1
|
Xing M, Wang F, Chu R, Wang H, Sun Y, Qian M, Jiang H, Midgley AC, Dai G, Zhao Q. Localized COUP-TFII pDNA Delivery Modulates Stem/Progenitor Cell Differentiation to Enhance Endothelialization and Inhibit Calcification of Decellularized Allografts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409744. [PMID: 39656938 PMCID: PMC11792037 DOI: 10.1002/advs.202409744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/12/2024] [Indexed: 12/17/2024]
Abstract
Decellularized allografts have emerged as promising candidates for vascular bypass grafting, owing to their inherent bioactivity and minimal immunogenicity. However, graft failure that results from suboptimal regeneration and pathological remodeling has hindered their clinical adoption. Recent advances in vascular biology highlight the pivotal role of COUP-TFII in orchestrating endothelial identity, angiogenesis, safeguarding against atherosclerosis, and mitigating vascular calcification. Here, plasmid DNA (pDNA) encoding COUP-TFII is incorporated into decellularized allografts to realize localized delivery. Comprehensive in vitro investigation complemented by a bone marrow transplantation model on genetic-lineage-tracing mouse revealed the underlying mechanisms of COUP-TFII in regulating vascular regeneration and remodeling. COUP-TFII augmented endothelialization and inhibited calcification in decellularized allografts by modulating the Ang1/Tie2/PI3K/AKT signaling pathway that dictates the fate of Sca-1+ stem/progenitor cells. Heparin-polyethyleneimine nanoparticles (HEPI) are prepared as COUP-TFII pDNA nanocarriers (COUP-TFII@HPEI) and used to modify decellularized allografts, achieving long-term and stable overexpression of COUP-TFII. Functionalized grafts are evaluated in rat abdominal artery replacement models, demonstrating enhanced neo-artery regeneration without calcification. The study provides an effective strategy to enhance the applicability of decellularized allograft and illustrates their translational prospects for vascular bypass grafting.
Collapse
Affiliation(s)
- Mengmeng Xing
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Fei Wang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Ruowen Chu
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - He Wang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Yuyao Sun
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Huan Jiang
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
| | - Guohao Dai
- Department of BioengineeringNortheastern UniversityBostonMA02115USA
| | - Qiang Zhao
- State key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for Cell ResponsesKey Laboratory of Bioactive Materials (Ministry of Education), College of Life SciencesNankai UniversityTianjin300071China
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and RemodelingHealth Science CenterPeking UniversityBeijing100191China
| |
Collapse
|
2
|
Han Y, Cui Y, Liu J, Wang D, Zou G, Qi X, Meng J, Huang X, He H, Li X. Single-Cell RNA-Seq Reveals Injuries in Aortic Dissection and Identifies PDGF Signalling Pathway as a Potential Therapeutic Target. J Cell Mol Med 2024; 28:e70293. [PMID: 39720896 DOI: 10.1111/jcmm.70293] [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: 04/27/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Aortic dissection (AD) represents a critical condition characterised by a tear in the inner lining of the aorta, leading to the leakage of blood into the layers of the aortic wall, posing a significant risk to life. However, the pathogenesis is unclear. In this study, scRNA-seq was applied to cells derived from aortas of both AD and non-AD donors (control) to unveil the cellular landscape. ScRNA-seq data uncover significant cellular heterogeneity in AD aortas. Specifically, we observed an accumulation of CD4+ T cells, which contributed to inflammation and cell death, and abnormal collagen formation mediated by fibroblast cells in AD. Moreover, we revealed a greater prevalence of cell death, oxidative stress and senescence in AD aorta cells. Furthermore, we found a decrease in the percentage of vascular stem cells (VSCs), along with a repression in their ability to differentiate into contractile vascular smooth muscle cells (VSMCs). Finally, our data demonstrated that the PDGF signalling pathway was activated in AD. We found that PDGF activation could lead to VSMCs aberrant switch from contractile to synthetic phenotype, which could be ameliorated by PDGF inhibitor. This underscores the potential of the PDGF as a therapeutic target for AD. In summary, our study highlights the cellular heterogeneity and associated injuries within aortas affected by AD, including cell death, oxidative stress, senescence and dysregulation of signalling pathways influencing the aberrant phenotypic switch of VSMCs. These insights offer valuable contributions to understanding the molecular mechanisms underlying AD and present new avenues for therapeutic intervention in this condition.
Collapse
Affiliation(s)
- Yichi Han
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Afliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yongji Cui
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Juli Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Dingchen Wang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guoxiang Zou
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Qi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Jinxiu Meng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaoran Huang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Haiwei He
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Ying Z, Lyu L, Xu X, Wen Z, Xue J, Chen M, Li Z, Jiang L, Chen T. Resident vascular Sca1 + progenitors differentiate into endothelial cells in vascular remodeling via miR-145-5p/ERG signaling pathway. iScience 2024; 27:110080. [PMID: 38883819 PMCID: PMC11176791 DOI: 10.1016/j.isci.2024.110080] [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: 11/07/2023] [Revised: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Endothelial cell (EC) damage or dysfunction serves as the initial event in the pathogenesis of various cardiovascular diseases. Progenitor cells have been postulated to be able to differentiate into ECs, facilitate endothelial regeneration, and alleviate vascular pathological remodeling. However, the precise cellular origins and underlying mechanisms remain elusive. Through single-cell RNA sequencing (scRNA-seq), we identified an increasing population of progenitors expressing stem cell antigen 1 (Sca1) during vascular remodeling in mice. Using both mouse femoral artery injury and vein graft models, we determined that Sca1+ cells differentiate into ECs, restored endothelium in arterial and venous remodeling processes. Notably, we have observed that the differentiation of Sca1+ cells into ECs is negatively regulated by the microRNA-145-5p (miR-145-5p)-Erythroblast transformation-specific-related gene (ERG) pathway. Inhibiting miR-145-5p promoted Sca1+ cell differentiation and reduced neointimal formation after vascular injury. Finally, a similar downregulation of miR-145-5p in human arteriovenous fistula was observed comparing to healthy veins.
Collapse
Affiliation(s)
- Zhangquan Ying
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodong Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zuoshi Wen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianing Xue
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Mengjia Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhoubin Li
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo 315010, China
| |
Collapse
|
4
|
Dasgupta S, Reddy KP, Datta P, Barui A. Vitamin D3-incorporated chitosan/collagen/fibrinogen scaffolds promote angiogenesis and endothelial transition via HIF-1/IGF-1/VEGF pathways in dental pulp stem cells. Int J Biol Macromol 2023; 253:127325. [PMID: 37820916 DOI: 10.1016/j.ijbiomac.2023.127325] [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: 05/15/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Effective vascularization during wound healing remains a critical challenge in the regeneration of skin tissue. On the other hand, mesenchymal stem cell (MSC) to endothelial phenotype transition (MEnDoT) is a potential phenomenon grossly underexplored in vascularized skin tissue engineering. Vitamin D3 has a proven role in promoting MEnDoT. Hence, a D3-incorporated scaffold made with biocompatible materials such as chitosan, collagen and fibrinogen should be able to promote endothelial lineage transition in vitro for tissue engineering purposes. In this study, we developed vitamin D3 incorporated chitosan-collagen-fibrinogen (CCF-D3) scaffolds physically crosslinked under UV and conducted thorough physicochemical and biological assays on it compared to a control scaffold without vitamin D3. Our study for the first time reports the potential vascularization property of the CCF-D3 scaffold by inducing the transitions of dental pulp MSC to endothelial lineage via the HIF-1/IGF-1/VEGF pathways. MSC seeded on UV-exposed CCF-D3 scaffolds had higher cell viability and transitioned towards endothelial lineage was observed by elevated proliferative and endothelial-specific gene expressions and flow cytometric analysis of SCA-1+ antibody. The difference in VEGF-A and α-SMA expressions was also observed in the D3-CCF scaffold compared to the scaffolds without D3.
