1
|
Zou X, Yuan M, Zhou W, Cai A, Cheng Y, Zhan Z, Zhang Y, Pan Z, Hu X, Zheng S, Liu T, Huang P. SOX17 Prevents Endothelial-Mesenchymal Transition of Pulmonary Arterial Endothelial Cells in Pulmonary Hypertension through Mediating TGF-β/Smad2/3 Signaling. Am J Respir Cell Mol Biol 2025; 72:364-379. [PMID: 39392679 DOI: 10.1165/rcmb.2023-0355oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/11/2024] [Indexed: 10/12/2024] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) has been reported to contribute to pulmonary vascular remodeling in patients with pulmonary hypertension (PH). Our study demonstrates that SOX17, a member of the SOX (SRY-Box) transcription factor family, plays a role in regulating pulmonary arterial homeostasis through extracellular vesicles in an autocrine and paracrine manner. However, the role of SOX17 in mediating EndMT of pulmonary arterial endothelial cells (PAECs) and its intracellular mechanisms remain unclear. Here we present evidence showing that downregulation of SOX17 expression is accompanied by significant pulmonary arterial EndMT and activation of the TGF-β/Smad2/3 signaling pathway in patients with idiopathic PH and rats with PH induced by Sugen 5416/hypoxia. In primary human PAECs, canonical TGF-β (transforming growth factor-β) signaling inhibits the expression of SOX17. Overexpression of SOX17 reverses TGF-β- and hypoxia-induced EndMT. These findings suggest that SOX17 is essential for human PAECs to undergo TGF-β-mediated EndMT. Mechanistically, our data demonstrate that SOX17 prevents TGF-β-induced EndMT by suppressing ROCK1 (Rho-associated kinase 1) expression through binding to the specific promoter region of ROCK1, thereby inhibiting MYPT1 (myosin phosphatase target subunit 1) and MLC (myosin light chain) phosphorylation. Furthermore, we show that Tie2-Cre rats with endothelial cell-specific overexpression of SOX17 are protected against Sugen/hypoxia-induced EndMT and subsequent pulmonary vascular remodeling. Consistent with the in vitro results, compared with Tie2-Cre rats treated with Sugen/hypoxia alone, rats overexpressing SOX17 exhibited reduced levels of ROCK1 as well as decreased phosphorylation levels of MYPT1 and MLC. Overall, our studies unveil a novel TGF-β/SOX17/ROCK1 pathway involved in regulating PAECs' EndMT process, and we propose the targeting of SOX17 as a potential therapeutic strategy for alleviating pulmonary vascular remodeling in PH.
Collapse
Affiliation(s)
- Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Mengnan Yuan
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Wei Zhou
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Anqi Cai
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Yili Cheng
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Zibo Zhan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaoping Hu
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Shuilian Zheng
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Ting Liu
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China; and
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, and Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
2
|
Chen Y, Xu F, Chen F, Li S, Wu M, Chen S, Chen J, Yang Z, Sun Z, Chen Z. Hydrogen sulfide-mediated inhibition of ROCK 2 exerts a vasoprotective effect on ischemic brain injury. Am J Physiol Cell Physiol 2025; 328:C986-C1000. [PMID: 39716722 DOI: 10.1152/ajpcell.00708.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
As a gas molecule, hydrogen sulfide (H2S) exerts neuroprotective effects. Despite its recognized importance, there remains a need for a deeper understanding of H2S's impact on vascular smooth muscle cells and its role in ischemic brain injury. This study employs encompassing cultured primary cerebral vascular smooth muscle cells, oxygen-glucose deprivation/reoxygenation model, in vitro vascular tone assessments, in vivo middle cerebral artery occlusion and reperfusion experimentation in male rats, and the utilization of Rho-associated coiled-coil containing protein kinase 2 (ROCK2) knockout, to unravel the intricate relationship between H2S and cerebrovascular diastolic function. Our findings show that RhoA activation induces heightened vascular smooth muscle cell (VSMC) contraction, whereas the introduction of exogenous H2S mitigates the relaxant effect of the middle cerebral artery in rats through the downregulation of both ROCK1 and ROCK2, with ROCK2 exhibiting a more pronounced effect. Correspondingly, the attenuation of ROCK2 expression yields a more substantial reduction in the protective impact of H2S on cerebral blood flow, as well as learning and memory ability in ischemic injury, compared with the decrease in ROCK1 expression. Moreover, we demonstrate that H2S effectively mitigates the damage induced by oxygen-glucose deprivation/reoxygenation in male mouse primary vascular smooth muscle cells. This effect is characterized by enhanced cell proliferation, reduced lactate dehydrogenase leakage, elevated superoxide dismutase activity, and inhibited apoptosis. Notably, this protective effect is markedly diminished in cells derived from ROCK2 knockout mice. Our study reveals that H2S can relax cerebral vascular smooth muscle and ameliorate ischemic stroke injury by inhibiting the ROCK, with a particular emphasis on the role of ROCK2.NEW & NOTEWORTHY This study employs a diverse array of methods; our collective findings indicate that H2S safeguards against ischemic brain injury by inhibiting ROCK activity, thereby promoting relaxation of cerebral smooth muscle and mitigating the impairment of cerebral smooth muscle cell function caused by oxygen-glucose deprivation/reoxygenation. In addition, our data underscore the critical role of ROCK2 in mediating the cerebral protective effects of H2S, surpassing that of ROCK1.
Collapse
MESH Headings
- Animals
- rho-Associated Kinases/metabolism
- rho-Associated Kinases/genetics
- rho-Associated Kinases/antagonists & inhibitors
- Hydrogen Sulfide/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Rats
- Rats, Sprague-Dawley
- Mice
- Brain Ischemia/pathology
- Brain Ischemia/drug therapy
- Brain Ischemia/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Mice, Knockout
- Cerebrovascular Circulation/drug effects
- Infarction, Middle Cerebral Artery/enzymology
- Infarction, Middle Cerebral Artery/pathology
- Neuroprotective Agents/pharmacology
- Mice, Inbred C57BL
- Glucose/deficiency
- Cells, Cultured
- Reperfusion Injury
Collapse
Affiliation(s)
- Ye Chen
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Fangfang Xu
- Department of Phase I Clinical Trials Laboratory, The First Affiliated of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Fang Chen
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Shuaishuai Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, People's Republic of China
| | - Miao Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Shuo Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Jinhua Chen
- Department of Clinical Central Laboratory, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - Zhaoyi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, People's Republic of China
- Department of Phase I Clinical Trials Laboratory, The First Affiliated of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
3
|
Lange M, Francis C, Furtado J, Kim YB, Liao JK, Eichmann A. Endothelial Rho kinase controls blood vessel integrity and angiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624343. [PMID: 39605538 PMCID: PMC11601598 DOI: 10.1101/2024.11.19.624343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background The Rho kinases 1 and 2 (ROCK1/2) are serine-threonine specific protein kinases that control actin cytoskeleton dynamics. They are expressed in all cells throughout the body, including cardiomyocytes, smooth muscle cells and endothelial cells, and intimately involved in cardiovascular health and disease. Pharmacological ROCK inhibition is beneficial in mouse models of hypertension, atherosclerosis, and neointimal thickening that display overactivated ROCK. However, the consequences of endothelial ROCK signaling deficiency in vivo remain unknown. To address this issue, we analyzed endothelial cell (EC) specific ROCK1 and 2 deletions. Methods We generated Cdh5-CreERT2 driven, tamoxifen inducible loss of function alleles of ROCK1 and ROCK2 and analyzed mouse survival and vascular defects through cellular, biochemical, and molecular biology approaches. Results We observed that postnatal or adult loss of endothelial ROCK1 and 2 was lethal within a week. Mice succumbed to multi-organ hemorrhage that occurred because of loss of vascular integrity. ECs displayed deficient cytoskeletal actin polymerization that prevented focal adhesion formation and disrupted junctional integrity. Retinal sprouting angiogenesis was also perturbed, as sprouting vessels exhibited lack of polymerized actin and defective lumen formation. In a three-dimensional endothelial sprouting assay, combined knockdown of ROCK1/2 or knockdown or ROCK2 but not ROCK1 led to reduced sprouting, lumenization and cell polarization defects caused by defective actin and altered VE-cadherin dynamics. The isoform specific role of endothelial ROCK2 correlated with ROCK2 substrate specificity for FAK and LIMK. By analyzing single and three allele mutants we show that one intact allele of ROCK2 is sufficient to maintain vascular integrity in vivo. Conclusion Endothelial ROCK1 and 2 maintain junctional integrity and ensure proper angiogenesis and lumen formation. The presence of one allele of ROCK2 is sufficient to maintain vascular growth and integrity. These data indicate the need of careful consideration for the use of ROCK inhibitors in disease settings.
Collapse
Affiliation(s)
- Martin Lange
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Caitlin Francis
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Mass, USA
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - James K Liao
- Division of Cardiology/Sarver Heart Center, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| |
Collapse
|
4
|
Knipe RS, Gosens R. Paving the ROCKy Path to Novel Antifibrotics. Am J Respir Cell Mol Biol 2024; 71:381-382. [PMID: 38864857 PMCID: PMC11450308 DOI: 10.1165/rcmb.2024-0224ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Rachel S Knipe
- Division of Pulmonary and Critical Care Medicine Massachusetts General Hospital Boston, Massachusetts
| | - Reinoud Gosens
- Department of Molecular Pharmacology University of Groningen Groningen, the Netherlands
| |
Collapse
|
5
|
Bao W, Cheng M, Chen X, Wang T, Xu D, Liao H, Chen L, Wen F, He J, Chen J. Effect of fasudil on clinical outcomes of pulmonary hypertension: a systematic review and meta-analysis. Expert Rev Clin Pharmacol 2024:1-18. [PMID: 39269366 DOI: 10.1080/17512433.2024.2404688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a life-threatening condition with high mortality, categorized into Group 1-5 by distinct etiologies. Fasudil, a potent vasodilator targeting RhoA/Rho kinase pathway, holds promise for diverse PH pathologies. However, a systematic evaluation of its clinical benefits remains elusive. METHODS We conducted a systematic search in several databases. Meta-analysis using odds ratio and mean difference was performed, with an assessment of studies' quality and pooled evidences. RESULTS Inclusion of 3269 Group-3 PH patients demonstrated that Fasudil increased effective events, forced expiratory volume in one second (FEV1), 6-minute walking distance (6MWD) and arterial partial pressure of oxygen (PaO2), and decreased mean pulmonary artery pressure (mPAP) and pulmonary artery systolic pressure (PASP); Inclusion of 197 Group-2 PH patients suggested that Fasudil increased 6MWD and PaO2, and decreased PASP. Subgroup analysis revealed no significant difference between 30 and 60 mg/day dosages of Fasudil, while administration durations and methods might affect its effectiveness in treating Group-3 PH patients. CONCLUSIONS Our study favors the beneficial effects of Fasudil by enhancing FEV1, 6MWD and PaO2, and reducing mPAP and PASP on Group-3 PH patients, suggesting Fasudil as a viable treatment option and highlighting the need for further studies to inform healthcare policies. PROTOCOL REGISTRATION www.crd.york.ac.uk/prospero identifier is CRD42022308947.
