1
|
Chen Q, Zheng A, Xu X, Shi Z, Yang M, Sun S, Wang L, Wang Y, Zhao H, Xiao Q, Zhang L. Nrf3-Mediated Mitochondrial Superoxide Promotes Cardiomyocyte Apoptosis and Impairs Cardiac Functions by Suppressing Pitx2. Circulation 2025; 151:1024-1046. [PMID: 40099370 DOI: 10.1161/circulationaha.124.070286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/31/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Myocardial infarction (MI) elicits mitochondria reactive oxygen species (ROS) production and cardiomyocyte (CM) apoptosis. Nrf3 (nuclear factor erythroid 2-related factor 3) has an established role in regulating redox signaling and tissue homeostasis. Here, we aimed to evaluate the role and mechanism of Nrf3 in injury-induced pathological cardiac remodeling. METHODS Global (Nrf3-KO) and CM-specific (Nrf3△CM) Nrf3 knockout mice were subjected to MI or ischemia/reperfusion injury, followed by functional and histopathological analysis. Primary neonatal mouse and rat ventricular myocytes and CMs derived from human induced pluripotent stem cells were used to evaluate the impact of Nrf3 on CM apoptosis and mitochondrial ROS production. Chromatin immunoprecipitation sequencing and immunoprecipitation-mass spectrometry analysis were used to uncover potential targets of Nrf3. MitoParaquat administration and CM-specific adeno-associated virus vectors were used to further confirm the in vivo relevance of the identified signal pathways. RESULTS Nrf3 was expressed mainly in CMs in healthy human hearts, and an increased level of Nrf3 was observed in CMs within the border zone of infarcted human hearts and murine cardiac tissues after MI. Both global and CM-specific Nrf3 knockout significantly decreased injury-induced mitochondrial ROS production, CM apoptosis, and pathological cardiac remodeling, consequently improving cardiac functions. In addition, cardiac-specific Nrf3 overexpression reversed the ameliorative cardiac phenotypes observed in Nrf3-KO mice. Functional studies showed that Nrf3 promoted neonatal mouse ventricular myocyte, neonatal rat ventricular myocyte, and CMs derived from human induced pluripotent stem cell apoptosis by increasing mitochondrial ROS production. Critically, augmenting mitochondrial ROS with MitoParaquat blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling. Mechanistically, a redox regulator Pitx2 (paired-like homeodomain transcription factor 2) was identified as one of the main target genes of Nrf3. Specifically, Nrf3 binds to Pitx2 promoter, where it increases DNA methylation through recruiting heterogeneous nuclear ribonucleoprotein K and DNA-methyltransferase 1 complex, thereby inhibiting Pitx2 expression. CM-specific knockdown of Pitx2 blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling, and cardiac-specific Pitx2 overexpression attenuated MI-induced mitochondrial ROS production and CM apoptosis, as well as preserved cardiac functions after MI. CONCLUSIONS Nrf3 promotes injury-induced CM apoptosis and deteriorates cardiac functions by increasing mitochondrial ROS production through suppressing Pitx2 expression. Targeting the Nrf3-Pitx2-mitochondrial ROS signal axis may therefore represent a novel therapeutic approach for MI treatment.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Ancheng Zheng
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Xiaolei Xu
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Zhenning Shi
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Mei Yang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Shasha Sun
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Leyu Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Yumeng Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Haige Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (H.Z.)
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Li Zhang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| |
Collapse
|
2
|
Bai Y, Chen L, Guo F, Zhang J, Hu J, Tao X, Lu Q, Li W, Chen X, Gong T, Qiu N, Jin Y, Yang L, Lei Y, Ruan C, Jing Q, Cooke JP, Wang S, Zou Y, Ge J. EphrinB2-mediated CDK5/ISL1 pathway enhances cardiac lymphangiogenesis and alleviates ischemic injury by resolving post-MI inflammation. Signal Transduct Target Ther 2024; 9:326. [PMID: 39557830 PMCID: PMC11574162 DOI: 10.1038/s41392-024-02019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
EphrinB2 (erythropoietin-producing hepatoma interactor B2) is a key Eph/ephrin family member, promoting angiogenesis, vasculogenesis, and lymphangiogenesis during embryonic development. However, the role of EphrinB2 in cardiac lymphangiogenesis following myocardial infarction (MI) and the potential molecular mechanism remains to be demonstrated. This study revealed that EphrinB2 prevented ischemic heart post-MI from remodeling and dysfunction by activating the cardiac lymphangiogenesis signaling pathway. Deletion of EphrinB2 impaired cardiac lymphangiogenesis and aggravated adverse cardiac remodeling and ventricular dysfunction post-MI. At the same time, overexpression of EphrinB2 stimulated cardiac lymphangiogenesis which facilitated cardiac infiltrating macrophage drainage and reduced inflammation in the ischemic heart. The beneficial effects of EphrinB2 on improving clearance of inflammatory response and cardiac function were abolished in Lyve1 knockout mice. Mechanistically, EphrinB2 accelerated cell cycling and lymphatic endothelial cell proliferation and migration by activating CDK5 and CDK5-dependent ISL1 nuclear translocation. EphrinB2 enhanced the transcriptional activity of ISL1 at the VEGFR3 (FLT4) promoter, and VEGFR3 inhibitor MAZ51 significantly diminished the EphrinB2-mediated lymphangiogenesis and deteriorated the ischemic cardiac function. We uncovered a novel mechanism of EphrinB2-driven cardiac lymphangiogenesis in improving myocardial remodeling and function after MI.