Collapse
Affiliation(s)
- Shalini Dasgupta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India
| | | | - Pallab Datta
- National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India.
| |
Collapse
|
5
|
Gao J, Li L, Zhou D, Sun X, Cui L, Yang D, Wang X, Du P, Yuan W. Effects of norepinephrine‑induced activation of rat vascular adventitial fibroblasts on proliferation and migration of BMSCs involved in vascular remodeling. Exp Ther Med 2023; 25:290. [PMID: 37206559 PMCID: PMC10189611 DOI: 10.3892/etm.2023.11989] [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: 11/07/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Vascular remodeling caused by vascular injury such as hypertension and atherosclerosis is a complex process involving a variety of cells and factors, and the mechanism is unclear. A vascular injury model was simulated by adding norepinephrine (NE) to culture medium of vascular adventitial fibroblasts (AFs). NE induced activation and proliferation of AFs. To investigate the association between the AFs activation and bone marrow mesenchymal stem cells (BMSCs) differentiation in vascular remodeling. BMSCs were cultured with supernatant of the AFs culture medium. BMSC differentiation and migration were observed by immunostaining and Transwell assay, respectively, while cell proliferation was measured using the Cell Counting Kit-8. Expression levels of smooth muscle actin (α-SMA), TGF-β1 and SMAD3 were measured using western blot assay. The results indicated that compared with those in the control group, in which BMSCs were cultured in normal medium, expression levels of α-SMA, TGF-β1 and SMAD3 in BMSCs cultured in medium supplemented with supernatant of AFs, increased significantly (all P<0.05). Activated AFs induced the differentiation of BMSCs into vascular smooth muscle-like cells and promoted proliferation and migration. AFs activated by NE may induce BMSCs to participate in vascular remodeling. These findings may help design and develop new approaches and therapeutic strategies for vascular injury to prevent pathological remodeling.
Collapse
Affiliation(s)
- Jun Gao
- Medical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Li Li
- Pediatric Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Dongli Zhou
- Nurse's Office, Health School of Laiyang, Laiyang, Yantai, Shandong 265200, P.R. China
| | - Xuhong Sun
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Lilu Cui
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Donglin Yang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaohui Wang
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pengchao Du
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| | - Wendan Yuan
- Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Correspondence to: Professor Wendan Yuan or Professor Pengchao Du, Institute of Pathology and Pathophysiology, Basic Medical School, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong 264003, P.R. China E-mail: 981713509 @qq.com
| |
Collapse
|
6
|
Kim TM, Lee RH, Kim MS, Lewis CA, Park C. ETV2/ER71, the key factor leading the paths to vascular regeneration and angiogenic reprogramming. Stem Cell Res Ther 2023; 14:41. [PMID: 36927793 PMCID: PMC10019431 DOI: 10.1186/s13287-023-03267-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Extensive efforts have been made to achieve vascular regeneration accompanying tissue repair for treating vascular dysfunction-associated diseases. Recent advancements in stem cell biology and cell reprogramming have opened unforeseen opportunities to promote angiogenesis in vivo and generate autologous endothelial cells (ECs) for clinical use. We have, for the first time, identified a unique endothelial-specific transcription factor, ETV2/ER71, and revealed its essential role in regulating endothelial cell generation and function, along with vascular regeneration and tissue repair. Furthermore, we and other groups have demonstrated its ability to directly reprogram terminally differentiated non-ECs into functional ECs, proposing ETV2/ER71 as an effective therapeutic target for vascular diseases. In this review, we discuss the up-to-date status of studies on ETV2/ER71, spanning from its molecular mechanism to vasculo-angiogenic role and direct cell reprogramming toward ECs. Furthermore, we discuss future directions to deploy the clinical potential of ETV2/ER71 as a novel and potent target for vascular disorders such as cardiovascular disease, neurovascular impairment and cancer.
Collapse
Affiliation(s)
- Tae Min Kim
- Graduate School of International Agricultural Technology and Institutes of Green-Bio Science and Technology, Seoul National University, 1447 Pyeongchang-daero, Pyeongchang, Gangwon-do, 25354, Republic of Korea.
| | - Ra Ham Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Min Seong Kim
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chloe A Lewis
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Changwon Park
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
7
|
Ma Z, Mao C, Chen X, Yang S, Qiu Z, Yu B, Jia Y, Wu C, Wang Y, Wang Y, Gu R, Yu F, Yin Y, Wang X, Xu Q, Liu C, Liao Y, Zheng J, Fu Y, Kong W. Peptide Vaccine Against ADAMTS-7 Ameliorates Atherosclerosis and Postinjury Neointima Hyperplasia. Circulation 2023; 147:728-742. [PMID: 36562301 DOI: 10.1161/circulationaha.122.061516] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The metalloprotease ADAMTS-7 (a disintegrin and metalloproteinase with thrombospondin type 1 motif 7) is a novel locus associated with human coronary atherosclerosis. ADAMTS-7 deletion protects against atherosclerosis and vascular restenosis in rodents. METHODS We designed 3 potential vaccines consisting of distinct B cell epitopic peptides derived from ADAMTS-7 and conjugated with the carrier protein KLH (keyhole limpet hemocyanin) as well as aluminum hydroxide as an adjuvant. Arterial ligation or wire injury was used to induce neointima in mice, whereas ApoE-/- and LDLR-/- (LDLR [low-density lipoprotein receptor]) mice fed a high-fat diet were applied to assess atherosclerosis. In addition, coronary stent implantation was performed on vaccine-immunized Bama miniature pigs, followed by optical coherence tomography to evaluate coronary intimal hyperplasia. RESULTS A vaccine, ATS7vac, was screened out from 3 candidates to effectively inhibit intimal thickening in murine carotid artery ligation models after vaccination. As well, immunization with ATS7vac alleviated neointima formation in murine wire injury models and mitigated atherosclerotic lesions in both hyperlipidemic ApoE-/- and LDLR-/- mice without lowering lipid levels. Preclinically, ATS7vac markedly impeded intimal hyperplasia in swine stented coronary arteries, but without significant immune-related organ injuries. Mechanistically, ATS7vac vaccination produced specific antibodies against ADAMTS-7, which markedly repressed ADAMTS-7-mediated COMP (cartilage oligomeric matrix protein) and TSP-1 (thrombospondin-1) degradation and subsequently inhibited vascular smooth muscle cell migration but promoted re-endothelialization. CONCLUSIONS ATS7vac is a novel atherosclerosis vaccine that also alleviates in-stent restenosis. The application of ATS7vac would be a complementary therapeutic avenue to the current lipid-lowering strategy for atherosclerotic disease.
