1
|
Zheng Y, Yao M, Chen S, Li J, Wei X, Qiu Z, Chen L, Zhang L. HMGB2 promotes smooth muscle cell proliferation through PPAR-γ/PGC-1α pathway-mediated glucose changes in aortic dissection. Atherosclerosis 2024; 399:119044. [PMID: 39531897 DOI: 10.1016/j.atherosclerosis.2024.119044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Aortic dissection (AD) is a fatal condition with a complicated pathogenesis. High mobility group protein B2 (HMGB2) is a member of the high mobility group protein family; HMGB2 is involved in innate immunity and inflammatory diseases, but its role in AD remains unclear. METHODS HMGB2-/- mice were generated and treated with β-aminopropionitrile and angiotensin II (Ang II) to establish an AD model. An F12 gel containing AAV9-HMGB2 was applied to overexpress HMGB2 in mice. Pathological changes in the aorta were assessed by visualizing vascular collagen deposition and elastic fiber fracture via H&E, Masson and EVG staining. HMGB2 expression was measured by Western blotting and immunohistochemistry. MTS, CCK-8 and EdU assays were used to test cell proliferation. RESULTS HMGB2 expression was increased in samples from AD patients, samples from AD mouse modeland human aortic smooth muscle cells (HASMCs). HMGB2 promoted HASMC proliferation. Immunofluorescence staining and plasma membrane protein isolation revealed that HMGB2 decreased GLUT1 expression and promoted GLUT4 translocation. HMGB2 was also found to inhibit the expression of SIRT1/PGC-1α, but blocking the PPAR-γ pathway attenuated this effect. HMGB2-/- significantly reduced the incidence and mortality rates of AD, whereas treatment with AAV9-HMGB2 exacerbated AD. CONCLUSIONS This study suggests that HMGB2 promotes HASMC proliferation and vascular remodeling by regulating glucose metabolism through the PPAR-γ/SIRT1/PGC-1α pathway. HMGB2 knockdown reduces, while HMGB2 overexpression promotes, the occurrence of AD in mice. This study may help elucidate the underlying mechanisms and provide a new preventive target for AD.
Collapse
MESH Headings
- Animals
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- PPAR gamma/metabolism
- Cell Proliferation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Humans
- Signal Transduction
- Mice
- HMGB2 Protein/metabolism
- HMGB2 Protein/genetics
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Disease Models, Animal
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Glucose/metabolism
- Male
- Mice, Inbred C57BL
- Cells, Cultured
- Aorta/pathology
- Aorta/metabolism
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Glucose Transporter Type 4/metabolism
- Sirtuin 1/metabolism
Collapse
Affiliation(s)
- Yameng Zheng
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Mengge Yao
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Shaokun Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jiakang Li
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Xiaozhen Wei
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Zhihuang Qiu
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| | - Li Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Pathophysiology, The School of Basic Medical Sciences, China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| |
Collapse
|
2
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [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: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
3
|
Wu J, Wang W, Huang Y, Wu H, Wang J, Han M. Deletion of SM22α disrupts the structure and function of caveolae and T-tubules in cardiomyocytes, contributing to heart failure. PLoS One 2022; 17:e0271578. [PMID: 35849583 PMCID: PMC9292107 DOI: 10.1371/journal.pone.0271578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/04/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Smooth muscle 22-alpha (SM22α) is an actin-binding protein that plays critical roles in mediating polymerization of actin filaments and stretch sensitivity of cytoskeleton in vascular smooth muscle cells (VSMCs). Multiple lines of evidence indicate the existence of SM22α in cardiomyocytes. Here, we investigated the effect of cardiac SM22α on the membrane architecture and functions of cardiomyocytes to pressure overload. Methods SM22α knock-out (KO) mice were utilized to assess the role of SM22α in the heart. Echocardiography was used to evaluate cardiac function, transverse aortic constriction (TAC) was used to induce heart failure, cell shortening properties were measured by IonOptix devices in intact cardiomyocytes, Ca2+ sensitivity of myofilaments was measured in permeabilized cardiomyocytes. Confocal microscopy, electron microscopy, western blotting, co-immunoprecipitation (co-IP), Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) techniques were used to perform functional and structural analysis. Results SM22α ablation did not alter cardiac function at baseline, but mRNA levels of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and β-myosin heavy chain (β-MHC) were increased significantly compared with wild type (WT) controls. The membrane architecture was severely disrupted in SM22α KO cardiomyocytes, with disassembly and flattening of caveolae and disrupted T-tubules. Furthermore, SM22α was co-immunoprecipitated with caveolin-3 (Cav3), and the interaction between Cav3 and actin was significantly reduced in SM22α KO cells. SM22α KO cardiomyocytes displayed asynchronized SR Ca2+ release, significantly increased Ca2+ spark frequency. Additionally, the kinetics of sarcomere shortening was abnormal, accompanied with increased sensitivity and reduced maximum response of myofilaments to Ca2+ in SM22α KO cardiomyocytes. SM22α KO mice were more prone to heart failure after TAC. Conclusions Our findings identified that SM22α may be required for the architecture and function of caveolae and T-tubules in cardiomyocytes.