Collapse
Affiliation(s)
- Yingnan Bai
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Liming Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fanghao Guo
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinghong Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinlin Hu
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuefei Tao
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qing Lu
- Department of Radiology, Shanghai Dongfang Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Wenyi Li
- Department of Endocrinology, Tongren Hospital, Shanghai JiaoMo Tong University School of Medicine, Shanghai, China
| | - Xueying Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ting Gong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Nan Qiu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yawei Jin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lifan Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, USA
| | - Shijun Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
- Minhang Hospital, Fudan University, Shanghai, China.
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
- Institute of Advanced Medicine, Henan University, Kaifeng, Henan, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Shafi O, Siddiqui G, Jaffry HA. The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review. BMC Cancer 2023; 23:1245. [PMID: 38110859 PMCID: PMC10726542 DOI: 10.1186/s12885-023-11723-3] [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: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions. METHODS Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes. RESULTS The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others. The Nkx2-5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2-5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential. CONCLUSION The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| | - Hassam A Jaffry
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
4
|
Shen Y, Kim IM, Tang Y. Uncovering the Heterogeneity of Cardiac Lin-KIT+ Cells: A scRNA-seq Study on the Identification of Subpopulations. Stem Cells 2023; 41:958-970. [PMID: 37539750 PMCID: PMC11009691 DOI: 10.1093/stmcls/sxad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/11/2023] [Indexed: 08/05/2023]
Abstract
The reparative potential of cardiac Lin-KIT+ (KIT) cells is influenced by their population, but identifying their markers is challenging due to changes in phenotype during in vitro culture. Resolving this issue requires uncovering cell heterogeneity and discovering new subpopulations. Single-cell RNA sequencing (scRNA-seq) can identify KIT cell subpopulations, their markers, and signaling pathways. We used 10× genomic scRNA-seq to analyze cardiac-derived cells from adult mice and found 3 primary KIT cell populations: KIT1, characterized by high-KIT expression (KITHI), represents a population of cardiac endothelial cells; KIT2, which has low-KIT expression (KITLO), expresses transcription factors such as KLF4, MYC, and GATA6, as well as genes involved in the regulation of angiogenic cytokines; KIT3, with moderate KIT expression (KITMOD), expresses the cardiac transcription factor MEF2C and mesenchymal cell markers such as ENG. Cell-cell communication network analysis predicted the presence of the 3 KIT clusters as signal senders and receivers, including VEGF, CXCL, and BMP signaling. Metabolic analysis showed that KIT1 has the low activity of glycolysis and oxidative phosphorylation (OXPHOS), KIT2 has high glycolytic activity, and KIT3 has high OXPHOS and fatty acid degradation activity, indicating distinct metabolic adaptations of the 3 KIT populations. Through the systemic infusion of KIT1 cells in a mouse model of myocardial infarction, we observed their involvement in promoting the formation of new micro-vessels. In addition, in vitro spheroid culture experiments demonstrated the cardiac differentiation capacity of KIT2 cells.
