1
|
Tierney JW, Francisco RP, Yu F, Ma J, Cheung-Flynn J, Keech MC, D'Arcy R, Shah VM, Kittel AR, Chang DJ, McCune JT, Bezold MG, Aligwekwe AN, Cook RS, Beckman JA, Brophy CM, Duvall CL. Intravascular delivery of an MK2 inhibitory peptide to prevent restenosis after angioplasty. Biomaterials 2025; 313:122767. [PMID: 39216327 DOI: 10.1016/j.biomaterials.2024.122767] [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: 03/07/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Peripheral artery disease is commonly treated with balloon angioplasty, a procedure involving minimally invasive, transluminal insertion of a catheter to the site of stenosis, where a balloon is inflated to open the blockage, restoring blood flow. However, peripheral angioplasty has a high rate of restenosis, limiting long-term patency. Therefore, angioplasty is sometimes paired with delivery of cytotoxic drugs like paclitaxel to reduce neointimal tissue formation. We pursue intravascular drug delivery strategies that target the underlying cause of restenosis - intimal hyperplasia resulting from stress-induced vascular smooth muscle cell switching from the healthy contractile into a pathological synthetic phenotype. We have established MAPKAP kinase 2 (MK2) as a driver of this phenotype switch and seek to establish convective and contact transfer (coated balloon) methods for MK2 inhibitory peptide delivery to sites of angioplasty. Using a flow loop bioreactor, we showed MK2 inhibition in ex vivo arteries suppresses smooth muscle cell phenotype switching while preserving vessel contractility. A rat carotid artery balloon injury model demonstrated inhibition of intimal hyperplasia following MK2i coated balloon treatment in vivo. These studies establish both convective and drug coated balloon strategies as promising approaches for intravascular delivery of MK2 inhibitory formulations to improve efficacy of balloon angioplasty.
Collapse
Affiliation(s)
- J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - R Paolo Francisco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Chemical Engineering, School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA
| | - Veeraj M Shah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Devin J Chang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Adrian N Aligwekwe
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; North Carolina State University, Raleigh, NC, 27695, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua A Beckman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Gao XF, Chen AQ, Tang HY, Kong XQ, Zhang H, Wang ZM, Lu W, Wang LG, Wang F, Zhou WY, Gu Y, Zuo GF, Ge Z, Zhang JJ, Chen SL. m 6A Modification of Profilin-1 in Vascular Smooth Muscle Cells Drives Phenotype Switching and Neointimal Hyperplasia via Activation of the p-ANXA2/STAT3 Pathway. Arterioscler Thromb Vasc Biol 2024. [PMID: 39508106 DOI: 10.1161/atvbaha.124.321399] [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: 06/15/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND In-stent restenosis is characterized by a significant reduction in lumen diameter within the stented segment, primarily attributed to excessive proliferation of vascular smooth muscle cells (VSMCs) and neointimal hyperplasia. PFN1 (profilin-1), an actin-sequestering protein extensively studied in amyotrophic lateral sclerosis, remains less explored in neointimal hyperplasia. METHODS Utilizing single-cell RNA sequencing alongside data from in-stent restenosis patients and various experimental in-stent restenosis models (swine, rats, and mice), we investigated the role of PFN1 in promoting VSMC phenotype switching and neointimal hyperplasia. RESULTS Single-cell RNA sequencing of stenotic rat carotid arteries revealed a critical role for PFN1 in neointimal hyperplasia, a finding corroborated in stented swine coronary arteries, in-stent restenosis patients, PFN1SMC-IKO (SMC-specific PFN1 knockout) mice, and PFN1 overexpressed mice. PFN1 deletion was shown to suppress VSMC phenotype switching and neointimal hyperplasia in PFN1SMC-IKO mice subjected to a wire-injured model. To elucidate the observed discordance in PFN1 mRNA and protein levels, we identified that METTL3 (N6-methyladenosine methyltransferase) and YTHDF3 (N6-methyladenosine-specific reader) enhance PFN1 translation efficiency in an N6-methyladenosine-dependent manner, confirmed through experiments involving METTL3 knockout and YTHDF3 knockout mice. Furthermore, PFN1 was mechanistically found to interact with the phosphorylation of ANXA2 (annexin A2) by recruiting Src, promoting the phosphorylation of STAT3, a typical transcription factor known to induce VSMC phenotype switching. CONCLUSIONS This study unveils the significance of PFN1 N6-methyladenosine modification in VSMCs, demonstrating its role in promoting phenotype switching and neointimal hyperplasia through the activation of the p-ANXA2 (phospho-ANXA2)/STAT3 pathway.
Collapse
Affiliation(s)
- Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Ai-Qun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Hao-Yue Tang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Xiang-Quan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Huan Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Zhi-Mei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Wei Lu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Li-Guo Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Feng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Wen-Ying Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Guang-Feng Zuo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Zhen Ge
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Jun-Jie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, China
| |
Collapse
|
3
|
Yao G, Zhang X, Zhang T, Jin J, Qin Z, Ren X, Wang X, Zhang S, Yin X, Tian Z, Zhang Y, Zhang J, Wang Z, Zhang Q. The role of dysbiotic gut mycobiota in modulating risk for abdominal aortic aneurysm. Microbiol Spectr 2024; 12:e0177624. [PMID: 39315850 PMCID: PMC11537029 DOI: 10.1128/spectrum.01776-24] [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: 07/19/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a large-vessel disease with high mortality, characterized by complex pathogenic mechanisms. Current therapeutic approaches remain insufficient to halt its progression. Fungi are important members of the gut microbiota. However, their characteristic alterations and roles in AAA remain unclear. This study investigated the role of gut fungal communities in the development of AAA through metagenomic sequencing of fecal samples from 31 healthy individuals and 33 AAA patients. We observed significant dysbiosis in the gut mycobiomes of AAA patients compared to healthy individuals, characterized by an increase in pathogenic fungi like Candida species and a decrease in beneficial yeasts such as Saccharomyces cerevisiae. The changes in fungal populations correlated strongly with clinical indicators of AAA, highlighting their potential for diagnosing and predicting AAA progression. Furthermore, our animal experiments demonstrated that Saccharomyces cerevisiae significantly ameliorated pathological alterations in AAA mice, suggesting a protective role for specific yeast strains against AAA development. These findings underscore the significant impact of gut mycobiomes on AAA and suggest that modulating these fungal communities could offer a novel therapeutic approach. Our research advances the understanding of the influence of gut microbiome on vascular diseases and suggests potential non-surgical approaches for managing AAA. By elucidating the diagnostic and therapeutic potential of gut fungi in AAA, this study provided important clues for future clinical strategies and therapeutic developments in the field of vascular medicine. IMPORTANCE Our research highlights the crucial role of gut fungi in abdominal aortic aneurysm (AAA) development. By analyzing fecal samples from AAA patients and healthy controls, we discovered significant dysbiosis in gut fungal communities, characterized by an increase in harmful Candida species and a decrease in beneficial yeasts like Saccharomyces cerevisiae. This dysbiosis was correlated with the severity of AAA. Importantly, in animal experiments, supplementing with Saccharomyces cerevisiae significantly slowed AAA progression. These findings suggest that modulating gut fungi may offer a novel, non-surgical approach to the diagnosis and treatment of AAA, potentially reducing the need for invasive procedures.
Collapse
Affiliation(s)
- Guixiang Yao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinjie Zhang
- Department of Biology, University College London, London, United Kingdom
| | - Tongxue Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jiajia Jin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zihan Qin
- Department of Endocrinology & Geriatrics, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoyu Ren
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaowei Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shucui Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xianlun Yin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenyu Tian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyong Zhang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Wang
- Department of Endocrinology & Geriatrics, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qunye Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
4
|
Kumariya S, Grano de Oro A, Nestor-Kalinoski AL, Joe B, Osman I. Gut microbiota-derived Metabolite, Shikimic Acid, inhibits vascular smooth muscle cell proliferation and migration. Biochem Pharmacol 2024; 229:116524. [PMID: 39251142 DOI: 10.1016/j.bcp.2024.116524] [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: 05/07/2024] [Revised: 07/25/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Gut microbiota dysbiosis is linked to vascular wall disease, but the mechanisms by which gut microbiota cross-talk with the host vascular cells remain largely unknown. Shikimic acid (SA) is a biochemical intermediate synthesized in plants and microorganisms, but not mammals. Surprisingly, recent metabolomic profiling data demonstrate that SA is detectable in human and murine blood. In this study, analyzing data from germ-free rats, we provide evidence in support of SA as a bona fide gut microbiota-derived metabolite, emphasizing its biological relevance. Since vascular cells are the first cells exposed to circulating metabolites, in this study, we examined, for the first time, the effects and potential underlying molecular mechanisms of SA on vascular smooth muscle cell (VSMC) proliferation and migration, which play a key role in occlusive vascular diseases, such as post-angioplasty restenosis and atherosclerosis. We found that SA inhibits the proliferation and migration of human coronary artery SMCs. At the molecular level, unexpectedly, we found that SA activates, rather than inhibits, multiple pro-mitogenic signaling pathways in VSMCs, such as ERK1/2, AKT, and mTOR/p70S6K. Conversely, we found that SA activates the anti-proliferative AMP-activated protein kinase (AMPK) in VSMCs, a key cellular energy sensor and regulator. However, loss-of-function experiments demonstrate that AMPK does not mediate the inhibitory effects of SA on VSMC proliferation. In conclusion, these studies demonstrate that a microbiota-derived metabolite, SA, inhibits VSMC proliferation and migration in vitro and prompt further evaluation of the possible underlying molecular mechanisms and the potential protective role in VSMC-related vascular wall disease in vivo.