Collapse
Affiliation(s)
- Wanying Bao
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetrics and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Mengxin Cheng
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoye Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Dan Xu
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Hualin Liao
- Department of Respiratory Medicine, Hospital of Chengdu office of People's Government of Tibetan Autonomous Region of China, Chengdu, Sichuan, China
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Fuqiang Wen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Junyun He
- Department of Respiratory Medicine, Hospital of Chengdu office of People's Government of Tibetan Autonomous Region of China, Chengdu, Sichuan, China
| | - Jun Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Ouellette J, Lacoste B. Rock2 heterozygosity improves recognition memory and endothelial function in a mouse model of 16p11.2 deletion autism syndrome. Neurosci Lett 2024; 837:137904. [PMID: 39029613 DOI: 10.1016/j.neulet.2024.137904] [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: 07/06/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Rho-associated protein kinase-2 (ROCK2) is a critical player in many cellular processes and was incriminated in cardiovascular and neurological disorders. Recent evidence has shown that non-selective pharmacological blockage of ROCKs ameliorates behavioral alterations in a mouse model of 16p11.2 haploinsufficiency. We had revealed that 16p11.2-deficient mice also display cerebrovascular abnormalities, including endothelial dysfunction. To investigate whether genetic blockage of ROCK2 also exerts beneficial effects on cognition and angiogenesis, we generated mice with both 16p11.2 and Rock2 haploinsufficiency (16p11.2df/+;Rock2+/-). We find that Rock2 heterozygosity on a 16p11.2df/+ background significantly improved recognition memory. Furthermore, brain endothelial cells from 16p11.2df/+;Rock2+/- mice display improved angiogenic capacity compared to cells from 16p11.2df/+ littermates. Overall, this study implicates Rock2 gene as a modulator of 16p11.2-associated alterations, highlighting its potential as a target for treatment of autism spectrum disorders.
Collapse
Affiliation(s)
- Julie Ouellette
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
7
|
Akamine T, Terabayashi T, Sasaki T, Hayashi R, Abe I, Hirayama F, Nureki SI, Ikawa M, Miyata H, Tokunaga A, Kobayashi T, Hanada K, Thumkeo D, Narumiya S, Ishizaki T. Conditional deficiency of Rho-associated kinases disrupts endothelial cell junctions and impairs respiratory function in adult mice. FEBS Open Bio 2024; 14:906-921. [PMID: 38604990 PMCID: PMC11148122 DOI: 10.1002/2211-5463.13802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
The Ras homology (Rho) family of GTPases serves various functions, including promotion of cell migration, adhesion, and transcription, through activation of effector molecule targets. One such pair of effectors, the Rho-associated coiled-coil kinases (ROCK1 and ROCK2), induce reorganization of actin cytoskeleton and focal adhesion through substrate phosphorylation. Studies on ROCK knockout mice have confirmed that ROCK proteins are essential for embryonic development, but their physiological functions in adult mice remain unknown. In this study, we aimed to examine the roles of ROCK1 and ROCK2 proteins in normal adult mice. Tamoxifen (TAM)-inducible ROCK1 and ROCK2 single and double knockout mice (ROCK1flox/flox and/or ROCK2flox/flox;Ubc-CreERT2) were generated and administered a 5-day course of TAM. No deaths occurred in either of the single knockout strains, whereas all of the ROCK1/ROCK2 double conditional knockout mice (DcKO) had died by Day 11 following the TAM course. DcKO mice exhibited increased lung tissue vascular permeability, thickening of alveolar walls, and a decrease in percutaneous oxygen saturation compared with noninducible ROCK1/ROCK2 double-floxed control mice. On Day 3 post-TAM, there was a decrease in phalloidin staining in the lungs in DcKO mice. On Day 5 post-TAM, immunohistochemical analysis also revealed reduced staining for vascular endothelial (VE)-cadherin, β-catenin, and p120-catenin at cell-cell contact sites in vascular endothelial cells in DcKO mice. Additionally, VE-cadherin/β-catenin complexes were decreased in DcKO mice, indicating that ROCK proteins play a crucial role in maintaining lung function by regulating cell-cell adhesion.
Collapse
Affiliation(s)
- Takahiro Akamine
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takeshi Terabayashi
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takako Sasaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Riku Hayashi
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Ichitaro Abe
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Yufu, Japan
| | - Fumihiro Hirayama
- Department of Respiratory Medicine and Infectious Diseases, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shin-Ichi Nureki
- Department of Respiratory Medicine and Infectious Diseases, Faculty of Medicine, Oita University, Yufu, Japan
| | - Masahito Ikawa
- Animal Resource Center for Infectious Diseases, Research Institute for Microbial Diseases, Suita, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Suita, Japan
| | - Akinori Tokunaga
- Division of Laboratory Animal Resources, Life Science Research Laboratory, University of Fukui, Eiheiji-cho, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
- Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Yufu, Japan
| | - Katsuhiro Hanada
- Clinical Engineering Research Center, Faculty of Medicine, Oita University, Yufu, Japan
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshimasa Ishizaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
8
|
Rezaei M, Mehta JL, Zadeh GM, Khedri A, Rezaei HB. Myosin light chain phosphatase is a downstream target of Rho-kinase in endothelin-1-induced transactivation of the TGF-β receptor. Cell Biochem Biophys 2024; 82:1109-1120. [PMID: 38834831 DOI: 10.1007/s12013-024-01262-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Rho-kinase (ROCK) regulates actomyosin contraction, coronary vasospasm, and cytoskeleton dynamics. ROCK and of NADPH oxidase (NOX) play an essential role in cardiovascular disease and proteoglycan synthesis, which promotes atherosclerosis by trapping low density lipoprotein. ROCK is activated by endothelin-1 (ET1) and transactivates the transforming growth factor beta receptor (TGFβR1), intensifying Smad signaling and proteoglycan production. This study aimed to identify the role of myosin light chain phosphatase (MLCP) as a downstream target of ROCK in TβR1 transactivation. METHODS Vascular smooth muscle cells were treated with ET1 and inhibitors of ROCK and MLCP were added. The phosphorylation levels of Smad2C, myosin light chain (MLC), and MLCP were monitored by western blot, and the mRNA expression of chondroitin 4-O-sulfotransferase 1 (C4ST1) was assessed by quantitative real-time PCR. RESULTS We examined ROCK's role in ET1-induced TGFβR1 activation. ROCK phosphorylated MLCP at the MYPT1 T853 residue, blocked by the ROCK inhibitor Y27632. ROCK also increased MLC phosphorylation and actomyosin contraction in response to ET1, enhanced by the phosphatase inhibitor Calyculin A. Calyculin A also increased C4ST1 expression, GAG-chain synthesizing enzymes. CONCLUSIONS This work suggests that ROCK is involved in ET1-mediated TβR1 activation through increased MLCP phosphorylation, which leads to Smad2C phosphorylation and stimulates C4ST1 expression.
Collapse
Affiliation(s)
- Maryam Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jawahar Lal Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Ghorban Mohammad Zadeh
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azam Khedri
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
9
|
Bickel MA, Sherry DM, Bullen EC, Vance ML, Jones KL, Howard EW, Conley SM. Microvascular smooth muscle cells exhibit divergent phenotypic switching responses to platelet-derived growth factor and insulin-like growth factor 1. Microvasc Res 2024; 151:104609. [PMID: 37716411 PMCID: PMC10842624 DOI: 10.1016/j.mvr.2023.104609] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) phenotypic switching is critical for normal vessel formation, vascular stability, and healthy brain aging. Phenotypic switching is regulated by mediators including platelet derived growth factor (PDGF)-BB, insulin-like growth factor (IGF-1), as well as transforming growth factor-β (TGF-β) and endothelin-1 (ET-1), but much about the role of these factors in microvascular VSMCs remains unclear. METHODS We used primary rat microvascular VSMCs to explore PDGF-BB- and IGF-1-induced phenotypic switching. RESULTS PDGF-BB induced an early proliferative response, followed by formation of polarized leader cells and rapid, directionally coordinated migration. In contrast, IGF-1 induced cell hypertrophy, and only a small degree of migration by unpolarized cells. TGF-β and ET-1 selectively inhibit PDGF-BB-induced VSMC migration primarily by repressing migratory polarization and formation of leader cells. Contractile genes were downregulated by both growth factors, while other genes were differentially regulated by PDGF-BB and IGF-1. CONCLUSIONS These studies indicate that PDGF-BB and IGF-1 stimulate different types of microvascular VSMC phenotypic switching characterized by different modes of cell migration. Our studies are consistent with a chronic vasoprotective role for IGF-1 in VSMCs in the microvasculature while PDGF is more involved in VSMC proliferation and migration in response to acute activities such as neovascularization. Better understanding of the nuances of the phenotypic switching induced by these growth factors is important for our understanding of a variety of microvascular diseases.
Collapse
Affiliation(s)
- Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Ken L Jones
- Bioinformatic Solutions, LLC, Sheridan, WY 82801, United States of America
| | - Eric W Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America.
| |
Collapse
|
10
|
Raman-Nair J, Cron G, MacLeod K, Lacoste B. Sex-Specific Acute Cerebrovascular Responses to Photothrombotic Stroke in Mice. eNeuro 2024; 11:ENEURO.0400-22.2023. [PMID: 38164600 PMCID: PMC10849032 DOI: 10.1523/eneuro.0400-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/11/2023] [Accepted: 11/25/2023] [Indexed: 01/03/2024] Open
Abstract
Mechanisms underlying cerebrovascular stroke outcomes are poorly understood, and the effects of biological sex on cerebrovascular regulation post-stroke have yet to be fully comprehended. Here, we explore the overlapping roles of gonadal sex hormones and rho-kinase (ROCK), two important modulators of cerebrovascular tone, on the acute cerebrovascular response to photothrombotic (PT) focal ischemia in mice. Male mice were gonadectomized and female mice were ovariectomized to remove gonadal hormones, whereas control ("intact") animals received a sham surgery prior to stroke induction. Intact wild-type (WT) males showed a delayed drop in cerebral blood flow (CBF) compared with intact WT females, whereby maximal CBF drop was observed 48 h following stroke. Gonadectomy in males did not alter this response. However, ovariectomy in WT females produced a "male-like" phenotype. Intact Rock2+/- males also showed the same phenotypic response, which was not altered by gonadectomy. Alternatively, intact Rock2+/- females showed a significant difference in CBF values compared with intact WT females, displaying higher CBF values immediately post-stroke and showing a maximal CBF drop 48 h post-stroke. This pattern was not altered by ovariectomy. Altogether, these data illustrate sex differences in acute CBF responses to PT stroke, which seem to involve gonadal female sex hormones and ROCK2. Overall, this study provides a framework for exploring sex differences in acute CBF responses to focal ischemic stroke in mice.
Collapse
Affiliation(s)
- Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Gregory Cron
- Neurology Department, Stanford University, Stanford 94305, California
| | - Kathleen MacLeod
- Pharmaceutical Sciences, University of British Colombia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
11
|
Oh SB, Cho S, Kim HJ, Kim SJ. Differential expression of the enzymes regulating myosin light chain phosphorylation are responsible for the slower relaxation of pulmonary artery than mesenteric artery in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:49-57. [PMID: 38154964 PMCID: PMC10762492 DOI: 10.4196/kjpp.2024.28.1.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/30/2023]
Abstract
While arterial tone is generally determined by the phosphorylation of Ser19 in myosin light chain (p-MLC2), Thr18/Ser19 diphosphorylation of MLC2 (pp-MLC2) has been suggested to hinder the relaxation of smooth muscle. In a dual-wire myography of rodent pulmonary artery (PA) and mesenteric artery (MA), we noticed significantly slower relaxation in PA than in MA after 80 mM KCl-induced condition (80K-contraction). Thus, we investigated the MLC2 phosphorylation and the expression levels of its regulatory enzymes; soluble guanylate cyclase (sGC), Rho-A dependent kinase (ROCK) and myosin light chain phosphatase target regulatory subunit (MYPT1). Immunoblotting showed higher sGC-α and ROCK2 in PA than MA, while sGC-β and MYPT1 levels were higher in MA than in PA. Interestingly, the level of pp-MLC2 was higher in PA than in MA without stimulation. In the 80K-contraction state, the levels of p-MLC2 and pp-MLC2 were commonly increased. Treatment with the ROCK inhibitor (Y27632, 10 μM) reversed the higher pp-MLC2 in PA. In the myography study, pharmacological inhibition of sGC (ODQ, 10 μM) slowed relaxation during washout, which was more pronounced in PA than in MA. The simultaneous treatment of Y27632 and ODQ reversed the impaired relaxation in PA and MA. Although treatment of PA with Y27632 alone could increase the rate of relaxation, it was still slower than that of MA without Y27632 treatment. Taken together, we suggest that the higher ROCK and lower MYPT in PA would have induced the higher level of MLC2 phosphorylation, which is responsible for the characteristic slow relaxation in PA.