Collapse
Affiliation(s)
- Yan Shen
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-Man Kim
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Yaoliang Tang
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
5
|
Ren J, Miao D, Li Y, Gao R. Spotlight on Isl1: A Key Player in Cardiovascular Development and Diseases. Front Cell Dev Biol 2021; 9:793605. [PMID: 34901033 PMCID: PMC8656156 DOI: 10.3389/fcell.2021.793605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/10/2021] [Indexed: 02/01/2023] Open
Abstract
Cardiac transcription factors orchestrate a regulatory network controlling cardiovascular development. Isl1, a LIM-homeodomain transcription factor, acts as a key player in multiple organs during embryonic development. Its crucial roles in cardiovascular development have been elucidated by extensive studies, especially as a marker gene for the second heart field progenitors. Here, we summarize the roles of Isl1 in cardiovascular development and function, and outline its cellular and molecular modes of action, thus providing insights for the molecular basis of cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Ren
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - Danxiu Miao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China.,Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Yanshu Li
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Rui Gao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
6
|
Wang Z, Schwartz RJ, Liu J, Sun F, Li Q, Ma Y. Smyd1 Orchestrates Early Heart Development Through Positive and Negative Gene Regulation. Front Cell Dev Biol 2021; 9:654682. [PMID: 33869215 PMCID: PMC8047137 DOI: 10.3389/fcell.2021.654682] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/17/2021] [Indexed: 01/09/2023] Open
Abstract
SET and MYND domain-containing protein 1 (Smyd1) is a striated muscle-specific histone methyltransferase. Our previous work demonstrated that deletion of Smyd1 in either cardiomyocytes or the outflow tract (OFT) resulted in embryonic lethality at E9.5, with cardiac structural defects such as truncation of the OFT and right ventricle and impaired expansion and proliferation of the second heart field (SHF). The cardiac phenotype was accompanied by the downregulation of ISL LIM Homeobox 1 (Isl1) and upregulation of atrial natriuretic factor (ANF). However, the mechanisms of Smyd1 regulating Isl1 and ANF during embryonic heart development remain to be elucidated. Here, we employed various biochemical and molecular biological approaches including chromatin immunoprecipitation polymerase chain reaction (ChIP-PCR), pGL3 fluorescence reporter system, and co-immunoprecipitation (CoIP) and found that Smyd1 interacted with absent small homeotic-2-like protein (ASH2L) and activated the promoter of Isl1 by trimethylating H3K4. We also found that Smyd1 associated with HDAC to repress ANF expression using trichostatin A (TSA), a deacetylase inhibitor. In conclusion, Smyd1 participates in early heart development by upregulating the expression of Isl1 and downregulating the expression of ANF.
Collapse
Affiliation(s)
- Zhen Wang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Jing Liu
- Department of Reproductive Medicine Center, Zhengzhou University, Zhengzhou, China
| | - Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.,Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| |
Collapse
|
7
|
Wang L, Serpooshan V, Zhang J. Engineering Human Cardiac Muscle Patch Constructs for Prevention of Post-infarction LV Remodeling. Front Cardiovasc Med 2021; 8:621781. [PMID: 33718449 PMCID: PMC7952323 DOI: 10.3389/fcvm.2021.621781] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering combines principles of engineering and biology to generate living tissue equivalents for drug testing, disease modeling, and regenerative medicine. As techniques for reprogramming human somatic cells into induced pluripotent stem cells (iPSCs) and subsequently differentiating them into cardiomyocytes and other cardiac cells have become increasingly efficient, progress toward the development of engineered human cardiac muscle patch (hCMP) and heart tissue analogs has accelerated. A few pilot clinical studies in patients with post-infarction LV remodeling have been already approved. Conventional methods for hCMP fabrication include suspending cells within scaffolds, consisting of biocompatible materials, or growing two-dimensional sheets that can be stacked to form multilayered constructs. More recently, advanced technologies, such as micropatterning and three-dimensional bioprinting, have enabled fabrication of hCMP architectures at unprecedented spatiotemporal resolution. However, the studies working on various hCMP-based strategies for in vivo tissue repair face several major obstacles, including the inadequate scalability for clinical applications, poor integration and engraftment rate, and the lack of functional vasculature. Here, we review many of the recent advancements and key concerns in cardiac tissue engineering, focusing primarily on the production of hCMPs at clinical/industrial scales that are suitable for administration to patients with myocardial disease. The wide variety of cardiac cell types and sources that are applicable to hCMP biomanufacturing are elaborated. Finally, some of the key challenges remaining in the field and potential future directions to address these obstacles are discussed.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
8
|
Birla RK. A methodological nine-step process to bioengineer heart muscle tissue. Tissue Cell 2020; 67:101425. [PMID: 32853859 DOI: 10.1016/j.tice.2020.101425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 01/15/2023]
Abstract
Research in the field of heart muscle tissue engineering is focused on the fabrication of heart muscle tissue which can be utilized to repair, replace and/or augment functionality of damaged and/or diseased tissue. In the simplest embodiment, bioengineering heart muscle tissue constructs involves culture of cardiomyocytes within natural or synthetic scaffolds. Functional integration of the cells with the scaffold and subsequent remodeling lead to the formation of 3D heart muscle tissue and physiological cues like mechanical stretch, electrical stimulation and perfusion are necessary to guide tissue maturation and development. Potential applications for bioengineered heart muscle include use as grafts to repair or replace damaged tissue, as models for basic research and as tools for high-throughput screening of pharmacological agents. In this article, we provide a methodological process to bioengineer functional 3D heart muscle tissue and discuss state of the art and potential challenges in each of the nine-step tissue fabrication process.