Collapse
Affiliation(s)
- Sanjana Kumariya
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, USA
| | - Arturo Grano de Oro
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, USA
| | | | - Bina Joe
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, USA
| | - Islam Osman
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
5
|
Lu X, Wang Y, Geng N, Zou Z, Feng X, Wang Y, Xu Z, Zhang N, Pu J. Dysregulated Mitochondrial Calcium Causes Spiral Artery Remodeling Failure in Preeclampsia. Hypertension 2024; 81:2368-2382. [PMID: 39291377 DOI: 10.1161/hypertensionaha.124.23046] [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: 03/25/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Calcium deficiency in women is strongly linked to an increased risk of developing preeclampsia. Mitochondrial calcium ([Ca2+]m) homeostasis is essential to regulate vascular smooth muscle cell (VSMC) function. However, the role of [Ca2+]m in preeclampsia development remains largely unknown. METHODS To investigate this, human spiral arteries obtained from normotensive and preeclamptic women were collected for vascular function, RNA sequencing, and VSMC studies. N(ω)-nitro-L-arginine methyl ester-induced preeclampsia animal experiments were established to investigate the effects of intervening in [Ca2+]m to improve the outcome for preeclamptic mothers or their infants. RESULTS Our initial findings revealed compromised vessel function in spiral arteries derived from patients with preeclampsia, as evidenced by diminished vasoconstriction and vasodilation responses to angiotensin II and sodium nitroprusside, respectively. Moreover, the spiral artery VSMCs from patients with preeclampsia exhibited phenotypic transformation and proliferation associated with the disrupted regulatory mechanisms of [Ca2+]m uptake. Subsequent in vitro experiments employing gain- and loss-of-function approaches demonstrated that the mitochondrial Na+/Ca2+ exchanger played a role in promoting phenotypic switching and impaired mitochondrial functions in VSMCs. Furthermore, mtNCLX (mitochondrial Na+/Ca2+ exchanger) inhibitor CGP37157 significantly improved VSMC phenotypic changes and restored mitochondrial function in both patients with preeclampsia-derived VSMCs and the preeclampsia rat model. CONCLUSIONS This study provides comprehensive evidence supporting the disrupted regulatory mechanisms of [Ca2+]m uptake in VSMCs of spiral arteries of patients with preeclampsia and further elucidates its correlation with VSMC phenotypic switching and defective spiral artery remodeling. The findings suggest that targeting mtNCLX holds promise as a novel therapeutic approach for managing preeclampsia.
Collapse
Affiliation(s)
- Xiyuan Lu
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yifan Wang
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Na Geng
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhiguo Zou
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xueqing Feng
- Department of Obstetrics Affiliated Hospital of Jining Medical University, China (X.F.)
| | - Yuehong Wang
- State Key Laboratory of Systems Medicine for Cancer (Yuehong Wang), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| | - Zhice Xu
- Wuxi Maternity and Child Health Care Hospital, China (Z.X.)
| | - Ning Zhang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology (N.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital State Key Laboratory of Systems Medicine for Cancer (J.P.), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| |
Collapse
|
6
|
Kane J, Lemieux A, Baranwal G, Misra S. The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells 2024; 13:1637. [PMID: 39404400 PMCID: PMC11475948 DOI: 10.3390/cells13191637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Vascular access is an indispensable component of haemodialysis therapy for end-stage kidney disease patients. The arteriovenous fistula (AVF) is most common, but importantly, two-year failure rates are greater than fifty percent. AVF failure can occur due to a lack of suitable vascular remodelling, and inappropriate inflammation preventing maturation, or alternatively neointimal hyperplasia and vascular stenosis preventing long-term use. A comprehensive mechanistic understanding of these processes is still lacking, but recent studies highlight an essential role for inflammation from uraemia and the AVF itself. Inflammation affects each cell in the cascade of AVF failure, the endothelium, the infiltrating immune cells, and the vascular smooth muscle cells. This review examines the role of inflammation in each cell step by step and the influence on AVF failure. Inflammation resulting in AVF failure occurs initially via changes in endothelial cell activation, permeability, and vasoprotective chemokine secretion. Resultingly, immune cells can extravasate into the subendothelial space to release inflammatory cytokines and cause other deleterious changes to the microenvironment. Finally, all these changes modify vascular smooth muscle cell function, resulting in excessive and unchecked hyperplasia and proliferation, eventually leading to stenosis and the failure of the AVF. Finally, the emerging therapeutic options based off these findings are discussed, including mesenchymal stem cells, small-molecule inhibitors, and far-infrared therapies. Recent years have clearly demonstrated a vital role for inflammation in deciding the fate of the AVF, and future works must be centred on this to develop therapies for a hitherto unacceptably underserved patient population.
Collapse
Affiliation(s)
| | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; (J.K.); (A.L.); (G.B.)
| |
Collapse
|
7
|
Gou Y, Zhao A, Qin T, Yang B. Identification of the Neointimal Hyperplasia-Related LncRNA-mRNA-Immune Cell Regulatory Network in a Rat Carotid Artery Balloon Injury Model. Int Heart J 2024; 65:945-955. [PMID: 39261031 DOI: 10.1536/ihj.24-062] [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] [Indexed: 09/13/2024]
Abstract
Excessive neointimal hyperplasia (NIH) of coronary vessels in patients is the main cause of restenosis (RS) after percutaneous coronary intervention (PCI). This study aimed to identify the regulatory genes related to NIH in a rat carotid artery balloon injury model.We established a rat model and performed RNA sequencing to identify differentially expressed long non-coding RNAs (DElncRNAs) and differentially expressed message RNAs (DEmRNAs). Immune cells were analyzed using a murine Microenvironment Cell Population counter. The Pearson correlation between DEmRNAs, DElncRNAs, and immune cells was analyzed, followed by function enrichment analysis. Core DEmRNA was identified using Cytoscape. Next, a core lncRNAs-mRNAs-immune cell regulatory network was constructed. NIH-related gene sets from the Gene Expression Omnibus and GeneCards databases were used for validation.A total of 2,165 DEmRNAs and 705 DElncRNAs were identified in rat carotid artery tissue. Four key immune cells were screened out, including mast cells, vessels, endothelial cells, and fibroblasts. Based on the Pearson correlation between DEmRNAs, DElncRNAs and 4 key immune cells, 246 DEmRNAs and 93 DElncRNAs were obtained. DEmRNAs that interact with lncRNAs were mainly involved in the cell cycle, MAPK signaling pathway, and PI3K-Akt signaling pathway. A core lncRNA-mRNA-immune cell regulatory network was constructed, including 9 mRNAs, 4 lncRNAs, and fibroblasts. External datasets validation confirmed the significant correlation of both these mRNAs and lncRNAs with NIH.In this study, an lncRNA-mRNA-immune cell regulatory network related to NIH was constructed, which provided clues for exploring the potential mechanism of RS in cardiovascular diseases.
Collapse
Affiliation(s)
- Yuan Gou
- Department of Vascular Surgery, Jining Medical University Affiliated Jining No. 1 People's Hospital
| | - Anli Zhao
- Department of Cardiovascular Medicine, Affiliated Hospital of Jining Medical University
| | - Tao Qin
- Department of Vascular Surgery, Jining Medical University Affiliated Jining No. 1 People's Hospital
| | - Bin Yang
- Department of Vascular Surgery, Jining No. 1 People's Hospital
| |
Collapse
|
8
|
Tumenbayar BI, Pham K, Biber JC, Drewes R, Bae Y. Transcriptomic and Multi-scale Network Analyses Reveal Key Drivers of Cardiovascular Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612437. [PMID: 39345636 PMCID: PMC11429675 DOI: 10.1101/2024.09.11.612437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cardiovascular diseases (CVDs) and pathologies are often driven by changes in molecular signaling and communication, as well as in cellular and tissue components, particularly those involving the extracellular matrix (ECM), cytoskeleton, and immune response. The fine-wire vascular injury model is commonly used to study neointimal hyperplasia and vessel stiffening, but it is not typically considered a model for CVDs. In this paper, we hypothesize that vascular injury induces changes in gene expression, molecular communication, and biological processes similar to those observed in CVDs at both the transcriptome and protein levels. To investigate this, we analyzed gene expression in microarray datasets from injured and uninjured femoral arteries in mice two weeks post-injury, identifying 1,467 significantly and differentially expressed genes involved in several CVDs such as including vaso-occlusion, arrhythmia, and atherosclerosis. We further constructed a protein-protein interaction network with seven functionally distinct clusters, with notable enrichment in ECM, metabolic processes, actin-based process, and immune response. Significant molecular communications were observed between the clusters, most prominently among those involved in ECM and cytoskeleton organizations, inflammation, and cell cycle. Machine Learning Disease pathway analysis revealed that vascular injury-induced crosstalk between ECM remodeling and immune response clusters contributed to aortic aneurysm, neovascularization of choroid, and kidney failure. Additionally, we found that interactions between ECM and actin cytoskeletal reorganization clusters were linked to cardiac damage, carotid artery occlusion, and cardiac lesions. Overall, through multi-scale bioinformatic analyses, we demonstrated the robustness of the vascular injury model in eliciting transcriptomic and molecular network changes associated with CVDs, highlighting its potential for use in cardiovascular research.
Collapse
Affiliation(s)
- Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Khanh Pham
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
9
|
Rahimzadeh M, Tennstedt S, Aherrahrou Z. Nexilin in cardiomyopathy: unveiling its diverse roles with special focus on endocardial fibroelastosis. Heart Fail Rev 2024; 29:1025-1037. [PMID: 38985384 DOI: 10.1007/s10741-024-10416-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cardiac disorders exhibit considerable heterogeneity, and understanding their genetic foundations is crucial for their diagnosis and treatment. Recent genetic analyses involving a growing number of participants have uncovered novel mutations within both coding and non-coding regions of DNA, contributing to the onset of cardiac conditions. The NEXN gene, encoding the Nexilin protein, an actin filament-binding protein, is integral to normal cardiac function. Mutations in this gene have been linked to cardiomyopathies, cardiovascular disorders, and sudden deaths. Heterozygous or homozygous variants of the NEXN gene are associated with the development of endocardial fibroelastosis (EFE), a rare cardiac condition characterized by excessive collagen and elastin deposition in the left ventricular endocardium predominantly affecting infants and young children. EFE occurs both primary and secondary to other conditions and often leads to unfavorable prognoses and outcomes. This review explores the role of NEXN genetic variants in cardiovascular disorders, particularly EFE, revealing that functional mutations are not clustered in a specific domain of Nexilin based on the cardiac disorder phenotype. Our review underscores the importance of understanding genetic mutations for the diagnosis and treatment of cardiac conditions.
Collapse
Affiliation(s)
- Mahsa Rahimzadeh
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Stephanie Tennstedt
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562, Germany
- University Heart Center Lübeck, Lübeck, 23562, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 23562, Germany.