Collapse
Affiliation(s)
- Seung Beom Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Suhan Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
12
|
He X, Fang J, Gong M, Zhang J, Xie R, Zhao D, Gu Y, Ma L, Pang X, Cui Y. Identification of immune-associated signatures and potential therapeutic targets for pulmonary arterial hypertension. J Cell Mol Med 2023; 27:3864-3877. [PMID: 37753829 PMCID: PMC10718157 DOI: 10.1111/jcmm.17962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 08/09/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) comprises a heterogeneous group of diseases with diverse aetiologies. It is characterized by increased pulmonary arterial pressure and right ventricular (RV) failure without specific drugs for treatment. Emerging evidence suggests that inflammation and autoimmune disorders are common features across all PAH phenotypes. This provides a novel idea to explore the characteristics of immunological disorders in PAH and identify immune-related genes or biomarkers for specific anti-remodelling regimens. In this study, we integrated three gene expression profiles and performed Gene Ontology (GO) and KEGG pathway analysis. CIBERSORT was utilized to estimate the abundance of tissue-infiltrating immune cells in PAH. The PPI network and machine learning were constructed to identify immune-related hub genes and then evaluate the relationship between hub genes and differential immune cells using ImmucellAI. Additionally, we implemented molecular docking to screen potential small-molecule compounds based on the obtained genes. Our findings demonstrated the density and distribution of infiltrating CD4 T cells in PAH and identified four immune-related genes (ROCK2, ATHL1, HSP90AA1 and ACTR2) as potential targets. We also listed 20 promising molecules, including TDI01953, pemetrexed acid and radotinib, for PAH treatment. These results provide a promising avenue for further research into immunological disorders in PAH and potential novel therapeutic targets.
Collapse
Affiliation(s)
- Xu He
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| | - Jiansong Fang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Mingli Gong
- Department of PharmacyPeking University First HospitalBeijingChina
- School of PharmacyXu Zhou Medical UniversityXuzhouChina
| | - Juqi Zhang
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| | - Ran Xie
- Department of PharmacyPeking University First HospitalBeijingChina
| | - Dai Zhao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yanlun Gu
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| | - Lingyue Ma
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| | - Xiaocong Pang
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| | - Yimin Cui
- Department of PharmacyPeking University First HospitalBeijingChina
- Institute of Clinical PharmacologyPeking University First HospitalBeijingChina
| |
Collapse
|
13
|
Cho S, Oh SB, Kim HJ, Kim SJ. T18/S19 diphosphorylation of myosin regulatory light chain impairs pulmonary artery relaxation in monocrotaline-induced pulmonary hypertensive rats. Pflugers Arch 2023; 475:1097-1112. [PMID: 37422604 DOI: 10.1007/s00424-023-02836-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
Phosphorylation of Ser19 (S19-p) on the myosin regulatory light chain (MLC2) is critical for arterial contraction. It has been shown that elevated RhoA-dependent kinase (ROCK) activity or decreased MLC phosphatase (MLCP) activity leads to further phosphorylation of Thr18 (T18/S19-pp), which has been linked to vasospastic diseases. However, this phenomenon has not yet been studied in the context of pulmonary arterial hypertension (PAH). In the monocrotaline-induced PAH (PAH-MCT) rat model, we observed a significant delay in pulmonary artery (PA) relaxation following high potassium-induced contraction, which persisted even with the use of an L-type calcium channel blocker or in a calcium-free solution. Immunoblot analysis showed increased levels of both S19-p and T18/S19-pp in unstimulated PAs from PAH-MCT rats. Proteomics analysis revealed a reduction in soluble guanylate cyclase (sGC) and protein kinase G (PKG) levels, and immunoblotting confirmed decreased levels of MYPT1 (a component of MLCP) and increased ROCK in PAH-MCT. In the control PAs, the pharmacological inhibition of sGC with ODQ resulted in a prominent delay of relaxation and increased T18/S19-pp as in PAH-MCT. The delayed relaxation and the T18/S19-pp in PAH-MCT were reversed by ROCK inhibitor, Y27632, while not by membrane permeable 8-Br-cGMP. The delayed relaxation and T18/S19-diP in the ODQ-treated control PA were also reversed by Y27632. Taken together, the decreased sGC and MLCP, and increased ROCK increased T18/S19-pp, which leads to the decreased ability of PA to relax in PAH-MCT rats. PA specific inhibition of ROCK or activation of MLCP are expected to serve as potential drugs in the treatment of PAH.
Collapse
Affiliation(s)
- Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Seung Beom Oh
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Hae Jin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, MO, Columbia, USA
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
14
|
Nishimiya K, Takahashi J, Oyama K, Matsumoto Y, Yasuda S, Shimokawa H. Mechanisms of Coronary Artery Spasm. Eur Cardiol 2023; 18:e39. [PMID: 37456775 PMCID: PMC10345984 DOI: 10.15420/ecr.2022.55] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/22/2023] [Indexed: 07/18/2023] Open
Abstract
Recent clinical trials have highlighted that percutaneous coronary intervention in patients with stable angina provides limited additional benefits on top of optimal medical therapy. This has led to much more attention being paid to coronary vasomotion abnormalities regardless of obstructive or non-obstructive arterial segments. Coronary vasomotion is regulated by multiple mechanisms that include the endothelium, vascular smooth muscle cells (VSMCs), myocardial metabolic demand, autonomic nervous system and inflammation. Over the years, several animal models have been developed to explore the central mechanism of coronary artery spasm. This review summarises the landmark studies on the mechanisms of coronary vasospasm demonstrating the central role of Rho-kinase as a molecular switch of VSMC hypercontraction and the important role of coronary adventitial inflammation for Rho-kinase upregulation in VSMCs.
Collapse
Affiliation(s)
- Kensuke Nishimiya
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Jun Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Kazuma Oyama
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Yasuharu Matsumoto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| |
Collapse
|
15
|
Hu Z, Sitkoff D, Glunz PW, Zou Y, Wang C, Muckelbauer JK, Adam LP, Wexler RR, Quan ML. Phthalazinone-based lactams and cyclic ureas as ROCK2 selective inhibitors. Bioorg Med Chem Lett 2023; 88:129304. [PMID: 37119973 DOI: 10.1016/j.bmcl.2023.129304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Derivatives of lactam, cyclic urea and carbamate were explored as aniline amide replacements in a series of phthalazinone-based ROCK inhibitors. Potent ROCK2 inhibitors such as 22 were identified with excellent overall kinase selectivity as well as good isoform selectivity over ROCK1.
Collapse
Affiliation(s)
- Zilun Hu
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA.
| | - Doree Sitkoff
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Peter W Glunz
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Yan Zou
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Cailan Wang
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Jodi K Muckelbauer
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Leonard P Adam
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Ruth R Wexler
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| | - Mimi L Quan
- Research & Early Development, Bristol Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, USA
| |
Collapse
|
16
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
17
|
Lu Q, Sun X, Yegambaram M, Ornatowski W, Wu X, Wang H, Garcia-Flores A, Da Silva V, Zemskov EA, Tang H, Fineman JR, Tieu K, Wang T, Black SM. Nitration-mediated activation of the small GTPase RhoA stimulates cellular glycolysis through enhanced mitochondrial fission. J Biol Chem 2023; 299:103067. [PMID: 36841483 PMCID: PMC10060112 DOI: 10.1016/j.jbc.2023.103067] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/27/2023] Open
Abstract
Mitochondrial fission and a Warburg phenotype of increased cellular glycolysis are involved in the pathogenesis of pulmonary hypertension (PH). The purpose of this study was to determine whether increases in mitochondrial fission are involved in a glycolytic switch in pulmonary arterial endothelial cells (PAECs). Mitochondrial fission is increased in PAEC isolated from a sheep model of PH induced by pulmonary overcirculation (Shunt PAEC). In Shunt PAEC we identified increases in the S616 phosphorylation responsible for dynamin-related protein 1 (Drp1) activation, the mitochondrial redistribution of Drp1, and increased cellular glycolysis. Reducing mitochondrial fission attenuated cellular glycolysis in Shunt PAEC. In addition, we observed nitration-mediated activation of the small GTPase RhoA in Shunt PAEC, and utilizing a nitration-shielding peptide, NipR1 attenuated RhoA nitration and reversed the Warburg phenotype. Thus, our data identify a novel link between RhoA, mitochondrial fission, and cellular glycolysis and suggest that targeting RhoA nitration could have therapeutic benefits for treating PH.
Collapse
Affiliation(s)
- Qing Lu
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Xutong Sun
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | | | - Wojciech Ornatowski
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Xiaomin Wu
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Hui Wang
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Alejandro Garcia-Flores
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Victoria Da Silva
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Evgeny A Zemskov
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Haiyang Tang
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Ting Wang
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Stephen M Black
- Center of Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
18
|
Protective role of Decylubiquinone against secondary melanoma at lung in B16F10 induced mice by reducing E-cadherin expression and ameliorating ROCKII-Limk1/2-Cofiliin mediated metastasis. Cell Signal 2023; 101:110486. [PMID: 36208704 DOI: 10.1016/j.cellsig.2022.110486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Melanoma is one of the most consequential skin cancer with a rising death incidences. Silent but belligerent nature of metastatic sprouting is the leading cause of melanoma related mortality. Invasion of metastatic cells and re-expression of E-Cadherin play the crucial role in the establishment of secondary tumor at distal sites. Thus, manipulation of tumor cell invasion in parallel to regulation of E-Cadherin expression can be considered as potential anti-metastatic strategy. Evidences suggested key role of reactive oxygen species associated ROCK activities in the modulation of metastatic invasion via F-actin stabilization. Here, we first-time report Decylubiquinone, a dietary Coenzyme Q10 analog, as an effective attenuator of pulmonary metastatic melanoma in C57BL/6 mice. Current study depicted detailed molecular interplay associated with Decylubiquinone mediated phosphorylation of ROCKII at Tyr722 along with reduced phosphorylation of ROCKII Ser1366 leading to suppression of Limk1/2-Cofilin-F-actin stabilization axis that finally restricted B16F10 melanoma cell invasion at metastatic site. Analysis further deciphered the role of HNF4α as its nuclear translocation modulated E-Cadherin expression, the effect of reactive oxygen species dependent ROCKII activity in secondarily colonized B16F10 melanoma cells at lungs. Thus unbosoming of related signal orchestra represented Decylubiquinone as a potential remedial agent against secondary lung melanoma.
Collapse
|
19
|
Mechanism of Hypoxia-Mediated Smooth Muscle Cell Proliferation Leading to Vascular Remodeling. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3959845. [PMID: 36593773 PMCID: PMC9805398 DOI: 10.1155/2022/3959845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022]
Abstract
Vascular remodeling refers to changes in the size, contraction, distribution, and flow rate of blood vessels and even changes in vascular function. Vascular remodeling can cause cardiovascular and cerebrovascular diseases. It can also lead to other systemic diseases, such as pulmonary hypertension, pulmonary atherosclerosis, chronic obstructive pulmonary disease, stroke, and ascites of broilers. Hypoxia is one of the main causes of vascular remodeling. Prolonged hypoxia or intermittent hypoxia can lead to loss of lung ventilation, causing respiratory depression, irregular respiratory rhythms, and central respiratory failure. Animals that are unable to adapt to the highland environment are also prone to sustained constriction of the small pulmonary arteries, increased resistance to pulmonary circulation, and impaired blood circulation, leading to pulmonary hypertension and right heart failure if they live in a highland environment for long periods of time. However, limited studies have been found on the relationship between hypoxia and vascular remodeling. Therefore, this review will explore the relationship between hypoxia and vascular remodeling from the aspects of endoplasmic reticulum stress, mitochondrial dysfunction, abnormal calcium channel, disordered cellular metabolism, abnormal expression of miRNA, and other factors. This will help to understand the detailed mechanism of hypoxia-mediated smooth muscle cell proliferation and vascular remodeling for the better treatment and management of diseases due to vascular remodeling.