Collapse
Affiliation(s)
- Ravi K Birla
- BIOLIFE4D, 2450 Holcombe Blvd; Houston, TX, 77204, United States.
| |
Collapse
|
9
|
Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis. Cell Death Dis 2020; 11:354. [PMID: 32393784 PMCID: PMC7214429 DOI: 10.1038/s41419-020-2508-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs)-derived cardiovascular progenitor cells (CVPCs) are a promising source for myocardial repair, while the mechanisms remain largely unknown. Extracellular vesicles (EVs) are known to mediate cell–cell communication, however, the efficacy and mechanisms of hPSC-CVPC-secreted EVs (hCVPC-EVs) in the infarct healing when given at the acute phase of myocardial infarction (MI) are unknown. Here, we report the cardioprotective effects of the EVs secreted from hESC-CVPCs under normoxic (EV-N) and hypoxic (EV-H) conditions in the infarcted heart and the long noncoding RNA (lncRNA)-related mechanisms. The hCVPC-EVs were confirmed by electron microscopy, nanoparticle tracking, and immunoblotting analysis. Injection of hCVPC-EVs into acutely infracted murine myocardium significantly improved cardiac function and reduced fibrosis at day 28 post MI, accompanied with the improved vascularization and cardiomyocyte survival at border zones. Consistently, hCVPC-EVs enhanced the tube formation and migration of human umbilical vein endothelial cells (HUVECs), improved the cell viability, and attenuated the lactate dehydrogenase release of neonatal rat cardiomyocytes (NRCMs) with oxygen glucose deprivation (OGD) injury. Moreover, the improvement of the EV-H in cardiomyocyte survival and tube formation of HUVECs was significantly better than these in the EV-N. RNA-seq analysis revealed a high abundance of the lncRNA MALAT1 in the EV-H. Its abundance was upregulated in the infarcted myocardium and cardiomyocytes treated with hCVPC-EVs. Overexpression of human MALAT1 improved the cell viability of NRCM with OGD injury, while knockdown of MALAT1 inhibited the hCVPC-EV-promoted tube formation of HUVECs. Furthermore, luciferase activity assay, RNA pull-down, and manipulation of miR-497 levels showed that MALAT1 improved NRCMs survival and HUVEC tube formation through targeting miR-497. These results reveal that hCVPC-EVs promote the infarct healing through improvement of cardiomyocyte survival and angiogenesis. The cardioprotective effects of hCVPC-EVs can be enhanced by hypoxia-conditioning of hCVPCs and are partially contributed by MALAT1 via targeting the miRNA.
Collapse
|
10
|
Fields MA, Del Priore LV, Adelman RA, Rizzolo LJ. Interactions of the choroid, Bruch's membrane, retinal pigment epithelium, and neurosensory retina collaborate to form the outer blood-retinal-barrier. Prog Retin Eye Res 2019; 76:100803. [PMID: 31704339 DOI: 10.1016/j.preteyeres.2019.100803] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023]
Abstract
The three interacting components of the outer blood-retinal barrier are the retinal pigment epithelium (RPE), choriocapillaris, and Bruch's membrane, the extracellular matrix that lies between them. Although previously reviewed independently, this review integrates these components into a more wholistic view of the barrier and discusses reconstitution models to explore the interactions among them. After updating our understanding of each component's contribution to barrier function, we discuss recent efforts to examine how the components interact. Recent studies demonstrate that claudin-19 regulates multiple aspects of RPE's barrier function and identifies a barrier function whereby mutations of claudin-19 affect retinal development. Co-culture approaches to reconstitute components of the outer blood-retinal barrier are beginning to reveal two-way interactions between the RPE and choriocapillaris. These interactions affect barrier function and the composition of the intervening Bruch's membrane. Normal or disease models of Bruch's membrane, reconstituted with healthy or diseased RPE, demonstrate adverse effects of diseased matrix on RPE metabolism. A stumbling block for reconstitution studies is the substrates typically used to culture cells are inadequate substitutes for Bruch's membrane. Together with human stem cells, the alternative substrates that have been designed offer an opportunity to engineer second-generation culture models of the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Ron A Adelman
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA; Department of Surgery, Yale University School of Medicine, PO Box 208062, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
11
|
Cardiac Progenitors Induced from Human Induced Pluripotent Stem Cells with Cardiogenic Small Molecule Effectively Regenerate Infarcted Hearts and Attenuate Fibrosis. Shock 2019; 50:627-639. [PMID: 29485473 DOI: 10.1097/shk.0000000000001133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cardiac progenitor cells (CPCs) being multipotent offer a promising source for cardiac repair due to their ability to proliferate and multiply into cardiac lineage cells. Here, we explored a novel strategy for human CPCs generation from human induced pluripotent stem cells (hiPSCs) using a cardiogenic small molecule, isoxazole (ISX-9) and their ability to grow in the scar tissue for functional improvement in the infarcted myocardium. CPCs were induced from hiPSCs with ISX-9. CPCs were characterized by immunocytochemistry and RT-PCR. The CPC survival and differentiation in the infarcted hearts were determined by in vivo transplantation in immunodeficient mice following left anterior descending artery ligation and their effects were determined on fibrosis and functional improvement. ISX-9 simultaneously induced expression of cardiac transcription factors, NK2 homeobox 5, islet-1, GATA binding protein 4, myocyte enhancer factor-2 in hiPSCs within 3 days of treatment and successfully differentiated into three cardiac lineages in vitro. Messenger RNA and microRNA-sequencing results showed that ISX-9 targeted multiple cardiac differentiation, proliferation signaling pathways and upregulated myogenesis and cardiac hypertrophy related-microRNA. ISX-9 activated multiple pathways including transforming growth factor β induced epithelial-mesenchymal transition signaling, canonical, and non-canonical Wnt signaling at different stages of cardiac differentiation. CPCs transplantation promoted myoangiogenesis, attenuated fibrosis, and led to functional improvement in treated mice.