- University Heart Center Lübeck, Lübeck, 23562, Germany.
| |
Collapse
|
10
|
Mackay CDA, Meechem MB, Patel VB. Macrophages in vascular disease: Roles of mitochondria and metabolic mechanisms. Vascul Pharmacol 2024; 156:107419. [PMID: 39181483 DOI: 10.1016/j.vph.2024.107419] [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: 06/18/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Macrophages are a dynamic cell type of the immune system implicated in the pathophysiology of vascular diseases and are a major contributor to pathological inflammation. Excessive macrophage accumulation, activation, and polarization is observed in aortic aneurysm (AA), atherosclerosis, and pulmonary arterial hypertension. In general, macrophages become activated and polarized to a pro-inflammatory phenotype, which dramatically changes cell behavior to become pro-inflammatory and infiltrative. These cell types become cumbersome and fail to be cleared by normal mechanisms such as autophagy. The result is a hyper-inflammatory environment causing the recruitment of adjacent cells and circulating immune cells to further augment the inflammatory response. In AA, this leads to excessive ECM degradation and chemokine secretion, ultimately causing macrophages to dominate the immune cell landscape in the aortic wall. In atherosclerosis, monocytes are recruited to the vascular wall, where they polarize to the pro-inflammatory phenotype and induce inflammatory pathway activation. This leads to the development of foam cells, which significantly contribute to neointima and necrotic core formation in atherosclerotic plaques. Pro-inflammatory macrophages, which affect other vascular diseases, present with fragmented mitochondria and corresponding metabolic dysfunction. Targeting macrophage mitochondrial dynamics has proved to be an exciting potential therapeutic approach to combat vascular disease. This review will summarize mitochondrial and metabolic mechanisms of macrophage activation, polarization, and accumulation in vascular diseases.
Collapse
Affiliation(s)
- Cameron D A Mackay
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Megan B Meechem
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
11
|
Ghanem L, Essayli D, Kotaich J, Zein MA, Sahebkar A, Eid AH. Phenotypic switch of vascular smooth muscle cells in COVID-19: Role of cholesterol, calcium, and phosphate. J Cell Physiol 2024:e31424. [PMID: 39188012 DOI: 10.1002/jcp.31424] [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: 03/06/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Although the novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily manifests as severe respiratory distress, its impact on the cardiovascular system is also notable. Studies reveal that COVID-19 patients often suffer from certain vascular diseases, partly attributed to increased proliferation or altered phenotype of vascular smooth muscle cells (VSMCs). Although the association between COVID-19 and VSMCs is recognized, the precise mechanism underlying SARS-CoV-2's influence on VSMC phenotype remains largely under-reviewed. In this context, while there is a consistent body of literature dissecting the effect of COVID-19 on the cardiovascular system, few reports delve into the potential role of VSMC switching in the pathophysiology associated with COVID-19 and the molecular mechanisms involved therein. This review dissects and critiques the link between COVID-19 and VSMCs, with particular attention to pathways involving cholesterol, calcium, and phosphate. These pathways underpin the interaction between the virus and VSMCs. Such interaction promotes VSMC proliferation, and eventually potentiates vascular calcification as well as worsens prognosis in patients with COVID-19.
Collapse
Affiliation(s)
- Laura Ghanem
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Dina Essayli
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Jana Kotaich
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- MEDICA Research Investigation, Beirut, Lebanon
| | - Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
12
|
Deng Y, Huang J, Chen C, Wen Y, Qiu D. Fe 3O 4 coated stent prevent artery neointimal hyperplasia by inhibiting vascular smooth muscle cell proliferation. Mater Today Bio 2024; 27:101133. [PMID: 39027680 PMCID: PMC11254736 DOI: 10.1016/j.mtbio.2024.101133] [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: 03/17/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
In-stent restenosis (ISR), caused by aggressive vascular smooth muscle cell (VSMC) proliferation, is a serious complication of stenting. Therefore, developing therapeutic approaches that target VSMC inhibition is imperative. Our previous study showed that VSMC hyperplasia was attenuated after iron stent degradation, and VSMC proliferation around the stented section was arrested. The corrosion products of the iron stents were primarily Fe3O4 particles. Therefore, we hypothesized that Fe3O4 particles generated by iron stents would prevent neointimal hyperplasia by inhibiting VSMC proliferation. To test this hypothesis, culture assays and flow cytometry were performed to investigate the proliferation of VSMC. Global gene sequencing and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to investigate the underlying mechanisms. Fe3O4-coated stents were implanted into rabbit carotid arteries to evaluate the inhibitory effects of Fe3O4 on neointimal hyperplasia. The major findings of the study were as follows: 1) Fe3O4 attenuated neointimal hyperplasia by preventing VSMC proliferation after stenting; 2) Fe3O4 exerted inhibitory effects on VSMCs by downregulating proliferative genes such as SOX9, EGR4, and TGFB1, but upregulated inhibitory genes such as DNMT1, TIMP3, and PCNA; 3) Fe3O4 inhibited VSMCs by preventing phenotypic transformation from the contractile to the synthetic phase; and 4) Fe3O4-coated stents achieved satisfactory hemocompatibility in a rabbit model. Our study highlights the additional benefits of Fe3O4 particles in inhibiting VSMC proliferation, indicating that Fe3O4 coated stent potentially served as an attractive therapeutic approach for ISR prevention.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Jiabing Huang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, PR China
| | - Changqing Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Yanbing Wen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| |
Collapse
|
13
|
Lu Z, Zhu S, Wu Y, Xu X, Li S, Huang Q. Circ_0008571 modulates the phenotype of vascular smooth muscle cells by targeting miR-145-5p in intracranial aneurysms. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167278. [PMID: 38834101 DOI: 10.1016/j.bbadis.2024.167278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The dysfunction of human vascular smooth cells (hVSMCs) is significantly connected to the development of intracranial aneurysms (IAs). By suppressing the activity of microRNAs (miRNAs), circular RNAs (circRNAs) participate in IA pathogenesis. Nevertheless, the role of hsa_circ_0008571 in IAs remains unclear. METHODS circRNA sequencing was used to identify circRNAs from human IA tissues. To determine the function of circ_0008571, Transwell, wound healing, and cell proliferation assays were conducted. To identify the target of circ_0008571, the analyses of CircInteractome and TargetScan, as well as the luciferase assay were carried out. Furthermore, circ_0008571 knockdown and over-expression were performed to investigate its functions in IA development and the underlying molecular mechanisms. RESULTS Both hsa_circ_0008571 and Integrin beta 8 (ITGB8) were downregulated, while miR-145-5p transcription was elevated in the aneurysm wall of IAs patients compared to superficial temporal artery tissues. In vitro, cell migration and growth were dramatically suppressed after hsa_circ_0008571 overexpression. Mechanistically, has_circ_0008571 could suppress miR-145-5p activity by direct sponging. Moreover, we found that ITGB8 expression and the activation of the TGF-β-mediated signaling pathway were significantly enhanced. CONCLUSION The hsa_circ_0008571-miR-145-5p-ITGB8 axis plays an essential role in IA progression.
Collapse
Affiliation(s)
- Zhiwen Lu
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China; Department of Neurosurgery, Naval Medical Center, The PLA Naval Medical University, Shanghai 200052, China
| | - Shijie Zhu
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yina Wu
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaolong Xu
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Siqi Li
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qinghai Huang
- Department of Neurovascular Centre, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
14
|
Prochilo G, Pfeffer A, Du S, Kaneko N, Liebeskind DS, Hinman JD. Recent Translational Research Models of Intracranial Atherosclerotic Disease. Stroke 2024; 55:1707-1719. [PMID: 38738375 DOI: 10.1161/strokeaha.124.044520] [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] [Indexed: 05/14/2024]
Abstract
Intracranial atherosclerotic disease (ICAD) is a leading cause of ischemic stroke worldwide. However, research on the pathophysiology of ICAD is scarce due to the relative inaccessibility of histology samples and the lack of comprehensive experimental models. As a result, much of the current understanding of ICAD relies on research on extracranial atherosclerosis. This approach is problematic as intracranial and extracranial arteries are anatomically, structurally, physiologically, and metabolically distinct, indicating that intracranial and extracranial atherosclerosis likely develop through different biologic pathways. The current standard of care for ICAD treatment relies predominantly on therapeutics developed to treat extracranial atherosclerosis and is insufficient given the alarmingly high risk of stroke. To provide a definitive treatment for the disease, a deeper understanding of the pathophysiology underlying ICAD is specifically required. True mechanistic understanding of disease pathogenesis is only possible using robust experimental models. In this review, we aim to identify the advantages and limitations of the existing in vivo and in vitro models of ICAD and basic atherosclerotic processes, which may be used to inform better models of ICAD in the future and drive new therapeutic strategies to reduce stroke risk.
Collapse
Affiliation(s)
- Grace Prochilo
- Departments of Neurology (G.P., A.P., S.D., D.S.L., J.D.H.), David Geffen School of Medicine, University of California, Los Angeles
| | - Alissa Pfeffer
- Departments of Neurology (G.P., A.P., S.D., D.S.L., J.D.H.), David Geffen School of Medicine, University of California, Los Angeles
| | - Stephanie Du
- Departments of Neurology (G.P., A.P., S.D., D.S.L., J.D.H.), David Geffen School of Medicine, University of California, Los Angeles
| | - Naoki Kaneko
- Radiology (N.K.), David Geffen School of Medicine, University of California, Los Angeles
| | - David S Liebeskind
- Departments of Neurology (G.P., A.P., S.D., D.S.L., J.D.H.), David Geffen School of Medicine, University of California, Los Angeles
| | - Jason D Hinman
- Departments of Neurology (G.P., A.P., S.D., D.S.L., J.D.H.), David Geffen School of Medicine, University of California, Los Angeles
- Department of Neurology, Department of Veterans Affairs Medical Center, Los Angeles, CA (J.D.H.)
