51
|
Tang Y, Zhao L, Yu X, Zhang J, Qian L, Jin J, Lu R, Zhou Y. Inhibition of EZH2 primes the cardiac gene activation via removal of epigenetic repression during human direct cardiac reprogramming. Stem Cell Res 2021; 53:102365. [PMID: 34087994 PMCID: PMC8238038 DOI: 10.1016/j.scr.2021.102365] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease, until now, is still the leading cause of death in the United States. Due to the limited regenerative capacity of adult hearts, the damage caused by heart injury cannot be reversed and eventually progress into heart failure. In need of cardiovascular disease treatment, many therapies aimed at either cell transplantation or cell regeneration have been proposed. Direct reprogramming of somatic cells into induced cardiomyocytes (iCMs) is considered to be a promising strategy for regenerative medicine. The induction of cardiomyocytes from non-myocytes can be achieved efficiently via ectopic expression of reprogramming factors both in vitro and in vivo in the mouse model, however, the generation of human induced cardiomyocyte-like cells (hiCMs) remains challenging. The inefficiency of hiCMs production called for the identification of the additional epigenetic memories in non-myocytes which might be damping the hiCM reprogramming. Here, we conducted an unbiased loss-of-function screening focusing on epigenetic regulators and identified enhancer of zeste homolog 2 (EZH2) as an important epigenetic barrier during hiCM reprogramming. We found that the removal of EZH2 via genetic knockdown or treatment of EZH2 selective degrader significantly increased the hiCM reprogramming efficiency and led to profound activation of cardiac genes and repression of collagen and extracellular matrix genes. Furthermore, EZH2 inhibitors targeting its catalytic activity also promotes hiCM reprogramming, suggesting that EZH2 may restrain cardiac conversion through H3K27me3-mediated gene repression. Indeed, genomic profiling of H3K27me3 revealed a subset of cardiac genes that remain repressed with high levels of H3K27me3 despite of the delivery of the reprogramming factors. Inhibition of EZH2, however, leads to reduced H3K27me3 occupancy and robust activation of these cardiac genes. Taken together, our data suggested that EZH2 inhibition facilitates the activation of cardiac genes in fibroblasts and eases the production of hiCMs.
Collapse
Affiliation(s)
- Yawen Tang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Lianzhong Zhao
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rui Lu
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yang Zhou
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
52
|
Direct cell reprogramming: approaches, mechanisms and progress. Nat Rev Mol Cell Biol 2021; 22:410-424. [PMID: 33619373 DOI: 10.1038/s41580-021-00335-z] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The reprogramming of somatic cells with defined factors, which converts cells from one lineage into cells of another, has greatly reshaped our traditional views on cell identity and cell fate determination. Direct reprogramming (also known as transdifferentiation) refers to cell fate conversion without transitioning through an intermediary pluripotent state. Given that the number of cell types that can be generated by direct reprogramming is rapidly increasing, it has become a promising strategy to produce functional cells for therapeutic purposes. This Review discusses the evolution of direct reprogramming from a transcription factor-based method to a small-molecule-driven approach, the recent progress in enhancing reprogrammed cell maturation, and the challenges associated with in vivo direct reprogramming for translational applications. It also describes our current understanding of the molecular mechanisms underlying direct reprogramming, including the role of transcription factors, epigenetic modifications, non-coding RNAs, and the function of metabolic reprogramming, and highlights novel insights gained from single-cell omics studies.
Collapse
|
53
|
Riching AS, Song K. Cardiac Regeneration: New Insights Into the Frontier of Ischemic Heart Failure Therapy. Front Bioeng Biotechnol 2021; 8:637538. [PMID: 33585427 PMCID: PMC7873479 DOI: 10.3389/fbioe.2020.637538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Ischemic heart disease is the leading cause of morbidity and mortality in the world. While pharmacological and surgical interventions developed in the late twentieth century drastically improved patient outcomes, mortality rates over the last two decades have begun to plateau. Following ischemic injury, pathological remodeling leads to cardiomyocyte loss and fibrosis leading to impaired heart function. Cardiomyocyte turnover rate in the adult heart is limited, and no clinical therapies currently exist to regenerate cardiomyocytes lost following ischemic injury. In this review, we summarize the progress of therapeutic strategies including revascularization and cell-based interventions to regenerate the heart: transiently inducing cardiomyocyte proliferation and direct reprogramming of fibroblasts into cardiomyocytes. Moreover, we highlight recent mechanistic insights governing these strategies to promote heart regeneration and identify current challenges in translating these approaches to human patients.