Collapse
|
12
|
Wang J, Liu M, Wu Q, Li Q, Gao L, Jiang Y, Deng B, Huang W, Bi W, Chen Z, Chin YE, Paul C, Wang Y, Yang HT. Human Embryonic Stem Cell-Derived Cardiovascular Progenitors Repair Infarcted Hearts Through Modulation of Macrophages via Activation of Signal Transducer and Activator of Transcription 6. Antioxid Redox Signal 2019; 31:369-386. [PMID: 30854870 PMCID: PMC6602123 DOI: 10.1089/ars.2018.7688] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aims: Human embryonic stem cell derived-cardiovascular progenitor cells (hESC-CVPCs) are a promising cell source for cardiac repair, while the underlying mechanisms need to be elucidated. We recently observed cardioprotective effects of human pluripotent stem cell (hPSC)-CVPCs in infarcted nonhuman primates, but their effects on inflammation during early phase of myocardial infarction (MI) and the contribution of such effect to the cardioprotection are unclear. Results: Injection of hESC-CVPCs into acutely infarcted myocardium significantly ameliorated the functional worsening and scar formation, concomitantly with reduced inflammatory reactions and cardiomyocyte apoptosis as well as increased vascularization. Moreover, hESC-CVPCs modulated cardiac macrophages toward a reparative phenotype in the infarcted hearts, and such modulation was further confirmed in vitro using human cardiovascular progenitor cell (hCVPC)-conditioned medium (hCVPC-CdM) and highly contained interleukin (IL)-4/IL-13. Furthermore, signal transducer and activator of transcription 6 (STAT6) was activated in hCVPC-CdM- and IL-4/IL-13-treated macrophages in vitro and in hESC-CVPC-implanted MI hearts, resulting in the polarization of macrophages toward a reparative phenotype in the post-MI hearts. However, hESC-CVPC-mediated modulation on macrophages and cardioprotection were abolished in STAT6-deficient MI mice. Innovation: This is the first report about the immunoregulatory role played by hESC-CVPCs in the macrophage polarization in the infarcted hearts, its importance for the infarct repair, and the underlying signaling pathway. The findings provide new insight into the mechanism of microenvironmental regulation of stem cell-based therapy during acute MI. Conclusions: Implantion of hESC-CVPCs during the early phase of MI promotes infarct repair via the modulation of macrophage polarization through secreted cytokine-mediated STAT6 activation. The findings suggest a therapeutic potential by modulating macrophage polarization during acute phase of MI.