Collapse
Affiliation(s)
- Jun Wu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Wei Wang
- Department of Physiology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yaomeng Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Haochen Wu
- Department of Physiology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jiabin Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
- * E-mail:
| |
Collapse
|
4
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 400] [Impact Index Per Article: 133.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
5
|
Huang M, Dong Y, Sun G, Yu Y. Circ-Sirt1 inhibits vascular smooth muscle cells proliferation via the c-Myc/cyclin B1 axis. Cell Biol Int 2022; 46:628-636. [PMID: 34989469 DOI: 10.1002/cbin.11758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/24/2021] [Accepted: 01/01/2022] [Indexed: 11/09/2022]
Abstract
Vascular smooth muscle cells(VSMCs)are an important cellular component of vascular wall. Restenosis is mainly due to VSMC excessive proliferation. However, little is known about the role of circRNAs in VSMC proliferation and phenotypic switching. Herein, using FISH assay and RT-qPCR, we found that circ-Sirt1 was markedly downregulated in neointimal formation after injury and in VSMCs treated with PDGF-BB. BrdU and MTT assays confirmed the inhibitory role of circ-Sirt1 on cell proliferation. Mechanistically, circ-Sirt1 was mainly expressed in the cytoplasm of VSMCs. Through RIP and RNA pull-down assays, we found that circ-Sirt1 bound c-Myc, protein associated with proliferation of VSMCs. ChIP assay also provided evidence that the overexpression of circ-Sirt1 almost ceased PDGF-BB-induced binding of c-Myc to the promoter of cyclin B1 in VSMCs. These results indicated that circ-Sirt1 had an inhibitory effect on c-Myc activity, providing a mechanism for suppressing PDGF-BB-induced VSMC proliferation by direct interactions with c-Myc and its sequestration in the cytoplasm. Overall, our study demonstrated that a previously unrecognized circ-Sirt1/c-Myc/cyclin B1 axis in VSMCs mediates neointimal formation following injury. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Minhua Huang
- Binzhou Medical University, Yantai, Shandong, China
| | - Yujie Dong
- Binzhou Medical University, Yantai, Shandong, China
| | - Guangbin Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| | - Yuan Yu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
6
|
Zhang BF, Wu ZH, Deng J, Jin HJ, Chen WB, Zhang S, Liu XJ, Wang WT, Zheng XT. M 6A methylation-mediated elevation of SM22α inhibits the proliferation and migration of vascular smooth muscle cells and ameliorates intimal hyperplasia in type 2 diabetes mellitus. Biol Chem 2021; 403:317-329. [PMID: 34882999 DOI: 10.1515/hsz-2021-0296] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/18/2021] [Indexed: 01/02/2023]
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by insulin resistance facilitates intimal hyperplasia of type 2 diabetes mellitus (T2DM) and N6-methyladenosine (m6A) methylation modification mediates the VSMC proliferation. This study aimed to reveal the m6A methylation modification regulatory mechanism. In this study, m6A demethylase FTO was elevated in insulin-treated VSMCs and T2DM mice with intimal injury. Functionally, FTO knockdown elevated m6A methylation level and further restrained VSMC proliferation and migration induced by insulin. Mechanistically, FTO knockdown elevated Smooth muscle 22 alpha (SM22α) expression and m6A-binding protein IGF2BP2 enhanced SM22α mRNA stability by recognizing and binding to m6A methylation modified mRNA. In vivo studies confirmed that the elevated m6A modification level of SM22α mRNA mitigated intimal hyperplasia in T2DM mice. Conclusively, m6A methylation-mediated elevation of SM22α restrained VSMC proliferation and migration and ameliorated intimal hyperplasia in T2DM.