Collapse
|
20
|
Shen Y, Xu LR, Yan D, Zhou M, Han TL, Lu C, Tang X, Lin CP, Qian RZ, Guo DQ. BMAL1 modulates smooth muscle cells phenotypic switch towards fibroblast-like cells and stabilizes atherosclerotic plaques by upregulating YAP1. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166450. [PMID: 35598770 DOI: 10.1016/j.bbadis.2022.166450] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/03/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ischemic heart diseases and ischemic stroke are closely related to circadian clock and unstable atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) can stabilize or destabilize an atherosclerotic lesion through phenotypic switch. BMAL1 is not only an indispensable core component in circadian clock but also an important regulator in atherosclerosis and VSMCs proliferation. However, little is known about the modulation mechanisms of BMAL1 in VSMCs phenotypic switch and atherosclerotic plaque stability. METHODS We integrated histological analysis of human plaques, in vivo experiments of VSMC-specific Bmal1-/- mice, in vitro experiments, and gene set enrichment analysis (GSEA) of public datasets of human plaques to explore the function of BMAL1 in VSMCs phonotypic switch and plaque stability. FINDINGS Comparing to human unstable plaques, BMAL1 was higher in stable plaques, accompanied by elevated YAP1 and fibroblast maker FSP1 which were positively correlated with BMAL1. In response to Methyl-β-cyclodextrin-cholesterol, oxidized-low-density-lipoprotein and platelet-derived-growth-factor-BB, VSMCs embarked on phenotypic switch and upregulated BMAL, YAP1 and FSP1. Besides, BMAL1 overexpression promoted VSMCs phonotypic switch towards fibroblast-like cells by transcriptionally upregulating the expression of YAP1. BMAL1 or YAP1 knock-down inhibited VSMCs phonotypic switch and downregulated FSP1. Furthermore, VSMC-specific Bmal1-/- mice exhibited VSMCs with lower YAP1 and FSP1 levels, and more vulnerable plaques with less collagen content. In addition, BMAL1 suppressed the migration of VSMCs. The GSEA results of public datasets were consistent with our laboratory findings. INTERPRETATION Our results highlight the importance of BMAL1 as a major regulator in VSMCs phenotypic switch towards fibroblast-like cells which stabilize an atherosclerotic plaque.
Collapse
Affiliation(s)
- Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Li-Rong Xu
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Min Zhou
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Tong-Lei Han
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | - Chang-Po Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| | - Rui-Zhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Rd., Shanghai 200032, China.
| | - Da-Qiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China.
| |
Collapse
|
21
|
Wang W, He Q, Zhuang C, Zhang H, Fan X, Wang Q, Qi M, Sun R, Li C, Yu J. Apatinib Through Activating the RhoA/ROCK Signaling Pathway to Cause Dysfunction of Vascular Smooth Muscle Cells. Appl Biochem Biotechnol 2022; 194:5367-5385. [PMID: 35776338 DOI: 10.1007/s12010-022-04020-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/02/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are associated with differentiated, organized, and contractile phenotype under the effect of various types of physiological conditions those are associated with migratory, proliferative, and synthetic phenotype under the effect of various types of stimuli, which dysfunction drives many cardiovascular diseases. Abnormal cell proliferation and invasion of VSMCs are among the primary causes of hypertension. Apatinib is a small-molecule tyrosine kinase inhibitor (TKI) that highly selectively binds to and strongly inhibits VEGFR-2. Previous studies have confirmed that the TKIs can raise blood pressure through RhoA/ROCK pathway. LARG is a key gene in the RhoA/ROCK pathway and plays a critical role in the continuous vasoconstriction function because it regulates part of signal transduction in VSMCs. In this study, an in vitro experiment was conducted to observe that apatinib caused dysfunction of MOVAS cells through the RhoA/ROCK signalling pathway and Y27632, a nonspecific ROCK inhibitor, and knockout of LARG gene can improve the proliferation, antiapoptosis, oxidative stress, and mitochondrial autophagy of apatinib-induced MOVAS cells. These findings suggest that activation of the RhoA/ROCK signalling pathway could be the underlying mechanism of apatinib-induced dysfunction of MOVAS cells, while ROCK inhibitor and knockout of LARG gene have potential therapeutic value.
Collapse
Affiliation(s)
- Wenjuan Wang
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Qingjian He
- Department of Breast and Thyroid Surgery, Zhoushan Hospital of Zhejiang Province, Zhoushan, China
| | - Chenchen Zhuang
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Haodong Zhang
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Xin Fan
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Qiongying Wang
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Miaomiao Qi
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Runmin Sun
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Caie Li
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China
| | - Jing Yu
- Department of Hypertension Center, Lanzhou University Second Hospital, Chengguan District, CuiyingmenLanzhou, No. 82, China.
| |
Collapse
|
22
|
Valipour M. Different Aspects of Emetine's Capabilities as a Highly Potent SARS-CoV-2 Inhibitor against COVID-19. ACS Pharmacol Transl Sci 2022; 5:387-399. [PMID: 35702393 PMCID: PMC9159504 DOI: 10.1021/acsptsci.2c00045] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 01/18/2023]
Abstract
In the global movement to find the appropriate agents to fight the coronavirus disease of 2019 (COVID-19), emetine is one of the strongest anti-SARS-CoV-2 compounds with sub-micromolar EC50 values, identified in several studies and high-throughput screening efforts. The reported anti-SARS-CoV-2 mechanisms indicate the effect of this compound on both virus-based and host-based targets. In addition to having excellent antiviral effects, emetine can relieve COVID-19 patients by reducing inflammation through inhibitory activity against NF-κB by the mechanism of IκBα phosphorylation inhibition; it can also limit the lipopolysaccharide-induced expression of pro-inflammatory cytokines TNFα, IL-1β, and IL-6. Emetine also can well reduce pulmonary arterial hypertension as an important COVID-19 complication by modulating a variety of cellular processes such as the Rho-kinase/CyPA/Bsg signaling pathway. The therapeutic value of emetine for combating COVID-19 was highlighted when in vivo pharmacokinetic studies showed that the concentration of this compound in the lungs increases significantly higher than the EC50 of the drug. Despite its valuable therapeutic effects, emetine has some cardiotoxic effects that limit its use in high doses. However, high therapeutic capabilities make emetine a valuable lead compound that can be used for the design and development of less toxic anti-COVID-19 agents in the future. This Review provides a collection of information on the capabilities of emetine and its potential for the treatment of COVID-19, along with structural analysis which could be used for further research in the future.
Collapse
Affiliation(s)
- Mehdi Valipour
- Department of Medicinal Chemistry,
Faculty of Pharmacy, Mazandaran University
of Medical Sciences, 48175-866 Sari, Iran
| |
Collapse
|
23
|
Rho kinase inhibition ameliorates vascular remodeling and blood pressure elevations in a rat model of apatinib-induced hypertension. J Hypertens 2022; 40:675-684. [PMID: 34862331 PMCID: PMC8901036 DOI: 10.1097/hjh.0000000000003060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Hypertension is one of the major adverse effects of tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factors. However, the mechanism underlying TKIs-induced hypertension remains unclear. Here, we explored the role of the RhoA/Rho kinase (ROCK) signaling pathway in elevation of blood pressure (BP) induced by apatinib, a selective TKI approved in China for treatment of advanced or metastatic gastric cancer. A nonspecific ROCK inhibitor, Y27632, was then combined with apatinib and its efficacy in alleviating apatinib-induced hypertension was evaluated. METHODS Normotensive female Wistar-Kyoto rats were exposed to two different doses of apatinib, or apatinib combined with Y27632, or vehicle for 2 weeks. BP was monitored by a tail-cuff plethysmography system. The mRNA levels and protein expression in the RhoA/ROCK pathway were determined, and vascular remodeling assessed. RESULTS Administration of either a high or low dose of apatinib was associated with a rapid rise in BP, reaching a plateau after 12 days. Apatinib treatment mediated upregulation of RhoA and ROCK II in the mid-aorta, more significant in the high-dose group. However, ROCK I expression showed no statistically significant differences. Furthermore, the mRNA level of GRAF3 decreased dose-dependently. Apatinib administration was also associated with decreased levels of MLCP, and elevated endothelin-1 (ET-1) and collagen I, which were accompanied with increased mid-aortic media. However, treatment with Y27632 attenuated the above changes. CONCLUSION These findings suggest that activation of the RhoA/ROCK signaling pathway could be the underlying mechanism of apatinib-induced hypertension, while ROCK inhibitor have potential therapeutic value.
Collapse
|
24
|
Penumatsa KC, Singhal AA, Warburton RR, Bear MD, Bhedi CD, Nasirova S, Wilson JL, Qi G, Preston IR, Hill NS, Fanburg BL, Kim YB, Toksoz D. Vascular smooth muscle ROCK1 contributes to hypoxia-induced pulmonary hypertension development in mice. Biochem Biophys Res Commun 2022; 604:137-143. [PMID: 35303680 PMCID: PMC9047112 DOI: 10.1016/j.bbrc.2022.02.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
Rho kinase (ROCK) is implicated in the development of pulmonary arterial hypertension (PAH) in which abnormal pulmonary vascular smooth muscle (VSM) contractility and remodeling lead to right heart failure. Pharmacologic ROCK inhibitors block experimental pulmonary hypertension (PH) development in rodents but can have off-target effects and do not distinguish between the two ROCK forms, ROCK1 and ROCK2, encoded by separate genes. An earlier study using gene knock out (KO) in mice indicated that VSM ROCK2 is required for experimental PH development, but the role of ROCK1 is not well understood. Here we investigated the in vivo role of ROCK1 in PH development by generating a VSM-targeted homozygous ROCK1 gene KO mouse strain. Adult control mice exposed to Sugen5416 (Su)/hypoxia treatment to induce PH had significantly increased right ventricular systolic pressures (RVSP) and RV hypertrophy versus normoxic controls. In contrast, Su/hypoxia-exposed VSM ROCK1 KO mice did not exhibit significant RVSP elevation, and RV hypertrophy was blunted. Su/hypoxia-induced pulmonary small vessel muscularization was similarly elevated in both control and VSM ROCK1 KO animals. siRNA-mediated ROCK1 knock-down (KD) in human PAH pulmonary arterial SM cells (PASMC) did not affect cell growth. However, ROCK1 KD led to reduced AKT and MYPT1 signaling in serotonin-treated PAH PASMC. The findings suggest that like VSM ROCK2, VSM ROCK1 actively contributes to PH development, but in distinction acts via nonproliferative pathways to promote hypoxemia, and thus may be a distinct therapeutic target in PH.
Collapse
Affiliation(s)
- Krishna C Penumatsa
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Adit A Singhal
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Rod R Warburton
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Michael D Bear
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Chinmayee D Bhedi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Sabina Nasirova
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Jamie L Wilson
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Guanming Qi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Ioana R Preston
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Nicholas S Hill
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Barry L Fanburg
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave., Boston, MA, 02115, USA
| | - Deniz Toksoz
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA.
| |
Collapse
|
25
|
Regulation of myosin light-chain phosphorylation and its roles in cardiovascular physiology and pathophysiology. Hypertens Res 2022; 45:40-52. [PMID: 34616031 DOI: 10.1038/s41440-021-00733-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/19/2021] [Accepted: 07/08/2021] [Indexed: 01/22/2023]
Abstract
The regulation of muscle contraction is a critical function in the cardiovascular system, and abnormalities may be life-threatening or cause illness. The common basic mechanism in muscle contraction is the interaction between the protein filaments myosin and actin. Although this interaction is primarily regulated by intracellular Ca2+, the primary targets and intracellular signaling pathways differ in vascular smooth muscle and cardiac muscle. Phosphorylation of the myosin regulatory light chain (RLC) is a primary molecular switch for smooth muscle contraction. The equilibrium between phosphorylated and unphosphorylated RLC is dynamically achieved through two enzymes, myosin light chain kinase, a Ca2+-dependent enzyme, and myosin phosphatase, which modifies the Ca2+ sensitivity of contractions. In cardiac muscle, the primary target protein for Ca2+ is troponin C on thin filaments; however, RLC phosphorylation also plays a modulatory role in contraction. This review summarizes recent advances in our understanding of the regulation, physiological function, and pathophysiological involvement of RLC phosphorylation in smooth and cardiac muscles.