Collapse
Affiliation(s)
- Jinxi Wang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Meilan Liu
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Qiang Wu
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Qiang Li
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ling Gao
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yun Jiang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Boxiong Deng
- 3 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Tumor and Stem Cell, SIBS, Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China
| | - Wei Huang
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Wei Bi
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhongyan Chen
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| | - Y Eugene Chin
- 3 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Tumor and Stem Cell, SIBS, Chinese Academy of Sciences (CAS), Shanghai, People's Republic of China
| | - Christian Paul
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Yigang Wang
- 4 Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Huang-Tian Yang
- 1 CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Jiao Tong University School of Medicine and Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences (CAS), Chinese Academy of Sciences, Shanghai, People's Republic of China.,2 Institute for Stem Cell and Regeneration, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|
13
|
Camberos V, Baio J, Bailey L, Hasaniya N, Lopez LV, Kearns-Jonker M. Effects of Spaceflight and Simulated Microgravity on YAP1 Expression in Cardiovascular Progenitors: Implications for Cell-Based Repair. Int J Mol Sci 2019; 20:E2742. [PMID: 31167392 PMCID: PMC6600678 DOI: 10.3390/ijms20112742] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 01/06/2023] Open
Abstract
Spaceflight alters many processes of the human body including cardiac function and cardiac progenitor cell behavior. The mechanism behind these changes remains largely unknown; however, simulated microgravity devices are making it easier for researchers to study the effects of microgravity. To study the changes that take place in cardiac progenitor cells in microgravity environments, adult cardiac progenitor cells were cultured aboard the International Space Station (ISS) as well as on a clinostat and examined for changes in Hippo signaling, a pathway known to regulate cardiac development. Cells cultured under microgravity conditions, spaceflight-induced or simulated, displayed upregulation of downstream genes involved in the Hippo pathway such as YAP1 and SOD2. YAP1 is known to play a role in cardiac regeneration which led us to investigate YAP1 expression in a sheep model of cardiovascular repair. Additionally, to mimic the effects of microgravity, drug treatment was used to induce Hippo related genes as well as a regulator of the Hippo pathway, miRNA-302a. These studies provide insight into the changes that occur in space and how the effects of these changes relate to cardiac regeneration studies.
Collapse
Affiliation(s)
- Victor Camberos
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Jonathan Baio
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Leonard Bailey
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Nahidh Hasaniya
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Larry V Lopez
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
14
|
Hong SP, Song S, Lee S, Jo H, Kim HK, Han J, Park JH, Cho SW. Regenerative potential of mouse embryonic stem cell-derived PDGFRα + cardiac lineage committed cells in infarcted myocardium. World J Stem Cells 2019; 11:44-54. [PMID: 30705714 PMCID: PMC6354102 DOI: 10.4252/wjsc.v11.i1.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/06/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pluripotent stem cell-derived cardiomyocytes (CMs) have become one of the most attractive cellular resources for cell-based therapy to rescue damaged cardiac tissue.
AIM We investigated the regenerative potential of mouse embryonic stem cell (ESC)-derived platelet-derived growth factor receptor-α (PDGFRα)+ cardiac lineage-committed cells (CLCs), which have a proliferative capacity but are in a morphologically and functionally immature state compared with differentiated CMs.
METHODS We induced mouse ESCs into PDGFRα+ CLCs and αMHC+ CMs using a combination of the small molecule cyclosporin A, the rho-associated coiled-coil kinase inhibitor Y27632, the antioxidant Trolox, and the ALK5 inhibitor EW7197. We implanted PDGFRα+ CLCs and differentiated αMHC+ CMs into a myocardial infarction (MI) murine model and performed functional analysis using transthoracic echocardiography (TTE) and histologic analysis.
RESULTS Compared with the untreated MI hearts, the anterior and septal regional wall motion and systolic functional parameters were notably and similarly improved in the MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs based on TTE. In histologic analysis, the untreated MI hearts contained a thinner ventricular wall than did the controls, while the ventricular walls of MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs were similarly thicker compared with that of the untreated MI hearts. Furthermore, implanted PDGFRα+ CLCs aligned and integrated with host CMs and were mostly differentiated into α-actinin+ CMs, and they did not convert into CD31+ endothelial cells or αSMA+ mural cells.
CONCLUSION PDGFRα+ CLCs from mouse ESCs exhibiting proliferative capacity showed a regenerative effect in infarcted myocardium. Therefore, mouse ESC-derived PDGFRα+ CLCs may represent a potential cellular resource for cardiac regeneration.
Collapse
Affiliation(s)
- Seon Pyo Hong
- Center for Vascular Research, Institute of Basic Science (IBS), Daejeon 34141, South Korea
| | - Sukhyun Song
- Center for Vascular Research, Institute of Basic Science (IBS), Daejeon 34141, South Korea
| | - Seungjoo Lee
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hyeonju Jo
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Department of Physiology, Department of Health Sciences and Technology, BK21 plus Project Team, Inje University College of Medicine, Busan 47392, South Korea
| | - Jae-Hyeong Park
- Department of Cardiology in Internal Medicine, School of Medicine, Chungnam National University Hospital, Chungnam National University, Daejeon 35015, South Korea
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Seoul Paik Hospital, Seoul 04551, South Korea
- Cardiovascular and Metabolic Disease Center, Inje University College of Medicine, Busan 47392, South Korea
| |
Collapse
|
15
|
Hong SP, Song S, Lee S, Jo H, Kim HK, Han J, Park JH, Cho SW. Regenerative potential of mouse embryonic stem cell-derived PDGFRα + cardiac lineage committed cells in infarcted myocardium. World J Stem Cells 2019. [DOI: 10.4252/wjsc.v11.i1.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
16
|
Abstract
Some of the most significant leaps in the history of modern civilization-the development of article in China, the steam engine, which led to the European industrial revolution, and the era of computers-have occurred when science converged with engineering. Recently, the convergence of human pluripotent stem cell technology with biomaterials and bioengineering have launched a new medical innovation: functional human engineered tissue, which promises to revolutionize the treatment of failing organs including most critically, the heart. This compendium covers recent, state-of-the-art developments in the fields of cardiovascular tissue engineering, as well as the needs and challenges associated with the clinical use of these technologies. We have not attempted to provide an exhaustive review in stem cell biology and cardiac cell therapy; many other important and influential reports are certainly merit but already been discussed in several recent reviews. Our scope is limited to the engineered tissues that have been fabricated to repair or replace components of the heart (eg, valves, vessels, contractile tissue) that have been functionally compromised by diseases or developmental abnormalities. In particular, we have focused on using an engineered myocardial tissue to mitigate deficiencies in contractile function.