Collapse
Affiliation(s)
- Zihan Ma
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China.,Beijing Institute of Biotechnology, Beijing, China (C.M.)
| | - Xiao Chen
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Zhihua Qiu
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Baoqi Yu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (B.Y.), Ministry of Education, Beijing, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (B.Y.)
| | - Yiting Jia
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chao Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yiyi Wang
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Yuhui Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education (Yuhui Wang), Peking University, Beijing, China
| | - Rui Gu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Yanhui Yin
- Department of Immunology (Y.Y.), Peking University, Beijing, China.,Key Laboratory of Medical Immunology of Ministry of Health (Y.Y.), Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (Q.X.).,Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q.X.)
| | - Chuanju Liu
- Department of Orthopedic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY (C.L.)
| | - Yuhua Liao
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yi Fu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| |
Collapse
|
8
|
Shi C, Zhang K, Zhao Z, Wang Y, Xu H, Wei W. Correlation between stem cell molecular phenotype and atherosclerotic plaque neointima formation and analysis of stem cell signal pathways. Front Cell Dev Biol 2023; 11:1080563. [PMID: 36711040 PMCID: PMC9877345 DOI: 10.3389/fcell.2023.1080563] [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: 10/26/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Vascular stem cells exist in the three-layer structure of blood vessel walls and play an indispensable role in angiogenesis under physiological conditions and vascular remodeling under pathological conditions. Vascular stem cells are mostly quiescent, but can be activated in response to injury and participate in endothelial repair and neointima formation. Extensive studies have demonstrated the differentiation potential of stem/progenitor cells to repair endothelium and participate in neointima formation during vascular remodeling. The stem cell population has markers on the surface of the cells that can be used to identify this cell population. The main positive markers include Stem cell antigen-1 (Sca1), Sry-box transcription factor 10 (SOX10). Stromal cell antigen 1 (Stro-1) and Stem cell growth factor receptor kit (c-kit) are still controversial. Different parts of the vessel have different stem cell populations and multiple markers. In this review, we trace the role of vascular stem/progenitor cells in the progression of atherosclerosis and neointima formation, focusing on the expression of stem cell molecular markers that occur during neointima formation and vascular repair, as well as the molecular phenotypic changes that occur during differentiation of different stem cell types. To explore the correlation between stem cell molecular markers and atherosclerotic diseases and neointima formation, summarize the differential changes of molecular phenotype during the differentiation of stem cells into smooth muscle cells and endothelial cells, and further analyze the signaling pathways and molecular mechanisms of stem cells expressing different positive markers participating in intima formation and vascular repair. Summarizing the limitations of stem cells in the prevention and treatment of atherosclerotic diseases and the pressing issues that need to be addressed, we provide a feasible scheme for studying the signaling pathways of vascular stem cells involved in vascular diseases.
Collapse
Affiliation(s)
- Chuanxin Shi
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kefan Zhang
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenyu Zhao
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haozhe Xu
- Department of Biotherapy, Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Wei Wei,
| |
Collapse
|
9
|
Wang F, Qin K, Wang K, Wang H, Liu Q, Qian M, Chen S, Sun Y, Hou J, Wei Y, Hu Y, Li Z, Xu Q, Zhao Q. Nitric oxide improves regeneration and prevents calcification in bio-hybrid vascular grafts via regulation of vascular stem/progenitor cells. Cell Rep 2022; 39:110981. [PMID: 35732119 DOI: 10.1016/j.celrep.2022.110981] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 04/29/2022] [Accepted: 05/28/2022] [Indexed: 11/18/2022] Open
Abstract
Vascular bypass surgery continues to use autologous grafts and often suffers from a shortage of donor grafts. Decellularized xenografts derived from porcine veins provide a promising candidate because of their abundant availability and low immunogenicity. Unfortunately, transplantation outcomes are far from satisfactory because of insufficient regeneration and adverse pathologic remodeling. Herein, a nitrate-functionalized prosthesis has been incorporated into a decellularized porcine vein graft to fabricate a bio-hybrid vascular graft with local delivery of nitric oxide (NO). Exogenous NO efficiently promotes vascular regeneration and attenuates intimal hyperplasia and vascular calcification in both rabbit and mouse models. The underlying mechanism was investigated using a Sca1 2A-CreER; Rosa-RFP genetic-lineage-tracing mouse model that reveals that Sca1+ stem/progenitor cells (SPCs) are major contributors to vascular regeneration and remodeling, and NO plays a critical role in regulating SPC fate. These results support the translational potential of this off-the-shelf vascular graft.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou 256600, China
| | - Kang Qin
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - He Wang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qi Liu
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shang Chen
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yijin Sun
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jingli Hou
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanhua Hu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Sustainable Chemical Transformations, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
10
|
Wang H, Xing M, Deng W, Qian M, Wang F, Wang K, Midgley AC, Zhao Q. Anti-Sca-1 antibody-functionalized vascular grafts improve vascular regeneration via selective capture of endogenous vascular stem/progenitor cells. Bioact Mater 2022; 16:433-450. [PMID: 35415291 PMCID: PMC8965769 DOI: 10.1016/j.bioactmat.2022.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/17/2022] Open
|
11
|
Tao J, Cao X, Yu B, Qu A. Vascular Stem/Progenitor Cells in Vessel Injury and Repair. Front Cardiovasc Med 2022; 9:845070. [PMID: 35224067 PMCID: PMC8866648 DOI: 10.3389/fcvm.2022.845070] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular repair upon vessel injury is essential for the maintenance of arterial homeostasis and function. Stem/progenitor cells were demonstrated to play a crucial role in regeneration and replenishment of damaged vascular cells during vascular repair. Previous studies revealed that myeloid stem/progenitor cells were the main sources of tissue regeneration after vascular injury. However, accumulating evidences from developing lineage tracing studies indicate that various populations of vessel-resident stem/progenitor cells play specific roles in different process of vessel injury and repair. In response to shear stress, inflammation, or other risk factors-induced vascular injury, these vascular stem/progenitor cells can be activated and consequently differentiate into different types of vascular wall cells to participate in vascular repair. In this review, mechanisms that contribute to stem/progenitor cell differentiation and vascular repair are described. Targeting these mechanisms has potential to improve outcome of diseases that are characterized by vascular injury, such as atherosclerosis, hypertension, restenosis, and aortic aneurysm/dissection. Future studies on potential stem cell-based therapy are also highlighted.
Collapse
Affiliation(s)
- Jiaping Tao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Xuejie Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- *Correspondence: Baoqi Yu
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- Aijuan Qu
| |
Collapse
|
12
|
Wei Y, Wang F, Guo Z, Zhao Q. Tissue-engineered vascular grafts and regeneration mechanisms. J Mol Cell Cardiol 2021; 165:40-53. [PMID: 34971664 DOI: 10.1016/j.yjmcc.2021.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are life-threatening diseases with high morbidity and mortality worldwide. Vascular bypass surgery is still the ultimate strategy for CVD treatment. Autografts are the gold standard for graft transplantation, but insufficient sources limit their widespread application. Therefore, alternative tissue engineered vascular grafts (TEVGs) are urgently needed. In this review, we summarize the major strategies for the preparation of vascular grafts, as well as the factors affecting their patency and tissue regeneration. Finally, the underlying mechanisms of vascular regeneration that are mediated by host cells are discussed.