Collapse
|
26
|
Li Y, Tai HC, Sladojevic N, Kim HH, Liao JK. Vascular Stiffening Mediated by Rho-Associated Coiled-Coil Containing Kinase Isoforms. J Am Heart Assoc 2021; 10:e022568. [PMID: 34612053 PMCID: PMC8751888 DOI: 10.1161/jaha.121.022568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The pathogenesis of vascular stiffening and hypertension is marked by non‐compliance of vessel wall because of deposition of collagen fibers, loss of elastin fibers, and increased vascular thickening. Rho/Rho‐associated coiled‐coil containing kinases 1 and 2 (ROCK1 and ROCK2) have been shown to regulate cellular contraction and vascular remodeling. However, the role of ROCK isoforms in mediating pathogenesis of vascular stiffening and hypertension is not known. Methods and Results Hemizygous Rock mice (Rock1+/− and Rock2+/−) were used to determine the role of ROCK1 and ROCK2 in age‐related vascular dysfunction. Both ROCK activity and aortic stiffness increased to a greater extent with age in wild‐type mice compared with that of Rock1+/− and Rock2+/− mice. As a model for age‐related vascular stiffening, we administered angiotensin II (500 ng/kg per minute) combined with nitric oxide synthase inhibitor, L‐Nω‐nitroarginine methyl ester (0.5 g/L) for 4 weeks to 12‐week‐old male Rock1+/− and Rock2+/− mice. Similar to advancing age, angiotensin II/L‐Nω‐nitroarginine methyl ester caused increased blood pressure, aortic stiffening, and vascular remodeling, which were attenuated in Rock2+/−, and to a lesser extent, Rock1+/− mice. The reduction of aortic stiffening in Rock2+/− mice was accompanied by decreased collagen deposition, relatively preserved elastin content, and less aortic wall hypertrophy. Indeed, the upregulation of collagen I by transforming growth factor‐β1 or angiotensin II was greatly attenuated in Rock2−/− mouse embryonic fibroblasts. Conclusions These findings indicate that ROCK1 and ROCK2 mediate both age‐related and pharmacologically induced aortic stiffening, and suggest that inhibition of ROCK2, and to a lesser extent ROCK1, may have therapeutic benefits in preventing age‐related vascular stiffening.
Collapse
Affiliation(s)
- Yuxin Li
- Division of Cell Regeneration and Transplantation Department of Functional Morphology Nihon University School of Medicine Tokyo Japan
| | - Haw-Chih Tai
- Section of Cardiology Department of Medicine University of Chicago IL
| | - Nikola Sladojevic
- Section of Cardiology Department of Medicine University of Chicago IL
| | - Hyung-Hwan Kim
- Neurovascular Laboratory Massachusetts General Hospital and Harvard Medical School Boston MA
| | - James K Liao
- Section of Cardiology Department of Medicine University of Chicago IL
| |
Collapse
|
27
|
ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021; 10:cells10071648. [PMID: 34209333 PMCID: PMC8303917 DOI: 10.3390/cells10071648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disease caused by extensive vascular remodeling in the lungs, which ultimately leads to death in consequence of right ventricle (RV) failure. While current drugs for PH therapy address the sustained vasoconstriction, no agent effectively targets vascular cell proliferation and tissue inflammation. Rho-associated protein kinases (ROCKs) emerged in the last few decades as promising targets for PH therapy, since ROCK inhibitors demonstrated significant anti-remodeling and anti-inflammatory effects. In this review, current aspects of ROCK inhibition therapy are discussed in relation to the treatment of PH and RV dysfunction, from cell biology to preclinical and clinical studies.
Collapse
|
28
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
29
|
Cheng C, Xu DL, Liu XB, Bi SJ, Zhang J. MicroRNA-145-5p inhibits hypoxia/reoxygenation-induced apoptosis in H9c2 cardiomyocytes by targeting ROCK1. Exp Ther Med 2021; 22:796. [PMID: 34093752 PMCID: PMC8170661 DOI: 10.3892/etm.2021.10228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that microRNAs (miRs) play critical roles in the pathological and physiological processes associated with myocardial ischemia reperfusion (I/R). miR-145 has been extensively studied in the cardiovascular system; however, the role of miR-145 in myocardial I/R remains unclear. Therefore, the present study aimed to investigate the role and mechanism of miR-145-5p in myocardial I/R by establishing a hypoxia/reoxygenation (H/R) model using H9c2 cardiomyocytes. The expression of miR-145-5p was regulated by transfection and the potential target of miR-145-5p was identified. In addition, apoptosis of the cardiomyocytes was evaluated using flow cytometry and the detection of cleaved caspase-3 by western blotting. The results revealed that miR-145-5p expression was decreased while cell apoptosis and Rho-associated coiled-coil-containing kinase 1 (ROCK1) expression were increased in H/R-stimulated H9c2 cardiomyocytes. The upregulation of miR-145-5p reduced apoptosis and the expression of ROCK1 in H/R-stimulated H9c2 cardiomyocytes. Furthermore, the overexpression of ROCK1 significantly attenuated the miR-145-5p-induced reduction of apoptosis following H/R. In conclusion, the present study indicates that the overexpression of miR-145-5p inhibits H/R-induced cardiomyocyte apoptosis by targeting ROCK1.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Dong-Ling Xu
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiao-Bo Liu
- Shandong Blood Center, Jinan, Shandong 250012, P.R. China
| | - Shao-Jie Bi
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Juan Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
30
|
Implication of EZH2 in the Pro-Proliferative and Apoptosis-Resistant Phenotype of Pulmonary Artery Smooth Muscle Cells in PAH: A Transcriptomic and Proteomic Approach. Int J Mol Sci 2021; 22:ijms22062957. [PMID: 33803922 PMCID: PMC7999120 DOI: 10.3390/ijms22062957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by a sustained elevation of pulmonary artery (PA) pressure, right ventricular failure, and premature death. Enhanced proliferation and resistance to apoptosis (as seen in cancer cells) of PA smooth muscle cells (PASMCs) is a major pathological hallmark contributing to pulmonary vascular remodeling in PAH, for which current therapies have only limited effects. Emerging evidence points toward a critical role for Enhancer of Zeste Homolog 2 (EZH2) in cancer cell proliferation and survival. However, its role in PAH remains largely unknown. The aim of this study was to determine whether EZH2 represents a new factor critically involved in the abnormal phenotype of PAH-PASMCs. We found that EZH2 is overexpressed in human lung tissues and isolated PASMCs from PAH patients compared to controls as well as in two animal models mimicking the disease. Through loss- and gain-of-function approaches, we showed that EZH2 promotes PAH-PASMC proliferation and survival. By combining quantitative transcriptomic and proteomic approaches in PAH-PASMCs subjected or not to EZH2 knockdown, we found that inhibition of EZH2 downregulates many factors involved in cell-cycle progression, including E2F targets, and contributes to maintain energy production. Notably, we found that EZH2 promotes expression of several nuclear-encoded components of the mitochondrial translation machinery and tricarboxylic acid cycle genes. Overall, this study provides evidence that, by overexpressing EZH2, PAH-PASMCs remove the physiological breaks that normally restrain their proliferation and susceptibility to apoptosis and suggests that EZH2 or downstream factors may serve as therapeutic targets to combat pulmonary vascular remodeling.
Collapse
|
31
|
Zhu L, Liu F, Hao Q, Feng T, Chen Z, Luo S, Xiao R, Sun M, Zhang T, Fan X, Zeng X, He J, Yuan P, Liu J, Ruiz M, Dupuis J, Hu Q. Dietary Geranylgeranyl Pyrophosphate Counteracts the Benefits of Statin Therapy in Experimental Pulmonary Hypertension. Circulation 2021; 143:1775-1792. [PMID: 33660517 DOI: 10.1161/circulationaha.120.046542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The mevalonate pathway generates endogenous cholesterol and intermediates including geranylgeranyl pyrophosphate (GGPP). By reducing GGPP production, statins exert pleiotropic or cholesterol-independent effects. The potential regulation of GGPP homeostasis through dietary intake and the interaction with concomitant statin therapy is unknown. METHODS We developed a sensitive high-pressure liquid chromatography technique to quantify dietary GGPP and conducted proteomics, qualitative real-time polymerase chain reaction screening, and Western blot to determine signaling cascades, gene expression, protein-protein interaction, and protein membrane trafficking in wild-type and transgenic rats. RESULTS GGPP contents were highly variable depending on food source that differentially regulated blood GGPP levels in rats. Diets containing intermediate and high GGPP reduced or abolished the effects of statins in rats with hypoxia- and monocrotaline-induced pulmonary hypertension: this was rescuable by methyl-allylthiosulfinate and methyl-allylthiosulfinate-rich garlic extracts. In human pulmonary artery smooth muscle cells treated with statins, hypoxia activated RhoA in an extracellular GGPP-dependent manner. Hypoxia-induced ROCK2 (Rho associated coiled-coil containing protein kinase 2)/Rab10 (Ras-related protein rab-10) signaling was prevented by statin and recovered by exogenous GGPP. The hypoxia-activated RhoA/ROCK2 pathway in rat and human pulmonary artery smooth muscle cells upregulated the expression of Ca2+-sensing receptor (CaSR) and HIMF (hypoxia-induced mitogenic factor), a mechanism attenuated by statin treatment and regained with exogenous GGPP. Rab10 knockdown almost abrogated hypoxia-promoted CaSR membrane trafficking, a process diminished by statin and resumed by exogenous GGPP. Hypoxia-induced pulmonary hypertension was reduced in rats with CaSR mutated at the binding motif of HIMF and the interaction between dietary GGPP and statin efficiency was abolished. In humans fed a high GGPP diet, blood GGPP levels were increased. This abolished statin-lowering effects on plasma GGPP, and also on hypoxia-enhanced RhoA activity of blood monocytes that was rescued by garlic extracts. CONCLUSIONS There is important dietary regulation of GGPP levels that interferes with the effects of statin therapy in experimental pulmonary hypertension. These observations rely on a key and central role of RhoA-ROCK2 cascade activation and Rab10-faciliated CaSR membrane trafficking with subsequent overexpression and binding of HIMF to CaSR. These findings warrant clinical investigation for the treatment of pulmonary hypertension and perhaps other diseases by combining statin with garlic-derived methyl-allylthiosulfinate or garlic extracts and thus circumventing dietary GGPP variations.
Collapse
Affiliation(s)
- Liping Zhu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangbo Liu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Hao
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Feng
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeshuai Chen
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengquan Luo
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xiao
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxiang Sun
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhang
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohang Fan
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqin Zeng
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianguo He
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.H.)
| | - Ping Yuan
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, China (P.Y., J.L.)
| | - Jinming Liu
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, China (P.Y., J.L.)
| | - Matthieu Ruiz
- Departments of Nutrition (M.R.), Université de Montréal, Canada.,Montreal Heart Institute Research Center, Canada (M.R., J.D.)
| | - Jocelyn Dupuis
- Medicine (J.D.), Université de Montréal, Canada.,Montreal Heart Institute Research Center, Canada (M.R., J.D.)