| |
Collapse
|
15
|
Yin C, Ge Z, Yuan J, Chen Y, Tang Y, Xiang Y, Zhang Y. NEAT1 regulates VSMC differentiation and calcification in as long noncoding RNA NEAT1 enhances phenotypic and osteogenic switching of vascular smooth muscle cells in atherosclerosis via scaffolding EZH2. Am J Physiol Cell Physiol 2024; 326:C1721-C1734. [PMID: 38646788 PMCID: PMC11371316 DOI: 10.1152/ajpcell.00587.2023] [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: 11/02/2023] [Revised: 03/27/2024] [Accepted: 04/11/2024] [Indexed: 04/23/2024]
Abstract
Atherosclerosis (AS) is a significant contributor to cardio-cerebrovascular ischemia diseases, resulting in high mortality rates worldwide. During AS, vascular smooth muscle cells (VSMCs) play a crucial role in plaque formation by undergoing phenotypic and osteogenic switching. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has previously been identified as a nuclear regulator that promotes tumorigenesis and metastasis, but its role in regulating VSMCs in AS remains unclear. Our study aimed to investigate the biological functions and specific mechanisms of NEAT1 in regulating VSMCs in AS. We found that NEAT1 was upregulated in the aortas of AS mouse models and dedifferentiated primary VSMCs. Silencing NEAT1 in vitro attenuated the proliferation, migration, and osteogenic differentiation of VSMCs, while NEAT1 overexpression had the opposite effect. Furthermore, NEAT1 promoted VSMC osteogenic differentiation and vascular calcification in both in vivo and in vitro vascular calcification models. We also discovered that NEAT1 directly activates enhancer of zeste homolog 2 (EZH2), an epigenetic enzyme that suppresses the expression of senescence- and antimigration-related genes, by translocating it into the nucleus. CUT&Tag assay revealed that NEAT1 guides EZH2 to the promoters of senescence-related genes (P16, P21, and TIMP3), methylating local histones to reduce their transcription. Our findings suggest that NEAT1 functions in AS by modulating the epigenetic function of EZH2, which enhances the proliferation, migration, and osteogenic differentiation of VSMCs. This study provides new insights into the molecular mechanisms underlying the pathogenesis of AS and highlights the potential of NEAT1 as a therapeutic target of AS.NEW & NOTEWORTHY Our study demonstrates that the upregulation of long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) promotes proliferation and migration during phenotypic switching of vascular smooth muscle cells in atherosclerosis. We also provide in vivo and in vitro evidence that NEAT1 accelerates vascular calcification. Our findings identified the direct interaction between enhancer of zeste homolog 2 (EZH2) and NEAT1 during atherosclerosis. NEAT1 is necessary for EZH2 to translocate from the cytoplasm to the nucleus, where EZH2 epigenetically inhibits the expression of genes related to senescence and antimigration.
Collapse
MESH Headings
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/genetics
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Osteogenesis/genetics
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Differentiation
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Mice
- Male
- Mice, Inbred C57BL
- Cell Proliferation
- Phenotype
- Cells, Cultured
- Humans
- Cell Movement
Collapse
Affiliation(s)
- Chengye Yin
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhuowang Ge
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiali Yuan
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yuhan Chen
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yong Tang
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yin Xiang
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yachen Zhang
- Department of Cardiology, Xinhua HospitalShanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
16
|
Ma W, Liu Z, Zhu T, Wang L, Du J, Wang K, Xu C. Fabric-Enhanced Vascular Graft with Hierarchical Structure for Promoting the Regeneration of Vascular Tissue. Adv Healthc Mater 2024; 13:e2302676. [PMID: 38279911 DOI: 10.1002/adhm.202302676] [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: 08/14/2023] [Revised: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Natural blood vessels have completed functions, including elasticity, compliance, and excellent antithrombotic properties because of their mature structure. To replace damaged blood vessels, vascular grafts should perform these functions by simulating the natural vascular structures. Although the structures of natural blood vessels are thoroughly explored, constructing a small-diameter vascular graft that matches the mechanical and biological properties of natural blood vessels remains a challenge. A hierarchical vascular graft is fabricated by Electrospinning, Braiding, and Thermally induced phase separation (EBT) processes, which could simulate the structure of natural blood vessels. The internal electrospun structure facilitates the adhesion of endothelial cells, thereby accelerating endothelialization. The intermediate PLGA fabric exhibits excellent mechanical properties, which allow it to maintain its shape during long-term transplantation and prevent graft expansion. The external macroporous structure is beneficial for cell growth and infiltration. Blood vessel remodeling aims to combine a structure that promotes tissue regeneration with anti-inflammatory materials. The results in vitro demonstrated that it EBT vascular graft (EBTVG) has matched the mechanical properties, reliable cytocompatibility, and the strongest endothelialization in situ. The results in vitro and replacement of the resected artery in vivo suggest that the EBTVG combines different structural advantages with biomechanical properties and reliable biocompatibility, significantly promoting the stabilization and regeneration of vascular endothelial cells and vascular smooth muscle cells, as well as stabilizing the blood microenvironment.
Collapse
Affiliation(s)
- Wenxin Ma
- Multidisciplinary Centre for Advanced Materials, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, P. R. China
- School of Textiles and Fashion, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, P. R. China
| | - Zhuo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Xueyuan Rd., Shanghai, 200032, P. R. China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, P. R. China
| | - Liming Wang
- School of Textiles and Fashion, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, P. R. China
| | - Juan Du
- Multidisciplinary Centre for Advanced Materials, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, P. R. China
| | - Kun Wang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Key Laboratory of Metabolism and Gastrointestinal Tumors, the First Affiliated Hospital of Shandong First Medical University, Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, Shandong Medicine and Health Key Laboratory of General Surgery, 16766 Jingshi Rd., Jinan, 250014, P. R. China
| | - Chen Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Xueyuan Rd., Shanghai, 200032, P. R. China
| |
Collapse
|
17
|
Tian Y, Luan X, Yang K. Chronotherapy involving rosiglitazone regulates the phenotypic switch of vascular smooth muscle cells by shifting the phase of TNF-α rhythm through triglyceride accumulation in macrophages. Heliyon 2024; 10:e30708. [PMID: 38803898 PMCID: PMC11128472 DOI: 10.1016/j.heliyon.2024.e30708] [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: 12/14/2023] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives Vascular diseases are often caused by the interaction between macrophages and vascular smooth muscle cells (VSMCs). This study aims to elucidate whether chronotherapy with rosiglitazone (RSG) can regulate the secretion rhythm of macrophages, thereby controlling the phenotypic switch of VSMCs and clarifying the potential molecular mechanisms, providing a chronotherapeutic approach for the treatment of vascular diseases. Methods RAW264.7 cells and A7r5 cells were synchronized via a 50 % FBS treatment. M1-type macrophages were induced through Lipopolysaccharide (LPS) exposure. Additionally, siRNA and plasmids targeting PPARγ were transfected into macrophages. The assessment encompassed cell viability, migration, inflammatory factor levels, lipid metabolites, clock gene expression, and relative protein expression. Results We revealed that, in alignment with core clock genes Bmal1 and CLOCK, RSG administration at ZT2 advanced the phase of TNF-α release rhythm, while ZT12 administration shifted it backward. Incubation with TNF-α at ZT2 significantly promoted the phenotype switch of VSMCs. This effect diminished when incubated at ZT12, implicating the involvement of the clock-MAPK pathway in VSMCs. Furthermore, RSG administration at ZT2 advanced the phases of PPARγ and Bmal1 genes, whereas ZT12 administration shifted them backward. Additionally, PPARγ overexpression significantly induced triglyceride (TG) accumulation in macrophages. Exogenous TG upregulated Bmal1 and CLOCK gene expression in macrophages and significantly increased TNF-α release. Conclusion Chronotherapy involving RSG induces TG accumulation within macrophages, resulting in alterations in circadian gene rhythms. These changes, in turn, modulate the phase of rhythmic TNF-α release and play a regulatory role in VSMCs phenotype switch. Our study establishes a theoretical foundation for chronotherapy of PPARγ agonists.
Collapse
Affiliation(s)
- Yu Tian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241001. PR China
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, 241001, PR China
| | - Xuanyu Luan
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Kui Yang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, 241001, PR China
| |
Collapse
|
18
|
Xin Y, Zhang Z, Lv S, Xu S, Liu A, Li H, Li P, Han H, Liu Y. Elucidating VSMC phenotypic transition mechanisms to bridge insights into cardiovascular disease implications. Front Cardiovasc Med 2024; 11:1400780. [PMID: 38803664 PMCID: PMC11128571 DOI: 10.3389/fcvm.2024.1400780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, despite advances in understanding cardiovascular health. Significant barriers still exist in effectively preventing and managing these diseases. Vascular smooth muscle cells (VSMCs) are crucial for maintaining vascular integrity and can switch between contractile and synthetic functions in response to stimuli such as hypoxia and inflammation. These transformations play a pivotal role in the progression of cardiovascular diseases, facilitating vascular modifications and disease advancement. This article synthesizes the current understanding of the mechanisms and signaling pathways regulating VSMC phenotypic transitions, highlighting their potential as therapeutic targets in cardiovascular disease interventions.
Collapse
Affiliation(s)
- Yuning Xin
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zipei Zhang
- Traditional Chinese Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Shan Lv
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Shan Xu
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Aidong Liu
- Traditional Chinese Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Li
- Traditional Chinese Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Pengfei Li
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Huize Han
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yinghui Liu
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
19
|
Yuan X, Jiang C, Xue Y, Guo F, Luo M, Guo L, Gao Y, Yuan T, Xu H, Chen H. KLF13 promotes VSMCs phenotypic dedifferentiation by directly binding to the SM22α promoter. J Cell Physiol 2024; 239:e31251. [PMID: 38634445 DOI: 10.1002/jcp.31251] [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: 10/11/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
Krüppel-like factor 13 (KLF13), a zinc finger transcription factor, is considered as a potential regulator of cardiomyocyte differentiation and proliferation during heart morphogenesis. However, its precise role in the dedifferentiation of vascular smooth muscle cells (VSMCs) during atherosclerosis and neointimal formation after injury remains poorly understood. In this study, we investigated the relationship between KLF13 and SM22α expression in normal and atherosclerotic plaques by bioanalysis, and observed a significant increase in KLF13 levels in the atherosclerotic plaques of both human patients and ApoE-/- mice. Knockdown of KLF13 was found to ameliorate intimal hyperplasia following carotid artery injury. Furthermore, we discovered that KLF13 directly binds to the SM22α promoter, leading to the phenotypic dedifferentiation of VSMCs. Remarkably, we observed a significant inhibition of platelet-derived growth factor BB-induced VSMCs dedifferentiation, proliferation, and migration when knocked down KLF13 in VSMCs. This inhibitory effect of KLF13 knockdown on VCMC function was, at least in part, mediated by the inactivation of p-AKT signaling in VSMCs. Overall, our findings shed light on a potential therapeutic target for treating atherosclerotic lesions and restenosis after vascular injury.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Cell Dedifferentiation
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cells, Cultured
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice, Inbred C57BL
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Neointima/genetics
- Phenotype
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/genetics
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Cell Cycle Proteins
- Microfilament Proteins/genetics
Collapse
Affiliation(s)
- Xiaofan Yuan
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chuan Jiang
- Department of Neurosurgery, The Southwest Medical University, Luzhou, Sichuan, China
| | - Yuzhou Xue
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Fuqiang Guo
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Guo
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yang Gao
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tongling Yuan
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hui Xu
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hong Chen
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Camarda ND, Ibarrola J, Biwer LA, Jaffe IZ. Mineralocorticoid Receptors in Vascular Smooth Muscle: Blood Pressure and Beyond. Hypertension 2024; 81:1008-1020. [PMID: 38426347 PMCID: PMC11023801 DOI: 10.1161/hypertensionaha.123.21358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
After half a century of evidence suggesting the existence of mineralocorticoid receptors (MR) in the vasculature, the advent of technology to specifically knockout the MR from smooth muscle cells (SMCs) in mice has elucidated contributions of SMC-MR to cardiovascular function and disease, independent of the kidney. This review summarizes the latest understanding of the molecular mechanisms by which SMC-MR contributes to (1) regulation of vasomotor function and blood pressure to contribute to systemic and pulmonary hypertension; (2) vascular remodeling in response to hypertension, vascular injury, obesity, and aging, and the impact on vascular calcification; and (3) cardiovascular pathologies including aortic aneurysm, heart valve dysfunction, and heart failure. Data are reviewed from in vitro studies using SMCs and in vivo findings from SMC-specific MR-knockout mice that implicate target genes and signaling pathways downstream of SMC-MR. By regulating expression of the L-type calcium channel subunit Cav1.2 and angiotensin II type-1 receptor, SMC-MR contributes to myogenic tone and vasoconstriction, thereby contributing to systemic blood pressure. MR activation also promotes SMC proliferation, migration, production and degradation of extracellular matrix, and osteogenic differentiation by regulating target genes including connective tissue growth factor, osteopontin, bone morphogenetic protein 2, galectin-3, and matrix metallopeptidase-2. By these mechanisms, SMC-MR promotes disease progression in models of aging-associated vascular stiffness, vascular calcification, mitral and aortic valve disease, pulmonary hypertension, and heart failure. While rarely tested, when sexes were compared, the mechanisms of SMC-MR-mediated disease were sexually dimorphic. These advances support targeting SMC-MR-mediated mechanisms to prevent and treat diverse cardiovascular disorders.