Collapse
Affiliation(s)
- Bao-Fu Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Zi-Heng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jie Deng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Hao-Jie Jin
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Wei-Biao Chen
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Sai Zhang
- Institute of Ischemia-Reperfusion Injury, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiu-Jie Liu
- Institute of Ischemia-Reperfusion Injury, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wan-Tie Wang
- Institute of Ischemia-Reperfusion Injury, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiang-Tao Zheng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| |
Collapse
|
7
|
Panahi M, Rodriguez PR, Fereshtehnejad SM, Arafa D, Bogdanovic N, Winblad B, Cedazo-Minguez A, Rinne J, Darreh-Shori T, Hase Y, Kalaria RN, Viitanen M, Behbahani H. Insulin-Independent and Dependent Glucose Transporters in Brain Mural Cells in CADASIL. Front Genet 2020; 11:1022. [PMID: 33101365 PMCID: PMC7522350 DOI: 10.3389/fgene.2020.01022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/10/2020] [Indexed: 11/26/2022] Open
Abstract
Typical cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by mutations in the human NOTCH3 gene. Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy is characterized by subcortical ischemic strokes due to severe arteriopathy and fibrotic thickening of small vessels. Blood regulating vascular smooth muscle cells (VSMCs) appear as the key target in CADASIL but the pathogenic mechanisms remain unclear. With the hypothesis that brain glucose metabolism is disrupted in VSMCs in CADASIL, we investigated post-mortem tissues and VSMCs derived from CADASIL patients to explore gene expression and protein immunoreactivity of glucose transporters (GLUTs), particularly GLUT4 and GLUT2 using quantitative RT-PCR and immunohistochemical techniques. In vitro cell model analysis indicated that both GLUT4 and -2 gene expression levels were down-regulated in VSMCs derived from CADASIL patients, compared to controls. In vitro studies further indicated that the down regulation of GLUT4 coincided with impaired glucose uptake in VSMCs, which could be partially rescued by insulin treatment. Our observations on reduction in GLUTs in VSMCs are consistent with previous findings of decreased cerebral blood flow and glucose uptake in CADASIL patients. That impaired ability of glucose uptake is rescued by insulin is also consistent with previously reported lower proliferation rates of VSMCs derived from CADASIL subjects. Overall, these observations are consistent with the development of severe cerebral arteriopathy in CADASIL, in which VSMCs are replaced by widespread fibrosis.