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Satoh K. Drug discovery focused on novel pathogenic proteins for pulmonary arterial hypertension. J Cardiol 2021; 78:1-11. [PMID: 33563508 DOI: 10.1016/j.jjcc.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 10/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease in which the wall thickening and narrowing of pulmonary microvessels progress due to complicated interactions among processes such as endothelial dysfunction, the proliferation of pulmonary artery smooth muscle cells (PASMCs) and adventitial fibrocytes, and inflammatory cell infiltration. Early diagnosis of patients with PAH is difficult and lung transplantation is the only last choice to save severely ill patients. However, the number of donors is limited. Many patients with PAH show rapid progression and a high degree of pulmonary arterial remodeling characterized by the abnormal proliferation of PASMCs, which makes treatment difficult even with multidrug therapy comprising pulmonary vasodilators. Thus, it is important to develop novel therapy targeting factors other than vasodilation, such as PASMC proliferation. In the development of PAH, inflammation and oxidative stress are deeply involved in its pathogenesis. Excessive proliferation and apoptosis resistance in PASMCs are key mechanisms underlying PAH. Based on those characteristics, we recently screened novel pathogenic proteins and have performed drug discovery targeting those proteins. To confirm the clinical significance of this, we used patient-derived blood samples to evaluate biomarker potential for diagnosis and prognosis. Moreover, we conducted high throughput screening and found several inhibitors of the pathogenic proteins. In this review, we introduce the recent progress on basic and clinical PAH research, focusing on the screening of pathogenic proteins and drug discovery.
Collapse
Affiliation(s)
- Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.
| |
Collapse
|
33
|
Bai L, Kee HJ, Choi SY, Seok YM, Kim GR, Kee SJ, Kook H, Jeong MH. HDAC5 inhibition reduces angiotensin II-induced vascular contraction, hypertrophy, and oxidative stress in a mouse model. Biomed Pharmacother 2021; 134:111162. [PMID: 33360932 DOI: 10.1016/j.biopha.2020.111162] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 01/02/2023] Open
Abstract
Non-specific histone deacetylase (HDAC) inhibition reduces high blood pressure in essential hypertensive animal models. However, the exact HDAC isoforms that play a critical role in controlling hypertension are not known. Here, we investigated the role of HDAC5 in vascular contraction, hypertrophy, and oxidative stress in the context of angiotensin II (Ang II)-induced hypertension. Genetic deletion of HDAC5 and treatment with class IIa HDAC inhibitors (TMP269 and TMP195) prevented Ang II-induced increases in blood pressure and arterial wall thickness. Hdac5-knockout mice were also resistant to the thromboxane A2 agonist (U46619)-induced vascular contractile response. Furthermore, the expression of Rho-associated protein kinase (ROCK) 2 was downregulated in the aortas of Ang II-treated Hdac5-knockout mice. Knockdown of HDAC5, RhoA, or ROCK2 reduced collagen gel contraction, whereas silencing of ROCK1 increased it. VSMC hypertrophy reduced on knocking down HDAC5, ROCK1, and ROCK2. Here we showed that genetic deletion of HDAC5 and pharmacological inhibition of class IIa HDACs ameliorated Ang II-induced ROS generation. Moreover, ROCK1 and ROCK2, the downstream targets of HDAC5, influenced ROS generation. The relative protein levels of HDAC5, ROCK1, and ROCK2 were increased both in the cytoplasm and nuclear fraction in response to Ang II stimulation in vascular smooth muscle cells. Inhibition of HDAC5 expression or activity reduced vascular hypertrophy, vasoconstriction, and oxidative stress in the Ang II-induced hypertension model. These findings indicate that HDAC5 may serve as a potential target in the treatment of hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Antihypertensive Agents/pharmacology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/physiopathology
- Arterial Pressure/drug effects
- Benzamides/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylases/deficiency
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Hypertension/chemically induced
- Hypertension/enzymology
- Hypertension/physiopathology
- Hypertension/prevention & control
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Oxadiazoles/pharmacology
- Oxidative Stress/drug effects
- Vascular Remodeling/drug effects
- Vasoconstriction/drug effects
- rho-Associated Kinases/genetics
- rho-Associated Kinases/metabolism
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
- Mice
Collapse
Affiliation(s)
- Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea; Hypertension and Heart Failure, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea; Hypertension and Heart Failure, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.
| | - Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea; Hypertension and Heart Failure, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Young Mi Seok
- National Development Institute of Korean Medicine, Hwarang-ro, Gyeongsan-si, Gyeongsangbuk-do 38540, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea; Hypertension and Heart Failure, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University, Medical School and Hospital, Gwangju 61469, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanam-do 58128, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea; Hypertension and Heart Failure, Chonnam National University Hospital, Gwangju 61469, Republic of Korea.
| |
Collapse
|
34
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
35
|
Yamamura A, Nayeem MJ, Sato M. The Rho kinase 2 (ROCK2)-specific inhibitor KD025 ameliorates the development of pulmonary arterial hypertension. Biochem Biophys Res Commun 2021; 534:795-801. [PMID: 33160621 DOI: 10.1016/j.bbrc.2020.10.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease that is characterized by the irreversible remodeling of the pulmonary artery. Although several PAH drugs have been developed, additional drugs are needed. Rho kinases (ROCKs) are involved in the pathogenesis of PAH, and thus, their inhibitors may prevent the development of PAH. However, the therapeutic benefits of ROCK isoform-specific inhibitors for PAH remain largely unknown. The in vitro and in vivo effects of the ROCK2-specific inhibitor, KD025, were examined herein using pulmonary arterial smooth muscle cells (PASMCs) from idiopathic pulmonary arterial hypertension (IPAH) patients and monocrotaline (MCT)-induced pulmonary hypertensive (PH) rats. The expression of ROCK1 was similar between normal- and IPAH-PASMCs, whereas that of ROCK2 was markedly higher in IPAH-PASMCs than in normal-PASMCs. KD025 inhibited the accelerated proliferation of IPAH-PASMCs in a concentration-dependent manner (IC50 = 289 nM). Accelerated proliferation was also reduced by the siRNA knockdown of ROCK2. In MCT-PH rats, the expression of ROCK2 was up-regulated in PASMCs. Elevated right ventricular systolic pressure in MCT-PH rats was attenuated by KD025 (1 mg/kg/day). These results strongly suggest that enhanced ROCK2 signaling is involved in the pathogenic mechanism underlying the development of PAH, including accelerated PASMC proliferation and vascular remodeling in patients with PAH. Therefore, ROCK2 may be a novel therapeutic target for the treatment of PAH.
Collapse
Affiliation(s)
- Aya Yamamura
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| | - Md Junayed Nayeem
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
36
|
Lee JY, Stevens RP, Kash M, Zhou C, Koloteva A, Renema P, Paudel SS, Stevens T. KD025 Shifts Pulmonary Endothelial Cell Bioenergetics and Decreases Baseline Lung Permeability. Am J Respir Cell Mol Biol 2020; 63:519-530. [PMID: 32628869 PMCID: PMC7528923 DOI: 10.1165/rcmb.2019-0435oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
KD025 is a ROCK2 inhibitor currently being tested in clinical trials for the treatment of fibrotic lung diseases. The therapeutic effects of KD025 are partly due to its inhibition of profibrotic pathways and fat metabolism. However, whether KD025 affects pulmonary microvascular endothelial cell (PMVEC) function is unknown, despite evidence that alveolar-capillary membrane disruption constitutes major causes of death in fibrotic lung diseases. We hypothesized that KD025 regulates PMVEC metabolism, pH, migration, and survival, a series of interrelated functional characteristics that determine pulmonary barrier integrity. We used PMVECs isolated from Sprague Dawley rats. KD025 dose-dependently decreased lactate production and glucose consumption. The inhibitory effect of KD025 was more potent compared with other metabolic modifiers, including 2-deoxy-glucose, extracellular acidosis, dichloroacetate, and remogliflozin. Interestingly, KD025 increased oxidative phosphorylation, whereas 2-deoxy-glucose did not. KD025 also decreased intracellular pH and induced a compensatory increase in anion exchanger 2. KD025 inhibited PMVEC migration, but fasudil (nonspecific ROCK inhibitor) did not. We tested endothelial permeability in vivo using Evans Blue dye in the bleomycin pulmonary fibrosis model. Baseline permeability was decreased in KD025-treated animals independent of bleomycin treatment. Under hypoxia, KD025 increased PMVEC necrosis as indicated by increased lactate dehydrogenase release and propidium iodide uptake and decreased ATP; it did not affect Annexin V binding. ROCK2 knockdown had no effect on PMVEC metabolism, pH, and migration, but it increased nonapoptotic caspase-3 activity. Together, we report that KD025 promotes oxidative phosphorylation; decreases glycolysis, intracellular pH, and migration; and strengthens pulmonary barrier integrity in a ROCK2-independent manner.
Collapse
Affiliation(s)
- Ji Young Lee
- Department of Physiology and Cell Biology
- Department of Internal Medicine
- Division of Pulmonary and Critical Care Medicine
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Reece P. Stevens
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Mary Kash
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Chun Zhou
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Anna Koloteva
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Phoibe Renema
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Sunita S. Paudel
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology
- Department of Internal Medicine
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| |
Collapse
|
37
|
Matoba K, Takeda Y, Nagai Y, Sekiguchi K, Yokota T, Utsunomiya K, Nishimura R. The Physiology, Pathology, and Therapeutic Interventions for ROCK Isoforms in Diabetic Kidney Disease. Front Pharmacol 2020; 11:585633. [PMID: 33101039 PMCID: PMC7545791 DOI: 10.3389/fphar.2020.585633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023] Open
Abstract
Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine/threonine kinase that was originally identified as RhoA interacting protein. A diverse array of cellular functions, including migration, proliferation, and phenotypic modulation, are orchestrated by ROCK through a mechanism involving cytoskeletal rearrangement. Mammalian cells express two ROCK isoforms: ROCK1 (Rho-kinase β/ROKβ) and ROCK2 (Rho-kinase α/ROKα). While both isoforms have structural similarities and are widely expressed across multiple tissues, investigations in gene knockout animals and cell-based studies have revealed distinct functions of ROCK1 and ROCK2. With respect to the kidney, inhibiting ROCK activity has proven effective for the preventing diabetic kidney disease (DKD) in both type 1 and type 2 diabetic rodent models. However, despite significant progress in the understanding of the renal ROCK biology over the past decade, the pathogenic roles of the ROCK isoforms is only beginning to be elucidated. Recent studies have demonstrated the involvement of renal ROCK1 in mitochondrial dynamics and cellular transdifferentiation, whereas ROCK2 activation leads to inflammation, fibrosis, and cell death in the diabetic kidney. This review provides a conceptual framework for dissecting the molecular underpinnings of ROCK-driven renal injury, focusing on the differences between ROCK1 and ROCK2.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kensuke Sekiguchi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Takeda Y, Matoba K, Sekiguchi K, Nagai Y, Yokota T, Utsunomiya K, Nishimura R. Endothelial Dysfunction in Diabetes. Biomedicines 2020; 8:E182. [PMID: 32610588 PMCID: PMC7400447 DOI: 10.3390/biomedicines8070182] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes is a worldwide health issue closely associated with cardiovascular events. Given the pandemic of obesity, the identification of the basic underpinnings of vascular disease is strongly needed. Emerging evidence has suggested that endothelial dysfunction is a critical step in the progression of atherosclerosis. However, how diabetes affects the endothelium is poorly understood. Experimental and clinical studies have illuminated the tight link between insulin resistance and endothelial dysfunction. In addition, macrophage polarization from M2 towards M1 contributes to the process of endothelial damage. The possibility that novel classes of anti-hyperglycemic agents exert beneficial effects on the endothelial function and macrophage polarization has been raised. In this review, we discuss the current status of knowledge regarding the pathological significance of insulin signaling in endothelium. Finally, we summarize recent therapeutic strategies against endothelial dysfunction with an emphasis on macrophage polarity.