Collapse
Affiliation(s)
- Nicholas D. Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Lauren A. Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
21
|
Zhao Y, Wang Z, Chen Y, Feng M, Liu X, Chen H, Wang N, Wang Z, Cao S, Ren J, Liu X, Zhao Y, Zhang Y. Asprosin aggravates atherosclerosis via regulating the phenotype transformation of vascular smooth muscle cells. Int J Biol Macromol 2024; 268:131868. [PMID: 38677690 DOI: 10.1016/j.ijbiomac.2024.131868] [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: 03/09/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Phenotype transformation of vascular smooth muscle cells (VSMCs) plays an important role in the development of atherosclerosis. Asprosin is a newly discovered adipokine, which is critical in regulating metabolism. However, the relationship between asprosin and phenotype transformation of VSMCs in atherosclerosis remains unclear. The aim of this study is to investigate whether asprosin affects the progression of atherosclerosis by inducing phenotype transformation of VSMCs. We established an atherosclerosis model in ApoE-/- mice and administered asprosin recombinant protein and asprosin antibody to mice. Knocking down asprosin was also as an intervention. Interestingly, we found a correlation between asprosin levels and atherosclerosis. Asprosin promoted plaque formation and phenotype transformation of VSMCs. While, AspKD or asprosin antibody reduced the plaque lesion and suppressed vascular stiffness in ApoE-/- mice. Mechanistically, asprosin induced phenotype transformation of MOVAs by binding to GPR54, leading to Gαq/11 recruitment and activation of the PLC-PKC-ERK1/2-STAT3 signaling pathway. Si GPR54 or GPR54 antagonist partially inhibited the action of asprosin in MOVAs. Mutant GPR54-(267, 307) residue cancelled the binding of asprosin and GPR54. In summary, this study confirmed asprosin activated GPR54/Gαq/11-dependent ERK1/2-STAT3 signaling pathway, thereby promoting VSMCs phenotype transformation and aggravating atherosclerosis, thus providing a new target for the treatment of atherosclerosis.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Phenotype
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Fibrillin-1/metabolism
- Fibrillin-1/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Male
- Signal Transduction
- Disease Models, Animal
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Humans
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Mice, Knockout
Collapse
Affiliation(s)
- Yu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China; Department of Pathophysiology, Province Key Laboratory of Medicine-Food Homologous Resources and Prevention and Treatment of Metabolic Diseases, Basic Medical College, Qiqihar Medical University, Qiqihar 161000, China
| | - Zhengkai Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yi Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Min Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinxin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Huan Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Nannan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Zhiqi Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shifeng Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jing Ren
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xue Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yixiu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Yan Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
22
|
Teng L, Qin Q, Zhou ZY, Zhou F, Cao CY, He C, Ding JW, Yang J. Role of C/EBP Homologous Protein in Vascular Stenosis After Carotid Artery Injury. Biochem Genet 2024:10.1007/s10528-024-10713-9. [PMID: 38526708 DOI: 10.1007/s10528-024-10713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/22/2024] [Indexed: 03/27/2024]
Abstract
The study aims to explore the fluctuating expression of C/EBP Homologous Protein (CHOP) following rat carotid artery injury and its central role in vascular stenosis. Using in vivo rat carotid artery injury models and in vitro ischemia and hypoxia cell models employing human aortic endothelial cells (HAECs) and vascular smooth muscle cells (T/G HA-VSMCs), a comprehensive investigative framework was established. Histological analysis confirmed intimal hyperplasia in rat models. CHOP expression in vascular tissues was assessed using Western blot and immunohistochemical staining, and its presence in HAECs and T/G HA-VSMCs was determined through RT-PCR and Western blot. The study evaluated HAEC apoptosis, inflammatory cytokine secretion, cell proliferation, and T/G HA-VSMCs migration through Western blot, ELISA, CCK8, and Transwell migration assays. The rat carotid artery injury model revealed substantial fibrous plaque formation and vascular stenosis, resulting in an increased intimal area and plaque-to-lumen area ratio. Notably, CHOP is markedly elevated in vessels of the carotid artery injury model compared to normal vessels. Atorvastatin effectively mitigated vascular stenosis and suppresses CHOP protein expression. In HAECs, ischemia and hypoxia-induced CHOP upregulation, along with heightened TNFα, IL-6, caspase3, and caspase8 levels, while reducing cell proliferation. Atorvastatin demonstrated a dose-dependent suppression of CHOP expression in HAECs. Downregulation of CHOP or atorvastatin treatment led to reduced IL-6 and TNFα secretion, coupled with augmented cell proliferation. Similarly, ischemia and hypoxia conditions increased CHOP expression in T/G HA-VSMCs, which was concentration-dependently inhibited by atorvastatin. Furthermore, significantly increased MMP-9 and MMP-2 concentrations in the cell culture supernatant correlated with enhanced T/G HA-VSMCs migration. However, interventions targeting CHOP downregulation and atorvastatin usage curtailed MMP-9 and MMP-2 secretion and suppressed cell migration. In conclusion, CHOP plays a crucial role in endothelial injury, proliferation, and VSMCs migration during carotid artery injury, serving as a pivotal regulator in post-injury fibrous plaque formation and vascular remodeling. Statins emerge as protectors of endothelial cells, restraining VSMCs migration by modulating CHOP expression.
Collapse
Affiliation(s)
- Lin Teng
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, SE5 9NU, UK
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Zi-Yi Zhou
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Cun-Yu Cao
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
- Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Chao He
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Jia-Wang Ding
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Jian Yang
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China.
| |
Collapse
|
23
|
Sun X, Yang Y, Zhao W, Wang M, Chen Y, Wang J, Yang D, Yang Y. MTMR7 suppresses the phenotypic switching of vascular smooth muscle cell and vascular intimal hyperplasia after injury via regulating p62/mTORC1-mediated glucose metabolism. Atherosclerosis 2024; 390:117470. [PMID: 38342025 DOI: 10.1016/j.atherosclerosis.2024.117470] [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: 09/24/2023] [Revised: 12/24/2023] [Accepted: 01/30/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND AND AIMS Myotubularin-related protein 7 (MTMR7) suppresses proliferation in various cell types and is associated with cardiovascular and cerebrovascular diseases. However, whether MTMR7 regulates vascular smooth muscle cell (VSMC) and vascular intimal hyperplasia remains unclear. We explored the role of MTMR7 in phenotypic switching of VSMC and vascular intimal hyperplasia after injury. METHODS AND RESULTS MTMR7 expression was significantly downregulated in injured arteries. Compared to wild type (WT) mice, Mtmr7-transgenic (Mtmr7-Tg) mice showed reduced intima/media ratio, decreased percentage of Ki-67-positive cells within neointima, and increased Calponin expression in injured artery. In vitro, upregulating MTMR7 by Len-Mtmr7 transfection inhibited platelet derived growth factor (PDGF)-BB-induced proliferation, migration of VSMC and reversed PDGF-BB-induced decrease in expression of Calponin and SM-MHC. Microarray, single cell sequence, and other bioinformatics analysis revealed that MTMR7 is highly related to glucose metabolism and mammalian target of rapamycin complex 1 (mTORC1). Further experiments confirmed that MTMR7 markedly repressed glycolysis and mTORC1 activity in PDGF-BB-challenged VSMC in vitro. Restoring mTORC1 activity abolished MTMR7-mediated suppression of glycolysis, phenotypic shift in VSMC in vitro and protection against vascular intimal hyperplasia in vivo. Furthermore, upregulating MTMR7 in vitro led to dephosphorylation and dissociation of p62 from mTORC1 in VSMC. External expression of p62 in vitro also abrogated the inhibitory effects of MTMR7 on glycolysis and phenotypic switching in PDGF-BB-stimulated VSMC. CONCLUSIONS Our study demonstrates that MTMR7 inhibits injury-induced vascular intimal hyperplasia and phenotypic switching of VSMC. Mechanistically, the beneficial effects of MTMR7 are conducted via suppressing p62/mTORC1-mediated glycolysis.
Collapse
Affiliation(s)
- Xiongshan Sun
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yao Yang
- From the Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Weiwei Zhao
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Mingliang Wang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yingmei Chen
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Jia Wang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China.