Collapse
Affiliation(s)
- Yongzhen Wei
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China; State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Fei Wang
- State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Zhikun Guo
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China
| | - Qiang Zhao
- Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, Henan Province, China; State key Laboratory of Medicinal Chemical Biology & Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
13
|
Jiang L, Sun X, Deng J, Hu Y, Xu Q. Different Roles of Stem/Progenitor Cells in Vascular Remodeling. Antioxid Redox Signal 2021; 35:192-203. [PMID: 33107320 DOI: 10.1089/ars.2020.8199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Since the discovery of vascular stem cells, there has been considerable advancement in comprehending the nature and functions of these cells. Due to their differentiation potential to repair endothelial cells and to participate in lesion formation during vascular remodeling, it is crucial to elucidate vascular stem cell behaviors and the mechanisms underlying this process, which could provide new chances for the design of clinical therapeutic application of stem cells. Recent Advances: Over the past decades, major progress has been made on progenitor/vascular stem cells in the field of cardiovascular research. Vascular stem cells are mostly latent in their niches and can be bioactivated in response to damage and get involved in endothelial repair and smooth muscle cell aggregation to generate neointima. Accumulating evidence has been shown recently, using genetic lineage tracing mouse models, to particularly provide solutions to the nature of vascular stem cells and to monitor both cell migration and the process of differentiation during physiological angiogenesis and in vascular diseases. Critical Issues: This article reviews and summarizes the current research progress of vascular stem cells in this field and highlights future prospects for stem cell research in regenerative medicine. Future Directions: Despite recent advances and achievements of stem cells in cardiovascular research, the nature and cell fate of vascular stem cells remain elusive. Further comprehensive studies using new techniques including genetic cell lineage tracing and single-cell RNA sequencing are essential to fully illuminate the role of stem cells in vascular development and diseases. Antioxid. Redox Signal. 35, 192-203.
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaolei Sun
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Jolly AJ, Lu S, Strand KA, Dubner AM, Mutryn MF, Nemenoff RA, Majesky MW, Moulton KS, Weiser-Evans MCM. Heterogeneous subpopulations of adventitial progenitor cells regulate vascular homeostasis and pathological vascular remodeling. Cardiovasc Res 2021; 118:1452-1465. [PMID: 33989378 DOI: 10.1093/cvr/cvab174] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are characterized by chronic vascular dysfunction and provoke pathological remodeling events such as neointima formation, atherosclerotic lesion development, and adventitial fibrosis. While lineage-tracing studies have shown that phenotypically modulated smooth muscle cells (SMCs) are the major cellular component of neointimal lesions, the cellular origins and microenvironmental signaling mechanisms that underlie remodeling along the adventitial vascular layer are not fully understood. However, a growing body of evidence supports a unique population of adventitial lineage-restricted progenitor cells expressing the stem cell marker, stem cell antigen-1 (Sca1; AdvSca1 cells) as important effectors of adventitial remodeling and suggests that they are at least partially responsible for subsequent pathological changes that occur in the media and intima. AdvSca1 cells are being studied in murine models of atherosclerosis, perivascular fibrosis, and neointima formation in response to acute vascular injury. Depending on the experimental conditions, AdvSca1 cells exhibit the capacity to differentiate into SMCs, endothelial cells, chondrocytes, adipocytes, and pro-remodeling cells such as myofibroblasts and macrophages. These data indicate that AdvSca1 cells may be a targetable cell population to influence the outcomes of pathologic vascular remodeling. Important questions remain regarding the origins of AdvSca1 cells and the essential signaling mechanisms and microenvironmental factors that regulate both maintenance of their stem-like, progenitor phenotype and their differentiation into lineage-specified cell types. Adding complexity to the story, recent data indicate that the collective population of adventitial progenitor cells is likely composed of several smaller, lineage-restricted subpopulations which are not fully defined by their transcriptomic profile and differentiation capabilities. The aim of this review is to outline the heterogeneity of Sca1+ adventitial progenitor cells, summarize their role in vascular homeostasis and remodeling, and comment on their translational relevance in humans.
Collapse
Affiliation(s)
- Austin J Jolly
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Keith A Strand
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Allison M Dubner
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Marie F Mutryn
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Raphael A Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension.,School of Medicine,Consortium for Fibrosis Research and Translation
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101.,Departments of Pediatrics and Pathology, University of Washington, Seattle, WA, 98195
| | | | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension.,School of Medicine,Consortium for Fibrosis Research and Translation.,Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| |
Collapse
|
15
|
Adventitial Progenitor Cells of Human Great Saphenous Vein Enhance the Resolution of Venous Thrombosis via Neovascularization. Stem Cells Int 2021; 2021:8816763. [PMID: 33679991 PMCID: PMC7926266 DOI: 10.1155/2021/8816763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/20/2021] [Accepted: 02/06/2021] [Indexed: 11/24/2022] Open
Abstract
Background Vascular adventitia contains progenitor cells and is shown to participate in vascular remolding. Progenitor cells are recruited into the venous thrombi in mice to promote neovascularization. We hypothesized that the adventitial progenitor cells of human great saphenous vein (HGSV-AdPC) enhance the resolution of venous thrombosis via neovascularization. Methods Human great saphenous vein (HGSV) was harvested from the patients with great saphenous vein varicose and sectioned for immunohistochemistry, or minced for progenitor cell primary culture, or placed in sodium dodecyl sulfate solution for decellularization. Human venous thrombi were collected from patients with great saphenous vein varicose and superficial thrombophlebitis. Infrarenal abdominal aorta of New Zealand white rabbits was replaced with interposing decellularized vessel, and the patency of the grafts was confirmed by ultrasonic examination. Animal venous thrombi in the left infrarenal vena cava of mice were produced with Prolene suture ligation and ophthalmic force clipping of this portion. After HGSVs were digested by collagenase, the CD34+CD117+ HGSV-AdPC were isolated on FACS system, labelled with CM-Dil, and transplanted into the adventitia of infrarenal vena cava of nude mice. The percentage of thrombus organization area to the thrombus area was calculated as the organization rate. The thrombus cell, endothelial cells, and macrophages in the thrombi were counted in sections. Cell smears and frozen sections of human saphenous veins and venous thrombi were labeled with Sca1, CD34, CD117, Flk1, CD31, and F4/80 antibodies. The CD34+CD117+ HGSV-AdPC were cultured in endothelial growth medium with vascular endothelial growth factor (VEGF) to induce endothelial cell differentiation and analyzed with real time-PCR, Western blotting, and tube formation assays. Results Immunohistochemical staining showed that the CD34+CD117+ cells were located within the adventitia of HGSVs, and many CD34+ and CD117+ cells have emerged in the human venous thrombi. The number of progenitor cells within the marginal area of 7 days mice thrombi was shown to be Sca1+ ≈21%, CD34+ ≈12%, CD117+ ≈9%, and Flk1+ ≈5%. Many CD34+adventitial progenitor cells have migrated into the decellularized vessels. FACS showed that the number of CD34+CD117+ HGSV-AdPC in primary cultured cells as 1.2 ± 0.07%. After CD34+CD117+HGSV-AdPC were transplanted into the adventitia of nude mice vena cava with venous thrombi, the organization rate, nucleate cell count, endothelial cells, and macrophage cells of thrombi were shown to be significantly increased. The transplanted CD34+CD117+ HGSV-AdPC at the adventitia have crossed the vein wall, entered the venous thrombi, and differentiated into endothelial cells. The CD34+CD117+ HGSV-AdPC in the culture medium in the presence of VEGF-promoted gene and protein expression of endothelial cell markers in vitro and induced tube formation. Conclusions HGSV-AdPC could cross the vein wall and migrate from the adventitia into the venous thrombi. Increased HGSV-AdPC in the adventitia has enhanced the resolution of venous thrombi via differentiating into endothelial cells of neovascularization.