Collapse
Affiliation(s)
- Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| | - Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| | - Kensuke Sekiguchi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan;
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan; (Y.T.); (K.S.); (Y.N.); (T.Y.); (R.N.)
| |
Collapse
|
39
|
Lei S, Peng F, Li ML, Duan WB, Peng CQ, Wu SJ. LncRNA-SMILR modulates RhoA/ROCK signaling by targeting miR-141 to regulate vascular remodeling in pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2020; 319:H377-H391. [PMID: 32559140 DOI: 10.1152/ajpheart.00717.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal progressive disease characterized by an increased blood pressure in the pulmonary arteries. RhoA/Rho-kinase (RhoA/ROCK) signaling activation is often associated with PAH. The purpose of this study is to investigate the role and mechanisms of long noncoding RNA (lncRNA) smooth muscle-induced lncRNA (SMILR) to activate the RhoA/ROCK pathway in PAH. SMILR, microRNA-141 (miR-141), and RhoA were identified by qRT-PCR in PAH patients' serum. 3-(4,5-Dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), wound-healing assay, cell counting kit-8 (CCK-8) assay, and flow cytometry were performed to determine cell viability, migration, proliferation, and cell cycle in human pulmonary arterial smooth muscle cells (hPASMCs) and primary PASMCs from PAH patients. We also performed bioinformatical prediction, luciferase reporter assay, and RNA-binding protein immunoprecipitation (RIP) to assess the interaction among SMILR, miR-141, and RhoA. The RhoA/ROCK pathway and proliferation-related proteins were measured by Western blotting. Finally, we introduced the small hairpin (sh)SMILR to monocrotaline-induced PAH rat model and used the hemodynamic measurement, qRT-PCR, and immunohistochemistry to examine the therapeutic effects of shSMILR. SMILR and RhoA expression were upregulated, while miR-141 expression was downregulated in PAH patients. SMILR directly interacted with miR-141 and negatively regulated its expression. Knockdown of SMILR suppressed PASMC proliferation and migration induced by hypoxia. Furthermore, overexpression of miR-141 could inhibit the RhoA/ROCK pathway by binding to RhoA, thereby repressing cell proliferation-related signals. Knockdown of SMILR significantly inhibited the Rho/ROCK activation and vascular remodeling in monocrotaline-induced rats. Knockdown of SMILR effectively elevated miR-141 expression and in turn inhibited the RhoA/ROCK pathway to regulate vascular remodeling and reduce blood pressure in PAH.NEW & NOTEWORTHY Smooth muscle enriched long noncoding RNA (SMILR), as a long noncoding RNA (lncRNA), was increased in pulmonary arterial hypertension (PAH) patients and in vitro and in vivo models. SMILR activated RhoA/ROCK signaling by targeting miR-141 to disinhibit its downstream target RhoA. SMILR knockdown or miR-141 overexpression inhibited hypoxia-induced cell proliferation and migration via repressing RhoA/ROCK signaling in pulmonary arterial smooth muscle cells (PASMCs), which was confirmed in vivo experiments that knockdown of SMILR inhibited vascular remodeling and alleviated PAH in rats. SMILR may be a promising and novel therapeutic target for the treatment and drug development of PAH.
Collapse
Affiliation(s)
- Si Lei
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Fei Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Mei-Lei Li
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Wen-Bing Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Cai-Qin Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Shang-Jie Wu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| |
Collapse
|
40
|
Sharma P, Roy K. ROCK-2-selective targeting and its therapeutic outcomes. Drug Discov Today 2020; 25:446-455. [DOI: 10.1016/j.drudis.2019.11.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/08/2019] [Accepted: 11/30/2019] [Indexed: 01/21/2023]
|
41
|
Li C, Peng G, Long J, Xiao P, Zeng X, Yang H. Protective effects of resveratrol and SR1001 on hypoxia-induced pulmonary hypertension in rats. Clin Exp Hypertens 2020; 42:519-526. [PMID: 31973589 DOI: 10.1080/10641963.2020.1714643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hypoxic pulmonary hypertension (HPH) is a fatal disease with limited therapeutic strategies. Combination therapy is regarded as the standard of care in PH and becoming widely used in clinical practice. However, many PH patients treated with combinations of available clinical drugs still have a poor prognosis. Therefore, identifying innovative therapeutic strategies is essential for PH. This study is designed to examine the effects of combined prevention with resveratrol and SR1001 on HPH in rats. The effects of combined prevention with resveratrol and SR1001 and each mono-prevention on the development of HPH, Th17 cells differentiation, expression of guanine nucleotide exchange factor-H1 (GEF-H1), Ras homolog gene family member A (RhoA) and Phosphorylated myosin phosphatase target subunit (MYPT1) were examined. HPH and RV hypertrophy occurred in rats exposed to hypoxia. Compared with normoxia group, the hypoxia group showed significantly increased ratio of Th17 cells. After treatment with resveratrol, HPH rats showed an obvious reduction of Th17 cells. SR1001 significantly reduced the increased p-MYPY1, RhoA, and GEF-H1 expression in the hypoxic rats. The mono-prevention with resveratrol or SR1001 significantly inhibited the Th17 cells differentiation, p-STAT3, p-MYPY1, RhoA, and GEF-H1 protein expression, which was further inhibited by their combination prevention. The combination of resveratrol and SR1001 has a synergistic interaction, suggesting that combined use of these pharmacological targets may be an alternative to exert further beneficial effects on HPH.
Collapse
Affiliation(s)
- Cheng Li
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| | - Ganlin Peng
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| | - Jing Long
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| | - Pan Xiao
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| | - Xiaoyuan Zeng
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| | - Hongzhong Yang
- Department of Respiratory and Critical Care Medicine, Changsha Central Hospital, University of South China , Changsha, Hunan, P.R. China
| |
Collapse
|
42
|
Li L, Jing L, Zhao J, Lv J, Yang W, Li W, Zhou L. Valsartan inhibits RhoA-ROCK2-MYL pathway in rat model of alcoholic cardiomyopathy. Exp Ther Med 2019; 18:4313-4321. [PMID: 31777538 PMCID: PMC6862588 DOI: 10.3892/etm.2019.8079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/01/2018] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate variations in the Ras homolog gene family, member A (RhoA)-Rho-associated protein kinase 2 (ROCK2)-myosin light chain (MYL) pathway in a rat model of alcoholic cardiomyopathy (ACM) and the role of angiotensin-converting enzyme inhibitor drugs. Rat models of ACM were established via alcoholic gavage + free access to alcohol. The structural and functional changes of the heart were analyzed by hematoxylin-eosin staining, Masson's trichrome staining, immunohistochemistry staining, western blotting and fluorescence quantitative polymerase chain reaction. A total of 16 weeks later, a decreased ejection fraction and left ventricular fractional shortening in the alcohol group compared with the control group were demonstrated resulting in an increased left ventricular end diastolic diameter. These adverse effects were ameliorated following treatment with valsartan. In addition, the alcohol group revealed a disorganized arrangement of myocardial filaments, which was improved upon treatment with valsartan. RhoA and ROCK2 protein expression significantly increased in myocardial cells in the alcohol compared with the control group. Following drug intervention with valsartan, expression of RhoA and ROCK2 proteins were inhibited in the alcohol group. Furthermore, significantly elevated RhoA and ROCK2 and decreased MYL protein and mRNA expression in the alcohol group was demonstrated compared with the control group. Administration of valsartan reversed the expression profile of RhoA, ROCK and MYL in ACM. Expression of RhoA and ROCK were elevated with downregulation of MYL resulting in heart failure. However, the angiotensin receptor antagonist diminished the expression of RhoA and ROCK and enhanced the expression of MYL. The results of the present study suggest a curative effect of valsartan in ACM.
Collapse
Affiliation(s)
- Luyifei Li
- Department of Internal Critical Illness, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Ling Jing
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiyi Zhao
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jiachen Lv
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Wen Yang
- Department of The First Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Weimin Li
- Department of The Fifth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Lijun Zhou
- Department of The Fourth Cardiovascular, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
43
|
Siddique MAH, Satoh K, Kurosawa R, Kikuchi N, Elias-Al-Mamun M, Omura J, Satoh T, Nogi M, Sunamura S, Miyata S, Ueda H, Tokuyama H, Shimokawa H. Identification of Emetine as a Therapeutic Agent for Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:2367-2385. [DOI: 10.1161/atvbaha.119.313309] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective:
Excessive proliferation and apoptosis resistance are special characteristics of pulmonary artery smooth muscle cells (PASMCs) in pulmonary arterial hypertension (PAH). However, the drugs in clinical use for PAH target vascular dilatation, which do not exert adequate effects in patients with advanced PAH. Here, we report a novel therapeutic effect of emetine, a principal alkaloid extracted from the root of ipecac clinically used as an emetic and antiprotozoal drug.
Approach and Results:
We performed stepwise screenings for 5562 compounds from original library. First, we performed high-throughput screening with PASMCs from patients with PAH (PAH-PASMCs) and found 80 compounds that effectively inhibited proliferation. Second, we performed the repeatability and counter assay. Finally, we performed a concentration-dependent assay and found that emetine inhibits PAH-PASMC proliferation. Interestingly, emetine significantly reduced protein levels of HIFs (hypoxia-inducible factors; HIF-1α and HIF-2α) and downstream PDK1 (pyruvate dehydrogenase kinase 1). Moreover, emetine significantly reduced the protein levels of RhoA (Ras homolog gene family, member A), Rho-kinases (ROCK1 and ROCK2 [rho-associated coiled-coil containing protein kinases 1 and 2]), and their downstream CyPA (cyclophilin A), and Bsg (basigin) in PAH-PASMCs. Consistently, emetine treatment significantly reduced the secretion of cytokines/chemokines and growth factors from PAH-PASMCs. Interestingly, emetine reduced protein levels of BRD4 (bromodomain-containing protein 4) and downstream survivin, both of which are involved in many cellular functions, such as cell cycle, apoptosis, and inflammation. Finally, emetine treatment ameliorated pulmonary hypertension in 2 experimental rat models, accompanied by reduced inflammatory changes in the lungs and recovered right ventricular functions.
Conclusions:
Emetine is an old but novel drug for PAH that reduces excessive proliferation of PAH-PASMCs and improves right ventricular functions.
Collapse
Affiliation(s)
- Mohammad Abdul Hai Siddique
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Ryo Kurosawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Md. Elias-Al-Mamun
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Junichi Omura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Taijyu Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Masamichi Nogi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| | - Hirofumi Ueda
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.U., H.T.)
| | - Hidetoshi Tokuyama
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.U., H.T.)
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (M.A.H.S., K.S., R.K., N.K., M.E.-A.-M., J.O., T.S., M.N., S.S., S.M., H.S.)
| |
Collapse
|
44
|
Zhao X, Nedvetsky P, Stanchi F, Vion AC, Popp O, Zühlke K, Dittmar G, Klussmann E, Gerhardt H. Endothelial PKA activity regulates angiogenesis by limiting autophagy through phosphorylation of ATG16L1. eLife 2019; 8:e46380. [PMID: 31580256 PMCID: PMC6797479 DOI: 10.7554/elife.46380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 10/01/2019] [Indexed: 12/15/2022] Open
Abstract
The cAMP-dependent protein kinase A (PKA) regulates various cellular functions in health and disease. In endothelial cells PKA activity promotes vessel maturation and limits tip cell formation. Here, we used a chemical genetic screen to identify endothelial-specific direct substrates of PKA in human umbilical vein endothelial cells (HUVEC) that may mediate these effects. Amongst several candidates, we identified ATG16L1, a regulator of autophagy, as novel target of PKA. Biochemical validation, mass spectrometry and peptide spot arrays revealed that PKA phosphorylates ATG16L1α at Ser268 and ATG16L1β at Ser269, driving phosphorylation-dependent degradation of ATG16L1 protein. Reducing PKA activity increased ATG16L1 protein levels and endothelial autophagy. Mouse in vivo genetics and pharmacological experiments demonstrated that autophagy inhibition partially rescues vascular hypersprouting caused by PKA deficiency. Together these results indicate that endothelial PKA activity mediates a critical switch from active sprouting to quiescence in part through phosphorylation of ATG16L1, which in turn reduces endothelial autophagy.