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China.
| |
Collapse
|
24
|
Hu RT, Deng HW, Teng WB, Zhou SD, Ye ZM, Dong ZM, Qin C. ADORA3: A Key Player in the Pathogenesis of Intracranial Aneurysms and a Potential Diagnostic Biomarker. Mol Diagn Ther 2024; 28:225-235. [PMID: 38341835 DOI: 10.1007/s40291-024-00694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND The effects of genes on the development of intracranial aneurysms (IAs) remain to be elucidated, and reliable blood biomarkers for diagnosing IAs are yet to be established. This study aimed to identify genes associated with IAs pathogenesis and explore their diagnostic value by analyzing IAs datasets, conducting vascular smooth muscle cells (VSMC) experiments, and performing blood detection. METHODS IAs datasets were collected and the differentially expressed genes were analyzed. The selected genes were validated in external datasets. Autophagy was induced in VSMC and the effect of selected genes was determined. The diagnostic value of selected gene on the IAs were explored using area under curve (AUC) analysis using IAs plasma samples. RESULTS Analysis of 61 samples (32 controls and 29 IAs tissues) revealed a significant increase in expression of ADORA3 compared with normal tissues using empirical Bayes methods of "limma" package; this was further validated by two external datasets. Additionally, induction of autophagy in VSMC lead to upregulation of ADORA3. Conversely, silencing ADORA3 suppressed VSMC proliferation and autophagy. Furthermore, analysis of an IAs blood sample dataset and clinical plasma samples demonstrated increased ADORA3 expression in patients with IA compared with normal subjects. The diagnostic value of blood ADORA3 expression in IAs was moderate when analyzing clinical samples (AUC: 0.756). Combining ADORA3 with IL2RB or CCR7 further enhanced the diagnostic ability for IAs, with the AUC value over 0.83. CONCLUSIONS High expression of ADORA3 is associated with IAs pathogenesis, likely through its promotion of VSMC autophagy. Furthermore, blood ADORA3 levels have the potential to serve as an auxiliary diagnostic biomarker for IAs.
Collapse
Affiliation(s)
- Rui-Ting Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Hao-Wei Deng
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Wen-Bin Teng
- Department of Neurology, Minzu Hospital of Guangxi Medical University, Nanning, 530001, China
| | - Shao-Dan Zhou
- Department of Neurology, Minzu Hospital of Guangxi Medical University, Nanning, 530001, China
| | - Zi-Ming Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Zi-Mei Dong
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Chao Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
25
|
Baldwin CS, Iyer S, Rao RR. The challenges and prospects of smooth muscle tissue engineering. Regen Med 2024; 19:135-143. [PMID: 38440898 PMCID: PMC10941056 DOI: 10.2217/rme-2023-0230] [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: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
Many vascular disorders arise as a result of dysfunctional smooth muscle cells. Tissue engineering strategies have evolved as key approaches to generate functional vascular smooth muscle cells for use in cell-based precision and personalized regenerative medicine approaches. This article highlights some of the challenges that exist in the field and presents some of the prospects for translating research advancements into therapeutic modalities. The article emphasizes the need for better developing synergetic intracellular and extracellular cues in the processes to generate functional vascular smooth muscle cells from different stem cell sources for use in tissue engineering strategies.
Collapse
Affiliation(s)
- Christofer S Baldwin
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Shilpa Iyer
- Department of Biological Sciences, Fulbright College of Arts & Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Raj R Rao
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
26
|
Tian Q, Chen JH, Ding Y, Wang XY, Qiu JY, Cao Q, Zhuang LL, Jin R, Zhou GP. EGR1 transcriptionally regulates SVEP1 to promote proliferation and migration in human coronary artery smooth muscle cells. Mol Biol Rep 2024; 51:365. [PMID: 38409611 DOI: 10.1007/s11033-024-09322-x] [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: 10/29/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
A low-frequency variant of sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SVEP1) is associated with the risk of coronary artery disease, as determined by a genome-wide association study. SVEP1 induces vascular smooth muscle cell proliferation and an inflammatory phenotype to promote atherosclerosis. In the present study, qRT‒PCR demonstrated that the mRNA expression of SVEP1 was significantly increased in atherosclerotic plaques compared to normal tissues. Bioinformatics revealed that EGR1 was a transcription factor for SVEP1. The results of the luciferase reporter assay, siRNA interference or overexpression assay, mutational analysis and ChIP confirmed that EGR1 positively regulated the transcriptional activity of SVEP1 by directly binding to its promoter. EGR1 promoted human coronary artery smooth muscle cell (HCASMC) proliferation and migration via SVEP1 in response to oxidized low-density lipoprotein (ox-LDL) treatment. Moreover, the expression level of EGR1 was increased in atherosclerotic plaques and showed a strong linear correlation with the expression of SVEP1. Our findings indicated that EGR1 binding to the promoter region drive SVEP1 transcription to promote HCASMC proliferation and migration.
Collapse
Affiliation(s)
- Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Yu Wang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Yun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Dai Y, Xu R, Yang K, Jiang T, Wei Y, Liu Y, Chen W, Fu Y, Zhao Y. Effect of tissue factor pathway inhibitor on the pyroptosis of vascular smooth muscle cells induced by angiotensin II. Cardiovasc Diagn Ther 2024; 14:72-83. [PMID: 38434568 PMCID: PMC10904294 DOI: 10.21037/cdt-23-355] [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: 08/25/2023] [Accepted: 12/08/2023] [Indexed: 03/05/2024]
Abstract
Background In recent years, a mass of studies have shown that pyroptosis plays an important role in the proliferation of vascular smooth muscle cells (VSMCs). We investigated whether angiotensin II (Ang II) induces the pyroptosis of rat aortic VSMCs and the role of NOD-like receptor family pyrin domain containing 3 (NLRP3) in this process. Additionally, we explored the effect and related mechanism of recombinant tissue factor pathway inhibitor (rTFPI) in Ang II-induced VSMC pyroptosis. Methods Cultured VSMCs were divided into five groups: control group, Ang II group (1×10-5 mol/L), MCC950 group (NLRP3 inhibitor, 15 nmol/L), Ang II + MCC950 group and Ang II + rTFPI (50 µg/L) group. Cell viability was measured by cell counting kit-8 (CCK8) assays and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays. Propidium iodide (PI) staining and immunofluorescence were performed to determine the pyroptosis of VSMCs. Changes in VSMC ultrastructure were evaluated through transmission electron microscopy. The expression levels of NLRP3, pro-caspase-1, gasdermin D-N (GSDMD-N), and interleukin-1β (IL-1β) were determined by western blot analysis. Results The cell viability, the positive rate of PI staining, and the expression level of GSDMD detected by immunofluorescence in the Ang II group were higher than that in the control group, whereas they all decreased in Ang II + MCC950 group and Ang II + rTFPI group compared with Ang II group (P<0.05). Electron microscopy analysis revealed less extracellular matrix, increased myofilaments, and decreased endoplasmic reticulum, Golgi complex, and mitochondria in Ang II + rTFPI-treated VSMCs than in Ang II-treated VSMCs. The protein expression levels of the pyroptosis-related molecules NLRP3, pro-caspase-1, GSDMD-N, and IL-1β in Ang II group showed an increasing trend compared with those in control group (P<0.05); however, these expression levels in Ang II + MCC950 and Ang II + rTFPI groups were significantly lower than those in Ang II group (P<0.05). Conclusions Ang II may induce pyroptosis in VSMCs by activating NLRP3. rTFPI can inhibit Ang II-induced VSMC pyroptosis. Furthermore, rTFPI might exert this effect by inhibiting the NLRP3 pathway and therefore play an important role in the treatment of vascular remodeling induced by hypertension.
Collapse
Affiliation(s)
- Yue Dai
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Xu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kelaier Yang
- Department of Endocrinology, Shenzhen University General Hospital, Shenzhen, China
| | - Tingting Jiang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongkang Wei
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
28
|
Ibarrola J, Jaffe IZ. The Mineralocorticoid Receptor in the Vasculature: Friend or Foe? Annu Rev Physiol 2024; 86:49-70. [PMID: 37788489 DOI: 10.1146/annurev-physiol-042022-015223] [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] [Indexed: 10/05/2023]
Abstract
Originally described as the renal aldosterone receptor that regulates sodium homeostasis, it is now clear that mineralocorticoid receptors (MRs) are widely expressed, including in vascular endothelial and smooth muscle cells. Ample data demonstrate that endothelial and smooth muscle cell MRs contribute to cardiovascular disease in response to risk factors (aging, obesity, hypertension, atherosclerosis) by inducing vasoconstriction, vascular remodeling, inflammation, and oxidative stress. Extrapolating from its role in disease, evidence supports beneficial roles of vascular MRs in the context of hypotension by promoting inflammation, wound healing, and vasoconstriction to enhance survival from bleeding or sepsis. Advances in understanding how vascular MRs become activated are also reviewed, describing transcriptional, ligand-dependent, and ligand-independent mechanisms. By synthesizing evidence describing how vascular MRs convert cardiovascular risk factors into disease (the vascular MR as a foe), we postulate that the teleological role of the MR is to coordinate responses to hypotension (the MR as a friend).
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| |
Collapse
|
29
|
Su C, Liu M, Yao X, Hao W, Ma J, Ren Y, Gao X, Xin L, Ge L, Yu Y, Wei M, Yang J. Vascular injury activates the ELK1/SND1/SRF pathway to promote vascular smooth muscle cell proliferative phenotype and neointimal hyperplasia. Cell Mol Life Sci 2024; 81:59. [PMID: 38279051 PMCID: PMC10817852 DOI: 10.1007/s00018-023-05095-x] [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: 08/15/2023] [Revised: 12/01/2023] [Accepted: 12/15/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) proliferation is the leading cause of vascular stenosis or restenosis. Therefore, investigating the molecular mechanisms and pivotal regulators of the proliferative VSMC phenotype is imperative for precisely preventing neointimal hyperplasia in vascular disease. METHODS Wire-induced vascular injury and aortic culture models were used to detect the expression of staphylococcal nuclease domain-containing protein 1 (SND1). SMC-specific Snd1 knockout mice were used to assess the potential roles of SND1 after vascular injury. Primary VSMCs were cultured to evaluate SND1 function on VSMC phenotype switching, as well as to investigate the mechanism by which SND1 regulates the VSMC proliferative phenotype. RESULTS Phenotype-switched proliferative VSMCs exhibited higher SND1 protein expression compared to the differentiated VSMCs. This result was replicated in primary VSMCs treated with platelet-derived growth factor (PDGF). In the injury model, specific knockout of Snd1 in mouse VSMCs reduced neointimal hyperplasia. We then revealed that ETS transcription factor ELK1 (ELK1) exhibited upregulation and activation in proliferative VSMCs, and acted as a novel transcription factor to induce the gene transcriptional activation of Snd1. Subsequently, the upregulated SND1 is associated with serum response factor (SRF) by competing with myocardin (MYOCD). As a co-activator of SRF, SND1 recruited the lysine acetyltransferase 2B (KAT2B) to the promoter regions leading to the histone acetylation, consequently promoted SRF to recognize the specific CArG motif, and enhanced the proliferation- and migration-related gene transcriptional activation. CONCLUSIONS The present study identifies ELK1/SND1/SRF as a novel pathway in promoting the proliferative VSMC phenotype and neointimal hyperplasia in vascular injury, predisposing the vessels to pathological remodeling. This provides a potential therapeutic target for vascular stenosis.