Collapse
Affiliation(s)
- Mahmod Panahi
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Patricia Rodriguez Rodriguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Seyed-Mohammad Fereshtehnejad
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Donia Arafa
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Nenad Bogdanovic
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden.,Neurogeriatric Clinic, Karolinska University Hospital, Huddinge, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Juha Rinne
- University of Turku, Turku University Hospital Kiinanmyllynkatu, Turku, Finland
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Yoshiki Hase
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matti Viitanen
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden.,Department of Geriatrics, Turun Kaupunginsairaala, University Hospital of Turku, University of Turku, Turku,Finland
| | - Homira Behbahani
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Gao Y, Liu G, Kong P, Song Y, Zhang D, Yin Y, Han M. Smooth muscle 22α deficiency impairs oxytocin-induced uterine contractility in mice at full-term pregnancy. Biochem Biophys Res Commun 2020; 529:884-889. [PMID: 32819594 DOI: 10.1016/j.bbrc.2020.05.220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 11/18/2022]
Abstract
Smooth muscle 22α (SM22α, namely Transgelin), as an actin-binding protein, regulates the contractility of vascular smooth muscle cells (VSMCs) by modulation of the stress fiber formation. However, little is known about the roles of SM22α in the regulation of uterine contraction during parturition. Here, we showed that contraction in response to oxytocin (OT) was significantly decreased in the uterine muscle strips from SM22α knockout (Sm22α-KO) mice, especially at full-term pregnancy, which may be resulted from impaired formation of stress fibers. Furthermore, serious mitochondrial damage such as the mitochondrial swelling, cristae disruption and even disappearance were observed in the myometrium of Sm22α-KO mice at full-term pregnancy, eventually resulting in the collapse of mitochondrial membrane potential and impairment in ATP synthesis. Our data indicate that SM22α is necessary to maintain uterine contractility at delivery in mice, and acts as a novel target for preventive or therapeutic manipulation of uterine atony during parturition.
Collapse
MESH Headings
- Adenosine Triphosphate/deficiency
- Animals
- Female
- Gene Expression Regulation
- Mice
- Mice, Knockout
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Mitochondria/drug effects
- Mitochondria/genetics
- Mitochondria/metabolism
- Mitochondrial Swelling/genetics
- Muscle Proteins/deficiency
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myometrium/drug effects
- Myometrium/metabolism
- Myometrium/pathology
- Oxytocin/pharmacology
- Parturition
- Pregnancy
- Primary Cell Culture
- Stress Fibers/drug effects
- Stress Fibers/metabolism
- Stress Fibers/pathology
- Tissue Culture Techniques
- Uterine Contraction/drug effects
- Uterine Inertia/genetics
- Uterine Inertia/metabolism
- Uterine Inertia/pathology
Collapse
Affiliation(s)
- Yakun Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Guixia Liu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, China
| | - Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yu Song
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Dandan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yajuan Yin
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
9
|
Qin X, Hou X, Zhang K, Li Q. α 1D-adrenoceptor involves the relaxation effect of farrerol in rat aortic vascular smooth muscle cells. Eur J Pharmacol 2019; 853:169-183. [PMID: 30768980 DOI: 10.1016/j.ejphar.2019.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/31/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022]
Abstract
The aim of this study was to investigate the relaxation effect of farrerol on rat aortic vascular smooth muscle cells (VSMCs) and its underlying mechanism. VSMCs were cultured primarily and were used to examine the relaxation effect of farrerol. Cells surface and length were measured by dynamic observation, or by rhodamine-phalloidin labeling and hematoxylin-eosin staining. Cells contractive activity were tested using collagen gel contraction assay. The [Ca2+]in was measured with molecular probe fluo-4-AM. The mRNA and protein expression of regulatory proteins for contraction were measured. In addition, rat aortic VSMCs were transfected with lentivirus-mediated α1D-adrenoceptor gene-shRNA, then the effect of farrerol were detected by the above experimental methods. The results revealed that 10 μΜ AngⅡ promoted cell contraction, increased [Ca2+]in and enhanced collagen contraction in rat aortic VSMCs. 10 μΜ AngⅡ not only increased expression of myosin light chain kinase (MLCK) and smooth muscle protein 22α (SM22α), but also increased phosphorylation level of myosin light chain (MLC) and myosin phosphatase target subunit 1 (MYPT1). The above effects induced by AngⅡ could be significantly inhibited by farrerol in a concentration dependent manner. When the cells were transfected with lentivirus mediated α1D-adrenoceptor gene-shRNA, the effects of farrerol on changes induced by AngⅡ in rat aortic VSMCs were markedly reversed. In conclusion, farrerol could produce relaxtion effect in rat aortic VSMCs precontracted by 10 μΜ AngⅡ, which was involved in downregulation expression of MLCK and SM22α, and inhibition phosphorylation level of MYPT1 and MLC via activating α1D-adrenoceptor gene.