Collapse
Affiliation(s)
- Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Wuqiang Zhu
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham (J.Z., W.Z.)
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada (M.R.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering and Department of Medicine, Columbia University, New York, NY (G.V.-N.)
| |
Collapse
|
17
|
Li B, Meng X, Zhang L. microRNAs and cardiac stem cells in heart development and disease. Drug Discov Today 2018; 24:233-240. [PMID: 29852125 DOI: 10.1016/j.drudis.2018.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/24/2018] [Accepted: 05/22/2018] [Indexed: 12/20/2022]
Abstract
Cumulative evidence has proven that proliferation, differentiation and migration of cardiac stem cells (CSCs) dominate early heart development and contribute to the later occurrence of heart disease. Among other mechanisms, microRNAs work as the 'fine-tuning' to modulate the levels of target genes in a specific cell type. The distinct microRNA signatures in CSCs reveal the stages and functions of CSCs. The focus of this review is to summarize recent knowledge advances in CSC proliferation, differentiation and migration and to discuss how microRNAs regulate these processes during heart development and in heart disease. Better understanding of microRNA regulation on CSCs under different situations will enable the unveiling of the mechanisms of heart disease and open new avenues in the therapeutic potentials of microRNA modulation to treat heart disease.
Collapse
Affiliation(s)
- Bo Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Xianmei Meng
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
18
|
Singh D, Wang SB, Xia T, Tainsh L, Ghiassi-Nejad M, Xu T, Peng S, Adelman RA, Rizzolo LJ. A biodegradable scaffold enhances differentiation of embryonic stem cells into a thick sheet of retinal cells. Biomaterials 2018; 154:158-168. [DOI: 10.1016/j.biomaterials.2017.10.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 12/25/2022]
|
19
|
Wang L, Meier EM, Tian S, Lei I, Liu L, Xian S, Lam MT, Wang Z. Transplantation of Isl1 + cardiac progenitor cells in small intestinal submucosa improves infarcted heart function. Stem Cell Res Ther 2017; 8:230. [PMID: 29037258 PMCID: PMC5644064 DOI: 10.1186/s13287-017-0675-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/02/2017] [Accepted: 09/14/2017] [Indexed: 11/21/2022] Open
Abstract
Background Application of cardiac stem cells combined with biomaterial scaffold is a promising therapeutic strategy for heart repair after myocardial infarction. However, the optimal cell types and biomaterials remain elusive. Methods In this study, we seeded Isl1+ embryonic cardiac progenitor cells (CPCs) into decellularized porcine small intestinal submucosa extracellular matrix (SIS-ECM) to assess the therapeutic potential of Isl1+ CPCs and the biocompatibility of SIS-ECM with these cells. Results We observed that SIS-ECM supported the viability and attachment of Isl1+ CPCs. Importantly, Isl1+ CPCs differentiated into cardiomyocytes and endothelial cells 7 days after seeding into SIS-ECM. In addition, SIS-ECM with CPC-derived cardiomyocytes showed spontaneous contraction and responded to β-adrenergic stimulation. Next, patches of SIS-ECM seeded with CPCs for 7 days were transplanted onto the outer surface of infarcted myocardium in mice. Four weeks after transplantation, the patches were tightly attached to the surface of the host myocardium and remained viable. Transplantation of patches improved cardiac function, decreased the left ventricular myocardial scarring area, and reduced fibrosis and heart failure. Conclusions Transplantation of Isl1+ CPCs seeded in SIS-ECM represents an effective approach for cell-based heart therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0675-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lingjun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elizabeth M Meier
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48201, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI, 48109, USA.,Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Liu Liu
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shaoxiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Mai T Lam
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48201, USA.,Cardiovascular Research Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
20
|
Cardiac stem cells for myocardial regeneration: promising but not ready for prime time. Curr Opin Biotechnol 2017; 47:30-35. [PMID: 28591641 DOI: 10.1016/j.copbio.2017.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 02/06/2023]
Abstract
Remarkable strides have been made in the treatment of ischemic heart disease in decades. As the initial loss of cardiomyocytes associated with myocardial infarction serves as an impetus for myocardial remodeling, the ability to replace these cells with healthy counterparts would represent an effective treatment for many forms of cardiovascular disease. The discovery of cardiac stem cells (that can differentiate into multiple lineages) highlighted the possibility for development of cell-based therapeutics to achieve this ultimate goal. Recent research features cardiac stem cell maintenance, proliferation, and differentiation, as well as direct reprogramming of various somatic cells into cardiomyocytes, all within the context of the holy grail of regeneration of the injured heart. Much work remains to be done, but the future looks bright!