Collapse
|
16
|
Zhou Y, Huang C, Hu Y, Xu Q, Hu X. Lymphatics in Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2020; 40:e275-e283. [PMID: 33085520 DOI: 10.1161/atvbaha.120.314735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Yijiang Zhou
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Chengchen Huang
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanhua Hu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Qingbo Xu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaosheng Hu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Nasser MI, Qi X, Zhu S, He Y, Zhao M, Guo H, Zhu P. Current situation and future of stem cells in cardiovascular medicine. Biomed Pharmacother 2020; 132:110813. [PMID: 33068940 DOI: 10.1016/j.biopha.2020.110813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Currently, many methods have been proposed by researchers for the prevention and treatment of CVD; among them, stem cell-based therapies are the most promising. As the cells of origin for various mature cells, stem cells have the ability to self-renew and differentiate. Stem cells have a powerful ability to regenerate biologically, self-repair, and enhance damaged functional tissues or organs. Allogeneic stem cells and somatic stem cells are two types of cells that can be used for cardiac repair. Theoretically, dilated cardiomyopathy and acute myocardial infarction can be treated with such cells. In addition, stem cell transplantation procedures, including intravenous, epicardial, cardiac, and endocardial injections, have been reported to provide significant benefits in clinical practice; however, there are still a number of issues that need further study and consideration, such as the form and quantity of transplanted cells and post-transplantation health. The goal of this analysis was to summarize the recent advances in stem cell-based therapies and their efficacy in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Xiao Qi
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Yin He
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Mingyi Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Huiming Guo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China.
| |
Collapse
|
18
|
Yan W, Li T, Yin T, Hou Z, Qu K, Wang N, Durkan C, Dong L, Qiu J, Gregersen H, Wang G. M2 macrophage-derived exosomes promote the c-KIT phenotype of vascular smooth muscle cells during vascular tissue repair after intravascular stent implantation. Theranostics 2020; 10:10712-10728. [PMID: 32929376 PMCID: PMC7482821 DOI: 10.7150/thno.46143] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: For intravascular stent implantation to be successful, the processes of vascular tissue repair and therapy are considered to be critical. However, the mechanisms underlying the eventual fate of vascular smooth muscle cells (VSMCs) during vascular tissue repair remains elusive. In this study, we hypothesized that M2 macrophage-derived exosomes to mediate cell-to-cell crosstalk and induce dedifferentiation phenotypes in VSMCs. Methods: In vivo, 316L bare metal stents (BMS) were implanted from the left iliac artery into the abdominal aorta of 12-week-old male Sprague-Dawley (SD) rats for 7 and 28 days. Hematoxylin and eosin (HE) were used to stain the neointimal lesions. En-face immunofluorescence staining of smooth muscle 22 alpha (SM22α) and CD68 showed the rat aorta smooth muscle cells (RASMCs) and macrophages. Immunohistochemical staining of total galactose-specific lectin 3 (MAC-2) and total chitinase 3-like 3 (YM-1) showed the total macrophages and M2 macrophages. In vitro, exosomes derived from IL-4+IL-13-treated macrophages (M2Es) were isolated by ultracentrifugation and characterized based on their specific morphology. Ki-67 staining was conducted to assess the effects of the M2Es on the proliferation of RASMCs. An atomic force microscope (AFM) was used to detect the stiffness of the VSMCs. GW4869 was used to inhibit exosome release. RNA-seq was performed to determine the mRNA profiles of the RASMCs and M2Es-treated RASMCs. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of the mRNAs. Western blotting was used to detect the candidate protein expression levels. T-5224 was used to inhibit the DNA binding activity of AP-1 in RASMCs. Results: M2Es promote c-KIT expression and softening of nearby VSMCs, hence accelerating the vascular tissue repair process. VSMCs co-cultured in vitro with M2 macrophages presented an increased capacity for de-differentiation and softening, which was exosome dependent. In addition, the isolated M2Es helped to promote VSMC dedifferentiation and softening. Furthermore, the M2Es enhanced vascular tissue repair potency by upregulation of VSMCs c-KIT expression via activation of the c-Jun/activator protein 1 (AP-1) signaling pathway. Conclusions: The findings of this study emphasize the prominent role of M2Es during VSMC dedifferentiation and vascular tissue repair via activation of the c-Jun/AP-1 signaling pathway, which has a profound impact on the therapeutic strategies of coronary stenting techniques.
Collapse
|
19
|
Yang F, Chen Q, Yang M, Maguire EM, Yu X, He S, Xiao R, Wang CS, An W, Wu W, Zhou Y, Xiao Q, Zhang L. Macrophage-derived MMP-8 determines smooth muscle cell differentiation from adventitia stem/progenitor cells and promotes neointima hyperplasia. Cardiovasc Res 2020; 116:211-225. [PMID: 30778537 DOI: 10.1093/cvr/cvz044] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/17/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
AIMS Emerging evidence has suggested that adventitia stem/progenitor cells (AdSPCs) migrate into the intima of arteries in response to injury, where they differentiate towards smooth muscle cells (SMCs) and participate in neointimal hyperplasia. We have previously identified matrix metalloproteinase-8 (MMP8) as a key player in atherogenesis. In this study, we aimed to investigate the functional roles of macrophage-derived MMP8 in AdSPC differentiation and injury-induced arterial remodelling. METHODS AND RESULTS We first observed an important role for MMP8 in SMC differentiation from embryonic stem cells, but this effect was not seen in AdSPCs. Instead, through macrophages/AdSPCs co-culture and macrophage conditional culture medium studies, we have demonstrated that the MMP8 protein secreted from macrophages promotes SMC differentiation from AdSPCs. Mechanistically, we showed that macrophage-derived MMP8 promotes SMC differentiation from AdSPCs through modulating transforming growth factor-β activity and a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10)/Notch1 signalling. We further demonstrated that the binding site for CBF1, Suppressor of Hairless, and Lag-1 (CSL) within SMC gene promoters is responsible for Notch1 mediated SMC differentiation. Finally, we demonstrated that macrophage-derived MMP8 increased injury-induced neointimal SMC hyperplasia by activating ADAM10/Notch1 signalling. CONCLUSIONS We have identified macrophage-derived MMP8 as a regulator in SMC differentiation from AdSPCs and neointimal SMC hyperplasia in response to injury. Our data provide new insights into the roles of MMP8 in AdSPC differentiation and the pathogenesis of neointima formation in the context of angiographic restenosis, and therefore may aid in the development of novel therapeutic agents for the prevention of this disease.