Collapse
Affiliation(s)
- Xiaocheng Zhao
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
| | - Pavel Nedvetsky
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
- Medical Cell Biology, Medical Clinic DUniversity Hospital MünsterMünsterGermany
| | - Fabio Stanchi
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
| | - Anne-Clemence Vion
- Integrative Vascular Biology LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- INSERM UMR-970, Paris Cardiovascular Research CenterParis Descartes UniversityParisFrance
| | - Oliver Popp
- ProteomicsMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Kerstin Zühlke
- Anchored Signaling LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Gunnar Dittmar
- ProteomicsMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- CRP Santé · Department of OncologyLIH Luxembourg Institute of HealthLuxembourgLuxembourg
| | - Enno Klussmann
- Anchored Signaling LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- DZHK (German Center for Cardiovascular Research)BerlinGermany
| | - Holger Gerhardt
- Vascular Patterning Laboratory, Center for Cancer BiologyVIBLeuvenBelgium
- Vascular Patterning Laboratory, Center for Cancer Biology, Department of OncologyVIBLeuvenBelgium
- Integrative Vascular Biology LabMax-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- DZHK (German Center for Cardiovascular Research)BerlinGermany
- Berlin Institute of Health (BIH)BerlinGermany
| |
Collapse
|
45
|
Satoh K, Shimokawa H. Recent Advances in the Development of Cardiovascular Biomarkers. Arterioscler Thromb Vasc Biol 2019; 38:e61-e70. [PMID: 29695533 DOI: 10.1161/atvbaha.118.310226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
46
|
Kurosawa R, Satoh K, Kikuchi N, Kikuchi H, Saigusa D, Al-Mamun ME, Siddique MAH, Omura J, Satoh T, Sunamura S, Nogi M, Numano K, Miyata S, Uruno A, Kano K, Matsumoto Y, Doi T, Aoki J, Oshima Y, Yamamoto M, Shimokawa H. Identification of Celastramycin as a Novel Therapeutic Agent for Pulmonary Arterial Hypertension. Circ Res 2019; 125:309-327. [PMID: 31195886 DOI: 10.1161/circresaha.119.315229] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is characterized by enhanced proliferation of pulmonary artery smooth muscle cells (PASMCs) accompanying increased production of inflammatory factors and adaptation of the mitochondrial metabolism to a hyperproliferative state. However, all the drugs in clinical use target pulmonary vascular dilatation, which may not be effective for patients with advanced PAH. OBJECTIVE We aimed to discover a novel drug for PAH that inhibits PASMC proliferation. METHODS AND RESULTS We screened 5562 compounds from original library using high-throughput screening system to discover compounds which inhibit proliferation of PASMCs from patients with PAH (PAH-PASMCs). We found that celastramycin, a benzoyl pyrrole-type compound originally found in a bacteria extract, inhibited the proliferation of PAH-PASMCs in a dose-dependent manner with relatively small effects on PASMCs from healthy donors. Then, we made 25 analogs of celastramycin and selected the lead compound, which significantly inhibited cell proliferation of PAH-PASMCs and reduced cytosolic reactive oxygen species levels. Mechanistic analysis demonstrated that celastramycin reduced the protein levels of HIF-1α (hypoxia-inducible factor 1α), which impairs aerobic metabolism, and κB (nuclear factor-κB), which induces proinflammatory signals, in PAH-PASMCs, leading to reduced secretion of inflammatory cytokine. Importantly, celastramycin treatment reduced reactive oxygen species levels in PAH-PASMCs with increased protein levels of Nrf2 (nuclear factor erythroid 2-related factor 2), a master regulator of cellular response against oxidative stress. Furthermore, celastramycin treatment improved mitochondrial energy metabolism with recovered mitochondrial network formation in PAH-PASMCs. Moreover, these celastramycin-mediated effects were regulated by ZFC3H1 (zinc finger C3H1 domain-containing protein), a binding partner of celastramycin. Finally, celastramycin treatment ameliorated pulmonary hypertension in 3 experimental animal models, accompanied by reduced inflammatory changes in the lungs. CONCLUSIONS These results indicate that celastramycin ameliorates pulmonary hypertension, reducing excessive proliferation of PAH-PASMCs with less inflammation and reactive oxygen species levels, and recovered mitochondrial energy metabolism. Thus, celastramycin is a novel drug for PAH that targets antiproliferative effects on PAH-PASMCs.
Collapse
Affiliation(s)
- Ryo Kurosawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan (R.K.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Haruhisa Kikuchi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Md Elias Al-Mamun
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Mohammad A H Siddique
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Junichi Omura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Taijyu Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Masamichi Nogi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Kazuhiko Numano
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Kuniyuki Kano
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yotaro Matsumoto
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Takayuki Doi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Junken Aoki
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yoshiteru Oshima
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| |
Collapse
|
47
|
ROCK2 Regulates Monocyte Migration and Cell to Cell Adhesion in Vascular Endothelial Cells. Int J Mol Sci 2019; 20:ijms20061331. [PMID: 30884801 PMCID: PMC6471293 DOI: 10.3390/ijms20061331] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
The small GTPase Rho and its downstream effector, Rho-kinase (ROCK), regulate various cellular functions, including organization of the actin cytoskeleton, cell adhesion and migration. A pro-inflammatory lipid mediator, lysophosphatidic acid (LPA), is a potent activator of the Rho/ROCK signalling pathway and has been shown to induce the expression of chemokines and cell adhesion molecules (CAMs). In the present study, we aimed to elucidate the precise mechanism by which ROCK regulates LPA-induced expressions and functions of chemokines and CAMs. We observed that ROCK blockade reduced LPA-induced phosphorylation of IκBα and inhibited NF-κB RelA/p65 phosphorylation, leading to attenuation of RelA/p65 nuclear translocation. Furthermore, small interfering RNA-mediated ROCK isoform knockdown experiments revealed that LPA induces the expression of monocyte chemoattractant protein-1 (MCP-1) and E-selectin via ROCK2 in human aortic endothelial cells (HAECs). Importantly, we found that ROCK2 but not ROCK1 controls LPA-induced monocytic migration and monocyte adhesion toward endothelial cells. These findings demonstrate that ROCK2 is a key regulator of endothelial inflammation. We conclude that targeting endothelial ROCK2 is potentially effective in attenuation of atherosclerosis.
Collapse
|
48
|
Kanazawa M, Matsumoto Y, Takahashi K, Suzuki H, Uzuka H, Nishimiya K, Shimokawa H. Treadmill exercise prevents reduction of bone mineral density after myocardial infarction in apolipoprotein E-deficient mice. Eur J Prev Cardiol 2019; 27:28-35. [PMID: 30857427 DOI: 10.1177/2047487319834399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIMS Recent clinical studies demonstrated the association between myocardial infarction (MI) and osteoporotic fractures. We examined whether MI causes bone loss and the effects of exercise training on bone in mice after MI. METHODS We created a MI model in 16-week-old male apolipoprotein E-deficient mice (n = 42), which were randomly assigned to exercise group (MI-Ex) and sedentary group (MI-Sed). We also performed sham operations in other mice (n = 10). Treadmill exercise training was performed from one week after operation to eight weeks. At eight weeks, the bone parameters of the femur were measured by quantitative computed tomography, followed by histological analysis (n = 10-17). RESULTS Bone mineral density (BMD) of the femur was significantly decreased in the MI-Sed group as compared with the sham group (P < 0.001), whereas the BMD was significantly increased in the MI-Ex group as compared with the MI-Sed group (P < 0.05). In histological analysis, Rho-associated coiled-coil kinase 2 and tartrate-resistant acid phosphate positive (bone resorptive) area in distal femur were significantly increased in the MI-Sed group as compared with the sham group (P < 0.05), whereas those parameters were significantly decreased in the MI-Ex group as compared with the MI-Sed group (P < 0.05). In contrast, alkaline phosphatase (ALP)-positive (bone-forming) area was significantly decreased in the MI-Sed group as compared with the sham group (P < 0.05), whereas ALP-positive area was significantly increased in the MI-Ex group as compared with the MI-Sed group (P < 0.05). CONCLUSIONS The present study demonstrates that MI reduces BMD and treadmill exercise training prevents the reduction of BMD in apolipoprotein E-deficient mice.
Collapse
Affiliation(s)
- Masanori Kanazawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuharu Matsumoto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kikuyo Takahashi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideaki Suzuki
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironori Uzuka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kensuke Nishimiya
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
49
|
Identification of Novel Therapeutic Targets for Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19124081. [PMID: 30562953 PMCID: PMC6321293 DOI: 10.3390/ijms19124081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) are fatal diseases; however, their pathogenesis still remains to be elucidated. We have recently screened novel pathogenic molecules and have performed drug discovery targeting those molecules. Pulmonary artery smooth muscle cells (PASMCs) in patients with PAH (PAH-PASMCs) have high proliferative properties like cancer cells, which leads to thickening and narrowing of distal pulmonary arteries. Thus, we conducted a comprehensive analysis of PAH-PASMCs and lung tissues to search for novel pathogenic proteins. We validated the pathogenic role of the selected proteins by using tissue-specific knockout mice. To confirm its clinical significance, we used patient-derived blood samples to evaluate the potential as a biomarker for diagnosis and prognosis. Finally, we conducted a high throughput screening and found inhibitors for the pathogenic proteins.
Collapse
|
50
|
Gong W, Qie S, Huang P, Xi J. Deletion of long noncoding RNA EFNA3 aggravates hypoxia-induced injury in PC-12 cells by upregulation of miR-101a. J Cell Biochem 2018; 120:836-847. [PMID: 30125989 DOI: 10.1002/jcb.27444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/16/2018] [Indexed: 11/07/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been reported to be involved in several neurological pathogenesis conditions including cerebral ischemia. In the current study, the functions of lncRNA EFNA3 on hypoxia-injured rat adrenal pheochromocytoma (PC-12) cells and the underlying molecular mechanism were studied. The expression of lncRNA EFNA3 was silenced by short hairpin RNA transfection, after which the cells were subjected with hypoxia. Cell viability, migration, invasion, and apoptosis were, respectively, determined by trypan blue staining, Transwell assay, annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double-staining, and Western blot analysis. The cross regulation between lncRNA EFNA3 and miR-101a, as well as between miR-101a and Rho associated coiled-coil containing protein kinase 2 (ROCK2) were detected by performing quantitative real-time polymerase chain reaction, RNA pull-down, RNA immunoprecipitation, luciferase activity assay, and Western blot analysis. Studies showed that lncRNA EFNA3 was highly expressed in response to hypoxia. Deletion of lncRNA EFNA3 significantly aggravated hypoxia-induced injury in PC-12 cells, as the impairment of cell viability, migration, and invasion, and the inducement of apoptosis. LncRNA EFNA3 worked as a sponging molecule for miR-101a and miR-101a suppression-protected PC-12 cells against hypoxia-induced injury even when lncRNA EFNA3 was silenced. ROCK2 was a target gene of miR-101a. ROCK2 overexpression exhibited neuroprotective activities. Besides, ROCK2 overexpression activated Wnt/β-catenin pathway whereas it deactivated JAK/STAT pathway upon hypoxia. Our study suggests that deletion of lncRNA EFNA3 aggravates hypoxia-induced injury in PC-12 cells by upregulating miR-101a, which further targets ROCK2.
Collapse
Affiliation(s)
- Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Shuyan Qie
- Department of Rehabilitation Therapy, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Peiling Huang
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Jianing Xi
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|