Collapse
|
21
|
Li Y, Tian S, Lei I, Liu L, Ma P, Wang Z. Transplantation of multipotent Isl1+ cardiac progenitor cells preserves infarcted heart function in mice. Am J Transl Res 2017; 9:1530-1542. [PMID: 28386378 PMCID: PMC5376043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/18/2017] [Indexed: 06/07/2023]
Abstract
Cell-based cardiac therapy is a promising therapeutic strategy to restore heart function after myocardial infarction (MI). However, the cell type selection and ensuing effects remain controversial. Here, we intramyocardially injected Isl1+ cardiac progenitor cells (CPCs) derived from EGFP/luciferase double-tagged mouse embryonic stem (dt-mES) cells with vehicle (fibrin gel) or phosphate-buffered saline (PBS) into the infarcted area in nude mice to assess the contribution of CPCs to the recovery of cardiac function post-MI. Our results showed that Isl1+ CPCs differentiated normally into three cardiac lineages (cardiomyocytes (CMs), endothelial cells and smooth muscle cells) both on cell culture plates and in fibrin gel. Cell retention was significantly increased when the transplanted cells were injected with vehicle. Importantly, 28 days after injection, CPCs were observed to differentiate into CMs within the infarcted area. Moreover, numerous CD31+ endothelial cells derived from endogenous revascularization and differentiation of the injected CPCs were detected. SMMHC-, Ki67- and CX-43-positive cells were identified in the injected CPC population, further demonstrating the proliferation, differentiation and integration of the transplanted CPCs in host cells. Furthermore, animal hearts injected with CPCs showed increased angiogenesis, decreased infarct size, and improved heart function. In conclusion, our studies showed that Isl1+ CPCs, when combined with a suitable vehicle, can produce notable therapeutic effects in the infarcted heart, suggesting that CPCs might be an ideal cell source for cardiac therapy.
Collapse
Affiliation(s)
- Yunpeng Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical UniversityXian 710032, Shaanxi, China
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
- Faculty of Health Sciences, University of MacauMacau SAR, China
| | - Liu Liu
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Peter Ma
- Department of Biologic and Materials Sciences, Biomedical Engineering, Macromolecular Science and Engineering Center, and Materials Science and Engineering, The University of MichiganAnn Arbor 48109, MI, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| |
Collapse
|
22
|
Anisotropic engineered heart tissue made from laser-cut decellularized myocardium. Sci Rep 2016; 6:32068. [PMID: 27572147 PMCID: PMC5004193 DOI: 10.1038/srep32068] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
We have developed an engineered heart tissue (EHT) system that uses laser-cut sheets of decellularized myocardium as scaffolds. This material enables formation of thin muscle strips whose biomechanical characteristics are easily measured and manipulated. To create EHTs, sections of porcine myocardium were laser-cut into ribbon-like shapes, decellularized, and mounted in specialized clips for seeding and culture. Scaffolds were first tested by seeding with neonatal rat ventricular myocytes. EHTs beat synchronously by day five and exhibited robust length-dependent activation by day 21. Fiber orientation within the scaffold affected peak twitch stress, demonstrating its ability to guide cells toward physiologic contractile anisotropy. Scaffold anisotropy also made it possible to probe cellular responses to stretch as a function of fiber angle. Stretch that was aligned with the fiber direction increased expression of brain natriuretic peptide, but off-axis stretches (causing fiber shear) did not. The method also produced robust EHTs from cardiomyocytes derived from human embryonic stem cells and induced pluripotent stem cells (hiPSC). hiPSC-EHTs achieved maximum peak stress of 6.5 mN/mm2 and twitch kinetics approaching reported values from adult human trabeculae. We conclude that laser-cut EHTs are a viable platform for novel mechanotransduction experiments and characterizing the biomechanical function of patient-derived cardiomyoctyes.
Collapse
|