Collapse
Affiliation(s)
- Feng Yang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Mei Yang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Eithne Margaret Maguire
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Xiaotian Yu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Shiping He
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Rui Xiao
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Claire S Wang
- Gonville & Caius College, University of Cambridge, Trinity Street, Cambridge, CB2 1TA, UK
| | - Weiwei An
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Wei Wu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Yijiang Zhou
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| | - Qingzhong Xiao
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.,Key Laboratory of Cardiovascular Diseases, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, Guangdong, 511436, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, Guangdong, 511436, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| |
Collapse
|
20
|
Wang K, Lin RZ, Hong X, Ng AH, Lee CN, Neumeyer J, Wang G, Wang X, Ma M, Pu WT, Church GM, Melero-Martin JM. Robust differentiation of human pluripotent stem cells into endothelial cells via temporal modulation of ETV2 with modified mRNA. SCIENCE ADVANCES 2020; 6:eaba7606. [PMID: 32832668 DOI: 10.1101/2020.03.02.973289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/09/2020] [Indexed: 05/23/2023]
Abstract
Human induced pluripotent stem cell (h-iPSC)-derived endothelial cells (h-iECs) have become a valuable tool in regenerative medicine. However, current differentiation protocols remain inefficient and lack reliability. Here, we describe a method for rapid, consistent, and highly efficient generation of h-iECs. The protocol entails the delivery of modified mRNA encoding the transcription factor ETV2 at the intermediate mesodermal stage of differentiation. This approach reproducibly differentiated 13 diverse h-iPSC lines into h-iECs with exceedingly high efficiency. In contrast, standard differentiation methods that relied on endogenous ETV2 were inefficient and notably inconsistent. Our h-iECs were functionally competent in many respects, including the ability to form perfused vascular networks in vivo. Timely activation of ETV2 was critical, and bypassing the mesodermal stage produced putative h-iECs with reduced expansion potential and inability to form functional vessels. Our protocol has broad applications and could reliably provide an unlimited number of h-iECs for vascular therapies.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Xuechong Hong
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Alex H Ng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Chin Nien Lee
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Neumeyer
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
| | - Gang Wang
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
21
|
Wang K, Lin RZ, Hong X, Ng AH, Lee CN, Neumeyer J, Wang G, Wang X, Ma M, Pu WT, Church GM, Melero-Martin JM. Robust differentiation of human pluripotent stem cells into endothelial cells via temporal modulation of ETV2 with modified mRNA. SCIENCE ADVANCES 2020; 6:eaba7606. [PMID: 32832668 PMCID: PMC7439318 DOI: 10.1126/sciadv.aba7606] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/09/2020] [Indexed: 05/04/2023]
Abstract
Human induced pluripotent stem cell (h-iPSC)-derived endothelial cells (h-iECs) have become a valuable tool in regenerative medicine. However, current differentiation protocols remain inefficient and lack reliability. Here, we describe a method for rapid, consistent, and highly efficient generation of h-iECs. The protocol entails the delivery of modified mRNA encoding the transcription factor ETV2 at the intermediate mesodermal stage of differentiation. This approach reproducibly differentiated 13 diverse h-iPSC lines into h-iECs with exceedingly high efficiency. In contrast, standard differentiation methods that relied on endogenous ETV2 were inefficient and notably inconsistent. Our h-iECs were functionally competent in many respects, including the ability to form perfused vascular networks in vivo. Timely activation of ETV2 was critical, and bypassing the mesodermal stage produced putative h-iECs with reduced expansion potential and inability to form functional vessels. Our protocol has broad applications and could reliably provide an unlimited number of h-iECs for vascular therapies.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Xuechong Hong
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Alex H. Ng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Chin Nien Lee
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Neumeyer
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Gang Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - George M. Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
22
|
Yang J, Moraga A, Xu J, Zhao Y, Luo P, Lao KH, Margariti A, Zhao Q, Ding W, Wang G, Zhang M, Zheng L, Zhang Z, Hu Y, Wang W, Shen L, Smith A, Shah AM, Wang Q, Zeng L. A histone deacetylase 7-derived peptide promotes vascular regeneration via facilitating 14-3-3γ phosphorylation. Stem Cells 2020; 38:556-573. [PMID: 31721359 PMCID: PMC7187271 DOI: 10.1002/stem.3122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Histone deacetylase 7 (HDAC7) plays a pivotal role in the maintenance of the endothelium integrity. In this study, we demonstrated that the intron-containing Hdac7 mRNA existed in the cytosol and that ribosomes bound to a short open reading frame (sORF) within the 5'-terminal noncoding area of this Hdac7 mRNA in response to vascular endothelial growth factor (VEGF) stimulation in the isolated stem cell antigen-1 positive (Sca1+ ) vascular progenitor cells (VPCs). A 7-amino acid (7A) peptide has been demonstrated to be translated from the sORF in Sca1+ -VPCs in vitro and in vivo. The 7A peptide was shown to receive phosphate group from the activated mitogen-activated protein kinase MEKK1 and transfer it to 14-3-3 gamma protein, forming an MEKK1-7A-14-3-3γ signal pathway downstream VEGF. The exogenous synthetic 7A peptide could increase Sca1+ -VPCs cell migration, re-endothelialization in the femoral artery injury, and angiogenesis in hind limb ischemia. A Hd7-7sFLAG transgenic mice line was generated as the loss-of-function model, in which the 7A peptide was replaced by a FLAG-tagged scrabbled peptide. Loss of the endogenous 7A impaired Sca1+ -VPCs cell migration, re-endothelialization of the injured femoral artery, and angiogenesis in ischemic tissues, which could be partially rescued by the addition of the exogenous 7A/7Ap peptide. This study provides evidence that sORFs can be alternatively translated and the derived peptides may play an important role in physiological processes including vascular remodeling.
Collapse
Affiliation(s)
- Junyao Yang
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK.,Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ana Moraga
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Jing Xu
- Institute of Bioengineering, Queen Mary University of London, London, UK
| | - Yue Zhao
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Peiyi Luo
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Ka Hou Lao
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Andriana Margariti
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Wei Ding
- Institute of Bioengineering, Queen Mary University of London, London, UK
| | - Gang Wang
- Department of Emergency Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Min Zhang
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Lei Zheng
- Southern Medical University, Guangzhou, People's Republic of China
| | - Zhongyi Zhang
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Yanhua Hu
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Wen Wang
- Institute of Bioengineering, Queen Mary University of London, London, UK
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Alberto Smith
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Qian Wang
- Southern Medical University, Guangzhou, People's Republic of China
| | - Lingfang Zeng
- School of Cardiovascular Medicine and Sciences, King's College - London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, UK
| |
Collapse
|
23
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
24
|
Lee DH, Kim TM, Kim JK, Park C. ETV2/ER71 Transcription Factor as a Therapeutic Vehicle for Cardiovascular Disease. Theranostics 2019; 9:5694-5705. [PMID: 31534512 PMCID: PMC6735401 DOI: 10.7150/thno.35300] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases have long been the leading cause of mortality and morbidity in the United States as well as worldwide. Despite numerous efforts over the past few decades, the number of the patients with cardiovascular disease still remains high, thereby necessitating the development of novel therapeutic strategies equipped with a better understanding of the biology of the cardiovascular system. Recently, the ETS transcription factor, ETV2 (also known as ER71), has been recognized as a master regulator of the development of the cardiovascular system and plays an important role in pathophysiological angiogenesis and the endothelial cell reprogramming. Here, we discuss the detailed mechanisms underlying ETV2/ER71-regulated cardiovascular lineage development. In addition, recent reports on the novel functions of ETV2/ER71 in neovascularization and direct cell reprogramming are discussed with a focus on its therapeutic potential for cardiovascular diseases.