Collapse
Affiliation(s)
- Chao Su
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Mingxia Liu
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xuyang Yao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
- Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Wei Hao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Jinzheng Ma
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Ren
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xingjie Gao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Lingbiao Xin
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Lin Ge
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Minxin Wei
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| | - Jie Yang
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China.
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), and Key Laboratory of Cellular and Molecular Immunology, Tianjin, China.
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
- State Key Laboratory of Experimental Hematology, Tianjin, China.
| |
Collapse
|
30
|
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
|
31
|
Chen X, Chen X. The Role of TRIM Proteins in Vascular Disease. Curr Vasc Pharmacol 2024; 22:11-18. [PMID: 38031766 DOI: 10.2174/0115701611241848231114111618] [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: 01/01/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
There are more than 80 different tripartite motifs (TRIM) proteins within the E3 ubiquitin ligase subfamily, including proteins that regulate intracellular signaling, apoptosis, autophagy, proliferation, inflammation, and immunity through the ubiquitination of target proteins. Studies conducted in recent years have unraveled the importance of TRIM proteins in the pathophysiology of vascular diseases. In this review, we describe the effects of TRIM proteins on vascular endothelial cells, smooth muscle cells, heart, and lungs. In particular, we discuss the potential mechanisms by which TRIMs regulate diseases and shed light on the potential therapeutic applications of TRIMs.
Collapse
Affiliation(s)
- Xinxin Chen
- Ophthalmology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| | - Xiaolong Chen
- Ophthalmology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| |
Collapse
|
32
|
Hamdin CD, Wu ML, Chen CM, Ho YC, Jiang WC, Gung PY, Ho HH, Chuang HC, Tan TH, Yet SF. Dual-Specificity Phosphatase 6 Deficiency Attenuates Arterial-Injury-Induced Intimal Hyperplasia in Mice. Int J Mol Sci 2023; 24:17136. [PMID: 38138967 PMCID: PMC10742470 DOI: 10.3390/ijms242417136] [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: 10/25/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
In response to injury, vascular smooth muscle cells (VSMCs) of the arterial wall dedifferentiate into a proliferative and migratory phenotype, leading to intimal hyperplasia. The ERK1/2 pathway participates in cellular proliferation and migration, while dual-specificity phosphatase 6 (DUSP6, also named MKP3) can dephosphorylate activated ERK1/2. We showed that DUSP6 was expressed in low baseline levels in normal arteries; however, arterial injury significantly increased DUSP6 levels in the vessel wall. Compared with wild-type mice, Dusp6-deficient mice had smaller neointima. In vitro, IL-1β induced DUSP6 expression and increased VSMC proliferation and migration. Lack of DUSP6 reduced IL-1β-induced VSMC proliferation and migration. DUSP6 deficiency did not affect IL-1β-stimulated ERK1/2 activation. Instead, ERK1/2 inhibitor U0126 prevented DUSP6 induction by IL-1β, indicating that ERK1/2 functions upstream of DUSP6 to regulate DUSP6 expression in VSMCs rather than downstream as a DUSP6 substrate. IL-1β decreased the levels of cell cycle inhibitor p27 and cell-cell adhesion molecule N-cadherin in VSMCs, whereas lack of DUSP6 maintained their high levels, revealing novel functions of DUSP6 in regulating these two molecules. Taken together, our results indicate that lack of DUSP6 attenuated neointima formation following arterial injury by reducing VSMC proliferation and migration, which were likely mediated via maintaining p27 and N-cadherin levels.
Collapse
Affiliation(s)
- Candra D. Hamdin
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
- National Health Research Institutes and Department of Life Sciences, National Central University Joint Ph.D. Program in Biomedicine, Zhongli District, Taoyuan 320317, Taiwan
| | - Meng-Ling Wu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.-L.W.); (Y.-C.H.)
| | - Chen-Mei Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (M.-L.W.); (Y.-C.H.)
| | - Wei-Cheng Jiang
- Department of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Pei-Yu Gung
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Hua-Hui Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan 350401, Taiwan; (H.-C.C.); (T.-H.T.)
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan 350401, Taiwan; (H.-C.C.); (T.-H.T.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 350401, Taiwan; (C.D.H.); (P.-Y.G.); (H.-H.H.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
33
|
Zhang M, Chen Y, Niu F, Luo X, Li J, Hu W. MicroRNA-30a-3p: a potential noncoding RNA target for the treatment of arteriosclerosis obliterans. Aging (Albany NY) 2023; 15:11875-11890. [PMID: 37899171 PMCID: PMC10683622 DOI: 10.18632/aging.205154] [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: 06/16/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023]
Abstract
An increasing number of studies have shown that noncoding RNAs are involved in cardiovascular diseases. Our study shows that the expression of microRNA-30a-3p (miR-30a-3p) in patients with arteriosclerosis obliterans (ASO) of the lower extremities is significantly decreased after endovascular treatment, but its role is unclear. This study aims to explore the role of microRNA-30a-3p in ASO and its related mechanisms. Immunofluorescence and in situ hybridization costaining indicated that microRNA-30a-3p mostly exists in vascular smooth muscle cells (VSMCs). Furthermore, after transfection into VSMCs, microRNA-30a-3p inhibited VSMC proliferation, migration and phenotype switching. In addition, luciferase reporter and western blot analyses indicated that ROCK2 (Rho-related spiral coil 2 containing protein kinase) is a microRNA-30a-3p target gene, and participates in the microRNA-30a-3p mediated cell inhibitory effect. At last, the rat carotid artery was infected by lentivirus after balloon injury, which increased microRNA-30a-3p levels and apparently suppressed the formation of neointima in vivo. Overall, exogenous introduction of microRNA-30a-3p, a noncoding RNA with unlimited potential, may be a new approach to treat ASO.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Vascular Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Chen
- Department of Cardiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Niu
- Department of Vascular Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaohui Luo
- Department of Gynaecology and Obstetrics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangping Li
- Department of Oncological Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Hu
- Department of Vascular Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
34
|
Hwang YJ, Park JH, Cho DH. Far-Infrared Irradiation Decreases Proliferation in Basal and PDGF-Stimulated VSMCs Through AMPK-Mediated Inhibition of mTOR/p70S6K Signaling Axis. J Korean Med Sci 2023; 38:e335. [PMID: 37873631 PMCID: PMC10593596 DOI: 10.3346/jkms.2023.38.e335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/15/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Far-infrared (FIR) irradiation has been reported to improve diverse cardiovascular diseases, including heart failure, hypertension, and atherosclerosis. The dysregulated proliferation of vascular smooth muscle cells (VSMCs) is well established to contribute to developing occlusive vascular diseases such as atherosclerosis and in-stent restenosis. However, the effects of FIR irradiation on VSMC proliferation and the underlying mechanism are unclear. This study investigated the molecular mechanism through which FIR irradiation inhibited VSMC proliferation. METHODS We performed cell proliferation and cell death assay, adenosine 5'-triphosphate (ATP) assay, inhibitor studies, transfection of dominant negative (dn)-AMP-activated protein kinase (AMPK) α1 gene, and western blot analyses. We also conducted confocal microscopic image analyses and ex vivo studies using isolated rat aortas. RESULTS FIR irradiation for 30 minutes decreased VSMC proliferation without altering the cell death. Furthermore, FIR irradiation accompanied decreases in phosphorylation of the mammalian target of rapamycin (mTOR) at Ser2448 (p-mTOR-Ser2448) and p70 S6 kinase (p70S6K) at Thr389 (p-p70S6K-Thr389). The phosphorylation of AMPK at Thr172 (p-AMPK-Thr172) was increased in FIR-irradiated VSMCs, which was accompanied by a decreased cellular ATP level. Similar to in vitro results, FIR irradiation increased p-AMPK-Thr172 and decreased p-mTOR-Ser2448 and p-p70S6K-Thr389 in isolated rat aortas. Pre-treatment with compound C, a specific AMPK inhibitor, or ectopic expression of dn-AMPKα1 gene, significantly reversed FIR irradiation-decreased VSMC proliferation, p-mTOR-Ser2448, and p-p70S6K-Thr389. On the other hand, hyperthermal stimulus (39°C) did not alter VSMC proliferation, cellular ATP level, and AMPK/mTOR/p70S6K phosphorylation. Finally, FIR irradiation attenuated platelet-derived growth factor (PDGF)-stimulated VSMC proliferation by increasing p-AMPK-Thr172, and decreasing p-mTOR-Ser2448 and p-p70S6K-Thr389 in PDGF-induced in vitro atherosclerosis model. CONCLUSION These results show that FIR irradiation decreases the basal and PDGF-stimulated VSMC proliferation, at least in part, by the AMPK-mediated inhibition of mTOR/p70S6K signaling axis irrespective of its hyperthermal effect. These observations suggest that FIR therapy can be used to treat arterial narrowing diseases, including atherosclerosis and in-stent restenosis.
Collapse
Affiliation(s)
- Yun-Jin Hwang
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Korea
| | | | - Du-Hyong Cho
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Korea.
| |
Collapse
|
35
|
van der Linden J, Trap L, Scherer CV, Roks AJM, Danser AHJ, van der Pluijm I, Cheng C. Model Systems to Study the Mechanism of Vascular Aging. Int J Mol Sci 2023; 24:15379. [PMID: 37895059 PMCID: PMC10607365 DOI: 10.3390/ijms242015379] [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: 08/31/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Within cardiovascular aging, arterial aging holds significant importance, as it involves structural and functional alterations in arteries that contribute substantially to the overall decline in cardiovascular health during the aging process. As arteries age, their ability to respond to stress and injury diminishes, while their luminal diameter increases. Moreover, they experience intimal and medial thickening, endothelial dysfunction, loss of vascular smooth muscle cells, cellular senescence, extracellular matrix remodeling, and deposition of collagen and calcium. This aging process also leads to overall arterial stiffening and cellular remodeling. The process of genomic instability plays a vital role in accelerating vascular aging. Progeria syndromes, rare genetic disorders causing premature aging, exemplify the impact of genomic instability. Throughout life, our DNA faces constant challenges from environmental radiation, chemicals, and endogenous metabolic products, leading to DNA damage and genome instability as we age. The accumulation of unrepaired damages over time manifests as an aging phenotype. To study vascular aging, various models are available, ranging from in vivo mouse studies to cell culture options, and there are also microfluidic in vitro model systems known as vessels-on-a-chip. Together, these models offer valuable insights into the aging process of blood vessels.
