, Till JH, Heinrich MC, Valent P, Superti-Furga G. A comprehensive target selectivity survey of the BCR-ABL kinase inhibitor INNO-406 by kinase profiling and chemical proteomics in chronic myeloid leukemia cells. Leukemia 2010; 24:44-50. [PMID: 19890374 DOI: 10.1038/leu.2009.228] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 09/09/2009] [Accepted: 10/02/2009] [Indexed: 12/27/2022]
Abstract
Resistance to the BCR-ABL tyrosine kinase inhibitor imatinib poses a pressing challenge in treating chronic myeloid leukemia (CML). This resistance is often caused by point mutations in the ABL kinase domain or by overexpression of LYN. The second-generation BCR-ABL inhibitor INNO-406 is known to inhibit most BCR-ABL mutants and LYN efficiently. Knowledge of its full target spectrum would provide the molecular basis for potential side effects or suggest novel therapeutic applications and possible combination therapies. We have performed an unbiased chemical proteomics native target profile of INNO-406 in CML cells combined with functional assays using 272 recombinant kinases thereby identifying several new INNO-406 targets. These include the kinases ZAK, DDR1/2 and various ephrin receptors. The oxidoreductase NQO2, inhibited by both imatinib and nilotinib, is not a relevant target of INNO-406. Overall, INNO-406 has an improved activity over imatinib but a slightly broader target profile than both imatinib and nilotinib. In contrast to dasatinib and bosutinib, INNO-406 does not inhibit all SRC kinases and most TEC family kinases and is therefore expected to elicit fewer side effects. Altogether, these properties may make INNO-406 a valuable component in the drug arsenal against CML.
Collapse
Affiliation(s)
- U Rix
- CeMM - Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Huang CY, Yang LC, Liu KY, Chang IC, Liao PH, Chou JIY, Chou MY, Lin WW, Yang JJ. ZAK negatively regulates RhoGDIbeta-induced Rac1-mediated hypertrophic growth and cell migration. J Biomed Sci 2009; 16:56. [PMID: 19538723 PMCID: PMC2703632 DOI: 10.1186/1423-0127-16-56] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Accepted: 06/18/2009] [Indexed: 11/10/2022] Open
Abstract
RhoGDIbeta, a Rho GDP dissociation inhibitor, induced hypertrophic growth and cell migration in a cultured cardiomyoblast cell line, H9c2. We demonstrated that RhoGDIbeta plays a previously undefined role in regulating Rac1 expression through transcription to induce hypertrophic growth and cell migration and that these functions are blocked by the expression of a dominant-negative form of Rac1. We also demonstrated that knockdown of RhoGDIbeta expression by RNA interference blocked RhoGDIbeta-induced Rac1 expression and cell migration. We demonstrated that the co-expression of ZAK and RhoGDIbeta in cells resulted in an inhibition in the activity of ZAK to induce ANF expression. Knockdown of ZAK expression in ZAK-RhoGDIbeta-expressing cells by ZAK-specific RNA interference restored the activities of RhoGDIbeta.
Collapse
Affiliation(s)
- Chih-Yang Huang
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 404, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cheng YC, Kuo WW, Wu HC, Lai TY, Wu CH, Hwang JM, Wang WH, Tsai FJ, Yang JJ, Huang CY, Chu CH. ZAK induces MMP-2 activity via JNK/p38 signals and reduces MMP-9 activity by increasing TIMP-1/2 expression in H9c2 cardiomyoblast cells. Mol Cell Biochem 2009; 325:69-77. [DOI: 10.1007/s11010-008-0021-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 12/30/2008] [Indexed: 10/21/2022]
|
27
|
Huang CY, Yang LC, Liu KY, Liao PH, Chou JIY, Chou MY, Lin WW, Yang JJ. RhoGDIbeta-induced hypertrophic growth in H9c2 cells is negatively regulated by ZAK. J Biomed Sci 2009; 16:11. [PMID: 19272173 PMCID: PMC2653512 DOI: 10.1186/1423-0127-16-11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 01/22/2009] [Indexed: 12/20/2022] Open
Abstract
We found that overexpression of RhoGDIβ, a Rho GDP dissociation inhibitor, induced hypertrophic growth and suppressed cell cycle progression in a cultured cardiomyoblast cell line. Knockdown of RhoGDIβ expression by RNA interference blocked hypertrophic growth. We further demonstrated that RhoGDIβ physically interacts with ZAK and is phosphorylated by ZAK in vitro, and this phosphorylation negatively regulates RhoGDIβ functions. Moreover, the ZAK-RhoGDIβ interaction may maintain ZAK in an inactive hypophosphorylated form. These two proteins could negatively regulate one another such that ZAK suppresses RhoGDIβ functions through phosphorylation and RhoGDIβ counteracts the effects of ZAK by physical interaction. Knockdown of ZAK expression in ZAK- and RhoGDIβ-expressing cells by ZAK-specific RNA interference restored the full functions of RhoGDIβ.
Collapse
Affiliation(s)
- Chih-Yang Huang
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 404, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Eisenberger S, Ackermann K, Voggenreiter G, Sültmann H, Kasperk C, Pyerin W. Metastases and multiple myeloma generate distinct transcriptional footprints in osteocytes in vivo. J Pathol 2008; 214:617-26. [DOI: 10.1002/path.2322] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|