Collapse
|
25
|
Gu W, Nowak WN, Xie Y, Le Bras A, Hu Y, Deng J, Issa Bhaloo S, Lu Y, Yuan H, Fidanis E, Saxena A, Kanno T, Mason AJ, Dulak J, Cai J, Xu Q. Single-Cell RNA-Sequencing and Metabolomics Analyses Reveal the Contribution of Perivascular Adipose Tissue Stem Cells to Vascular Remodeling. Arterioscler Thromb Vasc Biol 2019; 39:2049-2066. [PMID: 31340667 PMCID: PMC6766361 DOI: 10.1161/atvbaha.119.312732] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Perivascular adipose tissue (PVAT) plays a vital role in maintaining vascular homeostasis. However, most studies ascribed the function of PVAT in vascular remodeling to adipokines secreted by the perivascular adipocytes. Whether mesenchymal stem cells exist in PVAT and play a role in vascular regeneration remain unknown. Approach and Results: Single-cell RNA-sequencing allowed direct visualization of the heterogeneous PVAT-derived mesenchymal stem cells (PV-ADSCs) at a high resolution and revealed 2 distinct subpopulations, among which one featured signaling pathways crucial for smooth muscle differentiation. Pseudotime analysis of cultured PV-ADSCs unraveled their smooth muscle differentiation trajectory. Transplantation of cultured PV-ADSCs in mouse vein graft model suggested the contribution of PV-ADSCs to vascular remodeling through smooth muscle differentiation. Mechanistically, treatment with TGF-β1 (transforming growth factor β1) and transfection of microRNA (miR)-378a-3p mimics induced a similar metabolic reprogramming of PV-ADSCs, including upregulated mitochondrial potential and altered lipid levels, such as increased cholesterol and promoted smooth muscle differentiation. CONCLUSIONS Single-cell RNA-sequencing allows direct visualization of PV-ADSC heterogeneity at a single-cell level and uncovers 2 subpopulations with distinct signature genes and signaling pathways. The function of PVAT in vascular regeneration is partly attributed to PV-ADSCs and their differentiation towards smooth muscle lineage. Mechanistic study presents miR-378a-3p which is a potent regulator of metabolic reprogramming as a potential therapeutic target for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Witold N Nowak
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Yao Xie
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Alexandra Le Bras
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Yanhua Hu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Jiacheng Deng
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Shirin Issa Bhaloo
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| | - Yao Lu
- Center of Clinical Pharmacology, Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha, China (Y.L., H.Y., J.C.)
| | - Hong Yuan
- Center of Clinical Pharmacology, Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha, China (Y.L., H.Y., J.C.)
| | - Efthymios Fidanis
- Genomics Research Platform, Biomedical Research Centre at Guy's Hospital, London, United Kingdom (E.F., A.S.)
| | - Alka Saxena
- Genomics Research Platform, Biomedical Research Centre at Guy's Hospital, London, United Kingdom (E.F., A.S.)
| | - Tokuwa Kanno
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London, United Kingdom (T.K., A.J.M.)
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London, United Kingdom (T.K., A.J.M.)
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland (J. Dulak)
| | - Jingjing Cai
- Center of Clinical Pharmacology, Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha, China (Y.L., H.Y., J.C.)
| | - Qingbo Xu
- From the School of Cardiovascular Medicine and Sciences, King's College London, BHF Centre, United Kingdom (W.G., W.N.N., Y.X., A.L.B., Y.H., J. Deng, S.I.B., Q.X.)
| |
Collapse
|
26
|
Lu Y, Thavarajah T, Gu W, Cai J, Xu Q. Impact of miRNA in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e159-e170. [PMID: 30354259 DOI: 10.1161/atvbaha.118.310227] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yao Lu
- From the Center of Clinical Pharmacology (Y.L.)
| | - Tanuja Thavarajah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Jingjing Cai
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingbo Xu
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| |
Collapse
|
27
|
Hong X, Gu W. Plasticity of vascular resident mesenchymal stromal cells during vascular remodeling. VASCULAR BIOLOGY 2019; 1:H67-H73. [PMID: 32923956 PMCID: PMC7439836 DOI: 10.1530/vb-19-0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/11/2019] [Indexed: 11/26/2022]
Abstract
Vascular remodeling is a complex and dynamic pathological process engaging many different cell types that reside within the vasculature. Mesenchymal stromal/stem cells (MSCs) refer to a heterogeneous cell population with the plasticity to differentiate toward multiple mesodermal lineages. Various types of MSC have been identified within the vascular wall that actively contribute to the vascular remodeling process such as atherosclerosis. With the advances of genetic mouse models, recent findings demonstrated the crucial roles of MSCs in the progression of vascular diseases. This review aims to provide an overview on the current knowledge of the characteristics and behavior of vascular resident MSCs under quiescence and remodeling conditions, which may lead to the development of novel therapeutic approaches for cardiovascular diseases.
Collapse
Affiliation(s)
- Xuechong Hong
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenduo Gu
- Cardiovascular Division, BHF Centre for Vascular Regeneration, King's College London, London, UK
| |
Collapse
|
28
|
Vasculogenic properties of adventitial Sca-1 +CD45 + progenitor cells in mice: a potential source of vasa vasorum in atherosclerosis. Sci Rep 2019; 9:7286. [PMID: 31086203 PMCID: PMC6513996 DOI: 10.1038/s41598-019-43765-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/30/2019] [Indexed: 02/02/2023] Open
Abstract
The cellular origins of vasa vasorum are ill-defined and may involve circulating or local progenitor cells. We previously discovered that murine aortic adventitia contains Sca-1+CD45+ progenitors that produce macrophages. Here we investigated whether they are also vasculogenic. In aortas of C57BL/6 mice, Sca-1+CD45+ cells were localised to adventitia and lacked surface expression of endothelial markers (<1% for CD31, CD144, TIE-2). In contrast, they did show expression of CD31, CD144, TIE-2 and VEGFR2 in atherosclerotic ApoE-/- aortas. Although Sca-1+CD45+ cells from C57BL/6 aorta did not express CD31, they formed CD31+ colonies in endothelial differentiation media and produced interconnecting vascular-like cords in Matrigel that contained both endothelial cells and a small population of macrophages, which were located at branch points. Transfer of aortic Sca-1+CD45+ cells generated endothelial cells and neovessels de novo in a hindlimb model of ischaemia and resulted in a 50% increase in perfusion compared to cell-free control. Similarly, their injection into the carotid adventitia of ApoE-/- mice produced donor-derived adventitial and peri-adventitial microvessels after atherogenic diet, suggestive of newly formed vasa vasorum. These findings show that beyond its content of macrophage progenitors, adventitial Sca-1+CD45+ cells are also vasculogenic and may be a source of vasa vasorum during atherogenesis.