Collapse
Affiliation(s)
- Janette van der Linden
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline V. Scherer
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anton J. M. Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
36
|
You Q, Shao X, Wang J, Chen X. Progress on Physical Field-Regulated Micro/Nanomotors for Cardiovascular and Cerebrovascular Disease Treatment. SMALL METHODS 2023; 7:e2300426. [PMID: 37391275 DOI: 10.1002/smtd.202300426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/02/2023] [Indexed: 07/02/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) are two major vasculature-related diseases that seriously affect public health worldwide, which can cause serious death and disability. Lack of targeting effect of the traditional CCVD treatment drugs may damage other tissues and organs, thus more specific methods are needed to solve this dilemma. Micro/nanomotors are new materials that can convert external energy into driving force for autonomous movement, which can not only enhance the penetration depth and retention rates, but also increase the contact areas with the lesion sites (such as thrombus and inflammation sites of blood vessels). Physical field-regulated micro/nanomotors using the physical energy sources with deep tissue penetration and controllable performance, such as magnetic field, light, and ultrasound, etc. are considered as the emerging patient-friendly and effective therapeutic tools to overcome the limitations of conventional CCVD treatments. Recent efforts have suggested that physical field-regulated micro/nanomotors on CCVD treatments could simultaneously provide efficient therapeutic effect and intelligent control. In this review, various physical field-driven micro/nanomotors are mainly introduced and their latest advances for CCVDs are highlighted. Last, the remaining challenges and future perspectives regarding the physical field-regulated micro/nanomotors for CCVD treatments are discussed and outlined.
Collapse
Affiliation(s)
- Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Xinyue Shao
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore
| |
Collapse
|
37
|
Nolze A, Matern S, Grossmann C. Calcineurin Is a Universal Regulator of Vessel Function-Focus on Vascular Smooth Muscle Cells. Cells 2023; 12:2269. [PMID: 37759492 PMCID: PMC10528183 DOI: 10.3390/cells12182269] [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: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Calcineurin, a serine/threonine phosphatase regulating transcription factors like NFaT and CREB, is well known for its immune modulatory effects and role in cardiac hypertrophy. Results from experiments with calcineurin knockout animals and calcineurin inhibitors indicate that calcineurin also plays a crucial role in vascular function, especially in vascular smooth muscle cells (VSMCs). In the aorta, calcineurin stimulates the proliferation and migration of VSMCs in response to vascular injury or angiotensin II administration, leading to pathological vessel wall thickening. In the heart, calcineurin mediates coronary artery formation and VSMC differentiation, which are crucial for proper heart development. In pulmonary VSMCs, calcineurin/NFaT signaling regulates the release of Ca2+, resulting in increased vascular tone followed by pulmonary arterial hypertension. In renal VSMCs, calcineurin regulates extracellular matrix secretion promoting fibrosis development. In the mesenteric and cerebral arteries, calcineurin mediates a phenotypic switch of VSMCs leading to altered cell function. Gaining deeper insights into the underlying mechanisms of calcineurin signaling will help researchers to understand developmental and pathogenetical aspects of the vasculature. In this review, we provide an overview of the physiological function and pathophysiology of calcineurin in the vascular system with a focus on vascular smooth muscle cells in different organs. Overall, there are indications that under certain pathological settings reduced calcineurin activity seems to be beneficial for cardiovascular health.
Collapse
Affiliation(s)
| | | | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
38
|
Bi CX, Jin KQ, Yan J, Qin Y, Hong F, Huang WH, Liu YL. Nanofiber-based Stretchable Electrodes for Oriented Culture and Mechanotransduction Monitoring of Smooth Muscle Cells. ACS Sens 2023; 8:3248-3256. [PMID: 37581426 DOI: 10.1021/acssensors.3c01135] [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] [Indexed: 08/16/2023]
Abstract
Vascular smooth muscle cells (SMCs) are circumferentially oriented perpendicular to the blood vessel and maintain the contractile phenotype in physiological conditions. They can sense the mechanical forces of blood vessels expanding and contracting and convert them into biochemical signals to regulate vascular homeostasis. However, the real-time monitoring of mechanically evoked biochemical response while maintaining SMC oriented growth remains an important challenge. Herein, we developed a stretchable electrochemical sensor by electrospinning aligned and elastic polyurethane (PU) nanofibers on the surface of PDMS film and further modification of conductive polymer PEDOT:PSS-LiTFSI-CoPc (PPLC) on the nanofibers (denoted as PPLC/PU/PDMS). The aligned nanofibers on the electrode surface could guide the oriented growth of SMCs and maintain the contractile phenotype, and the modification of PPLC endowed the electrode with good electrochemical sensing performance and stability under mechanical deformation. By culturing cells on the electrode surface, the oriented growth of SMCs and real-time monitoring of stretch-induced H2O2 release were achieved. On this basis, the changes of H2O2 level released by SMCs under the pathology (hypertension) and intervention of natural product resveratrol were quantitatively monitored, which will be helpful to further understand the occurrence and development of vascular-related diseases and the mechanisms of pharmaceutical intervention.
Collapse
Affiliation(s)
- Chen-Xi Bi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Kai-Qi Jin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Yan
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Qin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Feng Hong
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yan-Ling Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
39
|
Ding YN, Wang TT, Lv SJ, Tang X, Wei ZY, Yao F, Xu HS, Chen YN, Wang XM, Wang HY, Wang HP, Zhang ZQ, Zhao X, Hao DL, Sun LH, Zhou Z, Wang L, Chen HZ, Liu DP. SIRT6 is an epigenetic repressor of thoracic aortic aneurysms via inhibiting inflammation and senescence. Signal Transduct Target Ther 2023; 8:255. [PMID: 37394473 DOI: 10.1038/s41392-023-01456-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 07/04/2023] Open
Abstract
Thoracic aortic aneurysms (TAAs) develop asymptomatically and are characterized by dilatation of the aorta. This is considered a life-threating vascular disease due to the risk of aortic rupture and without effective treatments. The current understanding of the pathogenesis of TAA is still limited, especially for sporadic TAAs without known genetic mutation. Sirtuin 6 (SIRT6) expression was significantly decreased in the tunica media of sporadic human TAA tissues. Genetic knockout of Sirt6 in mouse vascular smooth muscle cells accelerated TAA formation and rupture, reduced survival, and increased vascular inflammation and senescence after angiotensin II infusion. Transcriptome analysis identified interleukin (IL)-1β as a pivotal target of SIRT6, and increased IL-1β levels correlated with vascular inflammation and senescence in human and mouse TAA samples. Chromatin immunoprecipitation revealed that SIRT6 bound to the Il1b promoter to repress expression partly by reducing the H3K9 and H3K56 acetylation. Genetic knockout of Il1b or pharmacological inhibition of IL-1β signaling with the receptor antagonist anakinra rescued Sirt6 deficiency mediated aggravation of vascular inflammation, senescence, TAA formation and survival in mice. The findings reveal that SIRT6 protects against TAA by epigenetically inhibiting vascular inflammation and senescence, providing insight into potential epigenetic strategies for TAA treatment.
Collapse
Affiliation(s)
- Yang-Nan Ding
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting-Ting Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang-Jie Lv
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Yu Wei
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han-Shi Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Nan Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Man Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Yu Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He-Ping Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhu-Qin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - De-Long Hao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Hong Sun
- Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Beijing, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
40
|
Heuckeroth RO. Sometimes Gut Smooth Muscle Forget That They Are Supposed to Contract: CARMN and Visceral Myopathy. Gastroenterology 2023; 165:27-29. [PMID: 37172742 PMCID: PMC11302369 DOI: 10.1053/j.gastro.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Affiliation(s)
- Robert O Heuckeroth
- Perelman School of Medicine, University of Pennsylvania and, The Children's Hospital of Philadelphia-Research Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
41
|
Ibrahim AQ, Abdullah MS, Ahram M, Abdalla S. A Modified Protocol for the Isolation, Culture, and Characterization of Human Smooth Muscle Cells from the Umbilical Cord. Methods Protoc 2023; 6:54. [PMID: 37367998 DOI: 10.3390/mps6030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) and vascular endothelial cells are key participants in the pathogenesis of atherosclerosis. Human umbilical vein endothelial cells (HUVECs) and VSMCs are useful models to design therapeutic strategies for many cardiovascular diseases (CVDs). However, procuring a VSMC cell line by researchers, to model atherosclerosis, for example, is impeded by time and cost limitations, as well as by many other logistic problems in many countries. RESULTS This article describes a protocol for the quick and cheap isolation of VSMCs from human umbilical cords using a mechanical and enzymatic method. This VSMC protocol yields a confluent primary culture that could be obtained within 10 days and sub-cultured for 8-10 passages. The isolated cells are characterized by their morphology and the expression of mRNA of marker proteins analyzed by reverse transcription polymerase chain reaction (RT-qPCR). CONCLUSION The protocol described herein for the isolation of VSMCs from human umbilical cords is easy and is time- and cost-efficient. Isolated cells are useful models for understanding the mechanisms underlying many pathophysiological conditions.
Collapse
Affiliation(s)
- Asmaa Q Ibrahim
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Mohammed S Abdullah
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Shtaywy Abdalla
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
42
|
Yin L, Zhou T, Zheng Z, Long X, Qiu H, Lu HS. Editorial: Women in cardiovascular therapeutics. Front Cardiovasc Med 2023; 10:1183427. [PMID: 37234374 PMCID: PMC10207329 DOI: 10.3389/fcvm.2023.1183427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Affiliation(s)
- Liya Yin
- Department of Integrative Medical Sciences, North Ohio Medical University, Rootstown, OH, United States
| | - Ting Zhou
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ze Zheng
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Hongyu Qiu
- Translational Cardiovascular Research Center, Department of Internal Medicine, College of Medicine at Phoenix, University of Arizona, Phoenix, AZ, United States
| | - Hong S. Lu
- Department of Physiology, Saha Cardiovascular Research Center and Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|