Collapse
Affiliation(s)
- Xiaojiang Qin
- School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan 030001, Shanxi Province, China; Shanxi University of Chinese medicine, No. 121, Daxuejie, Jinzhong 030619, Shanxi Province, China
| | - Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan 030001, Shanxi Province, China
| | - Kun Zhang
- School of Pharmaceutical Science, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan 030001, Shanxi Province, China
| | - Qingshan Li
- Shanxi University of Chinese medicine, No. 121, Daxuejie, Jinzhong 030619, Shanxi Province, China; School of Pharmaceutical Science, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan 030001, Shanxi Province, China.
| |
Collapse
|
10
|
Li J, Wang H, Shi X, Zhao L, Lv T, Yuan Q, Hao W, Zhu J. Anti-proliferative and anti-migratory effects of Scutellaria strigillosa Hemsley extracts against vascular smooth muscle cells. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:155-163. [PMID: 30763696 DOI: 10.1016/j.jep.2019.02.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/30/2019] [Accepted: 02/09/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The abnormal increase in vascular smooth muscle cell (VSMC) proliferation and migration are critical events in the pathogenesis of cardiovascular diseases (CVDs) including restenosis and atherosclerosis. The dried roots of Scutellaria baicalensis Georgi (common name: Huangqin in China) have been confirmed to possess beneficial effects on CVD by clinical and modern pharmacological studies. Flavonoids in Huangqin exert anti-proliferative and anti-migratory effects. Similar to Huangqin, Scutellaria strigillosa Hemsley (SSH) has been used to clear heat and damp and is especially rich in flavonoids including wogonin, wogonoside, baicalein, and baicalin. However, there have been few of reports about pharmacological activities of SSH. AIM OF THE STUDY To investigate the anti-proliferative and anti-migratory properties of Scutellaria strigillosa Hemsley extract (SSHE) in vitro and in vivo and explore its possible mechanism of action. MATERIALS AND METHODS The chemical constituents of SSHE were analyzed by ultra-high performance liquid chromatography coupled with triple time-of-flight mass spectrometry (UPLC-Triple-TOF-MS/MS). Cell proliferation and migration were investigated using BrdU incorporation assay and cell scratch test, respectively. The protein expression was determined by western blotting. In vivo, we established an artery ligation model of C57BL/6 mice and orally administered them with 50 or 100 mg/kg/day of SSHE. The carotid arteries were harvested and the intima-media thickness was examined 28 days post-ligation. RESULTS Twelve compounds were identified and tentatively characterized. SSHE significantly inhibited the VSMC proliferation and migration stimulated by PDGF-BB and decreased the relative protein expression of regulatory signaling intermediates. Furthermore, the expression of SM22α was significantly elevated in SSHE-pretreated VSMCs, whereas knockdown of SM22α impaired the PDGF-BB-induced proliferation and migration arrest. Meanwhile, both ROS generation and the phosphorylation of ERK decreased in SSHE-pretreated VSMCs. In carotid artery ligation mice model, SSHE treatment significantly inhibited neointimal hyperplasia. CONCLUSIONS SSHE significantly inhibited the PDGF-BB-induced VSMC proliferation, migration, and neointimal hyperplasia of carotid artery caused by ligation. Upregulation of SM22α expression, inhibition of ROS generation and ERK phosphorylation were, at least, partly responsible for the effects of SSHE on VSMCs.
Collapse
MESH Headings
- Animals
- Becaplermin/administration & dosage
- Carotid Intima-Media Thickness
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Chromatography, High Pressure Liquid
- Dose-Response Relationship, Drug
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Plant Extracts/administration & dosage
- Plant Extracts/pharmacology
- Rats
- Scutellaria/chemistry
- Tandem Mass Spectrometry
Collapse
Affiliation(s)
- Jiankun Li
- The Forth Affiliated Hospital of Hebei Medical University, No. 12 Health Road, Shijiazhuang 050011, PR China.
| | - Hairong Wang
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Xiaowei Shi
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Lili Zhao
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Tao Lv
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Qi Yuan
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Wenyang Hao
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| | - Jing Zhu
- Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang 050017, PR China.
| |
Collapse
|