Collapse
|
29
|
Cheng J, Wen J, Wang N, Wang C, Xu Q, Yang Y. Ion Channels and Vascular Diseases. Arterioscler Thromb Vasc Biol 2019; 39:e146-e156. [DOI: 10.1161/atvbaha.119.312004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jun Cheng
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Jing Wen
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Na Wang
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Claire Wang
- Gonville and Caius College, University of Cambridge, United Kingdom (C.W.)
| | - Qingbo Xu
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yan Yang
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| |
Collapse
|
30
|
He H, Yu B, Liu Z, Ye G, You W, Hong Y, Lian Q, Zhang Y, Li X. Vascular progenitor cell senescence in patients with Marfan syndrome. J Cell Mol Med 2019; 23:4139-4152. [PMID: 30920150 PMCID: PMC6533473 DOI: 10.1111/jcmm.14301] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/29/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Vascular progenitor cells (VPCs) present in the adventitia of the vessel wall play a critical role in the regulation of vascular repair following injury. This study aimed to assess the function of VPCs isolated from patients with Marfan syndrome (MFS). VPCs were isolated from control and MFS donors and characterized. Compared with control‐VPCs, MFS‐VPCs exhibited cellular senescence as demonstrated by increased cell size, higher SA‐β‐gal activity and elevated levels of p53 and p21. RNA sequencing showed that several cellular process‐related pathways including cell cycle and cellular senescence were significantly enriched in MFP‐VPCs. Notably, the expression level of TGF‐β1 was much higher in MFS‐VPCs than control‐VPCs. Treatment of control‐VPCs with TGF‐β1 significantly enhanced mitochondrial reactive oxidative species (ROS) and induced cellular senescence whereas inhibition of ROS reversed these effects. MFS‐VPCs displayed increased mitochondrial fusion and decreased mitochondrial fission. Treatment of control‐VPCs with TGF‐β1 increased mitochondrial fusion and reduced mitochondrial fission. Nonetheless, treatment of mitofusin2 (Mfn2)‐siRNA inhibited TGF‐β1‐induced mitochondrial fusion and cellular senescence. Furthermore, TGF‐β1‐induced mitochondrial fusion was mediated by the AMPK signalling pathway. Our study shows that TGF‐β1 induces VPC senescence in patients with MFS by mediating mitochondrial dynamics via the AMPK signalling pathway.
Collapse
Affiliation(s)
- Haiwei He
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baoqi Yu
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodelling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zipeng Liu
- Center for Genomic Sciences, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Gen Ye
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wei You
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yimei Hong
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qizhou Lian
- Department of Medicine, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Li
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
31
|
Tao X, Sun M, Chen M, Ying R, Su W, Zhang J, Xie X, Wei W, Meng X. HMGB1-modified mesenchymal stem cells attenuate radiation-induced vascular injury possibly via their high motility and facilitation of endothelial differentiation. Stem Cell Res Ther 2019; 10:92. [PMID: 30867070 PMCID: PMC6416980 DOI: 10.1186/s13287-019-1197-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/28/2022] Open
Abstract
Background Vascular injury is one of the most common detrimental effects of cancer radiotherapy on healthy tissues. Since the efficacy of current preventive and therapeutic strategies remains limited, the exploration of new approaches to treat radiation-induced vascular injury (RIV) is on high demands. The use of mesenchymal stem cells (MSCs) to treat RIV holds great promise thanks to their well-documented function of mediating tissue regeneration after injury. Recently, we genetically modified MSCs with high mobility group box 1 (HMGB1) and demonstrated the high efficacy of these cells in treating graft atherosclerosis. The current study was to investigate the protective effect of HMGB1-modified MSCs (MSC-H) on RIV by using a rat model. Methods Female F344 rats received an intravenous injection of male F344 MSC-H cells or vehicle control at four doses of 2 × 106 cells with a 15-day interval starting from 30 days after irradiation to the abdominal aorta. The aortas were procured for histological and biomedical analysis at 90 days after irradiation. Cell migration to irradiated aortas was traced by green fluorescent protein and sex determination region on the Y chromosome. In vitro cell migration and endothelial differentiation of MSC-H cells were analyzed by stromal-derived factor 1-induced transwell assay and RNA microarray, respectively. The contribution of extracellular HMGB1 to the bioactivity of MSC-H cells was investigated by inhibition experiments with HMGB1 antibody. Result MSC-H cell infusion alleviated neointimal formation, vascular inflammation, and fibrosis in irradiated aortas, which was associated with local migration and endothelial differentiation of MSC-H cells. The MSC-H cells showed high motility and potential of endothelial differentiation in vitro. Microarray analysis suggested multiple pathways like MAPK and p53 signaling were activated during endothelial differentiation. MSC-H cells highly expressed CXC chemokine receptor 4 and migrated progressively after stromal-derived factor 1 stimulation, which was blocked by the antagonist of CXC chemokine receptor 4. Finally, the migration and endothelial differentiation of MSC-H cells were inhibited by HMGB1 antibody. Conclusion MSC-H cell infusion significantly attenuated RIV, which was associated with their high motility and endothelial differentiation potential. Multiple pathways that possibly contributed to the efficacy of MSC-H cells were suggested and deserved further investigation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1197-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuan Tao
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingyang Sun
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Chen
- Department of Gastroenterology, Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Rongchao Ying
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Su
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Xie
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Department of Gastroenterological Surgery, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China.
| | - Xiaohu Meng
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
32
|
Majesky MW. Vascular Development. Arterioscler Thromb Vasc Biol 2019; 38:e17-e24. [PMID: 29467221 DOI: 10.1161/atvbaha.118.310223] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The vascular system forms as a branching network of endothelial cells that acquire identity as arterial, venous, hemogenic, or lymphatic. Endothelial specification depends on gene targets transcribed by Ets domain-containing factors, including Ets variant gene 2 (Etv2), together with the activity of chromatin-remodeling complexes containing Brahma-related gene-1 (Brg1). Once specified and assembled into vessels, mechanisms regulating lumen diameter and axial growth ensure that the structure of the branching vascular network matches the need for perfusion of target tissues. In addition, blood vessels provide important morphogenic cues that guide or direct the development of organs forming around them. As the embryo grows and lumen diameters increase, smooth muscle cells wrap around the nascent vessel walls to provide mechanical strength and vasomotor control of the circulation. Increasing mechanical stretch and wall strain promote smooth muscle cell differentiation via coupling of actin cytoskeletal remodeling to myocardin and serum response factor-dependent transcription. Remodeling of artery walls by developmental signaling pathways reappears in postnatal blood vessels during physiological and pathological adaptation to vessel wall injury, inflammation, or chronic hypoxia. Recent reports providing insights into major steps in vascular development are reviewed here with a particular emphasis on studies that have been recently published in Arteriosclerosis, Thrombosis, and Vascular Biology.
Collapse
Affiliation(s)
- Mark W Majesky
- From the Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA; and Departments of Pediatrics and Pathology, University of Washington, Seattle.
| |
Collapse
|
33
|
Abstract
Vascular, resident stem cells are present in all 3 layers of the vessel wall; they play a role in vascular formation under physiological conditions and in remodeling in pathological situations. Throughout development and adult early life, resident stem cells participate in vessel formation through vasculogenesis and angiogenesis. In adults, the vascular stem cells are mostly quiescent in their niches but can be activated in response to injury and participate in endothelial repair and smooth muscle cell accumulation to form neointima. However, delineation of the characteristics and of the migration and differentiation behaviors of these stem cells is an area of ongoing investigation. A set of genetic mouse models for cell lineage tracing has been developed to specifically address the nature of these cells and both migration and differentiation processes during physiological angiogenesis and in vascular diseases. This review summarizes the current knowledge on resident stem cells, which has become more defined and refined in vascular biology research, thus contributing to the development of new potential therapeutic strategies to promote endothelial regeneration and ameliorate vascular disease development.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| | - Ting Chen
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences (B.Z.)
| | - Qingbo Xu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| |
Collapse
|