Collapse
Affiliation(s)
- Andrew S. Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
54
|
Regulation of cardiomyocyte fate plasticity: a key strategy for cardiac regeneration. Signal Transduct Target Ther 2021; 6:31. [PMID: 33500391 PMCID: PMC7838318 DOI: 10.1038/s41392-020-00413-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/11/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
With the high morbidity and mortality rates, cardiovascular diseases have become one of the most concerning diseases worldwide. The heart of adult mammals can hardly regenerate naturally after injury because adult cardiomyocytes have already exited the cell cycle, which subseqently triggers cardiac remodeling and heart failure. Although a series of pharmacological treatments and surgical methods have been utilized to improve heart functions, they cannot replenish the massive loss of beating cardiomyocytes after injury. Here, we summarize the latest research progress in cardiac regeneration and heart repair through altering cardiomyocyte fate plasticity, which is emerging as an effective strategy to compensate for the loss of functional cardiomyocytes and improve the impaired heart functions. First, residual cardiomyocytes in damaged hearts re-enter the cell cycle to acquire the proliferative capacity by the modifications of cell cycle-related genes or regulation of growth-related signals. Additionally, non-cardiomyocytes such as cardiac fibroblasts, were shown to be reprogrammed into cardiomyocytes and thus favor the repair of damaged hearts. Moreover, pluripotent stem cells have been shown to transform into cardiomyocytes to promote heart healing after myocardial infarction (MI). Furthermore, in vitro and in vivo studies demonstrated that environmental oxygen, energy metabolism, extracellular factors, nerves, non-coding RNAs, etc. play the key regulatory functions in cardiac regeneration. These findings provide the theoretical basis of targeting cellular fate plasticity to induce cardiomyocyte proliferation or formation, and also provide the clues for stimulating heart repair after injury.
Collapse
|
55
|
Varshney A, Chahal G, Santos L, Stolper J, Hallab JC, Nim HT, Nikolov M, Yip A, Ramialison M. Human Cardiac Transcription Factor Networks. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11597-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
56
|
Riching AS, Danis E, Zhao Y, Cao Y, Chi C, Bagchi RA, Klein BJ, Xu H, Kutateladze TG, McKinsey TA, Buttrick PM, Song K. Suppression of canonical TGF-β signaling enables GATA4 to interact with H3K27me3 demethylase JMJD3 to promote cardiomyogenesis. J Mol Cell Cardiol 2020; 153:44-59. [PMID: 33359755 DOI: 10.1016/j.yjmcc.2020.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 11/14/2020] [Accepted: 12/12/2020] [Indexed: 01/07/2023]
Abstract
Direct reprogramming of fibroblasts into cardiomyocytes (CMs) represents a promising strategy to regenerate CMs lost after ischemic heart injury. Overexpression of GATA4, HAND2, MEF2C, TBX5, miR-1, and miR-133 (GHMT2m) along with transforming growth factor beta (TGF-β) inhibition efficiently promote reprogramming. However, the mechanisms by which TGF-β blockade promotes cardiac reprogramming remain unknown. Here, we identify interactions between the histone H3 lysine 27 trimethylation (H3K27me3) demethylase JMJD3, the SWI/SNF remodeling complex subunit BRG1, and cardiac transcription factors. Furthermore, canonical TGF-β signaling regulates the interaction between GATA4 and JMJD3. TGF-β activation impairs the ability of GATA4 to bind target genes and prevents demethylation of H3K27 at cardiac gene promoters during cardiac reprogramming. Finally, a mutation in GATA4 (V267M) that is associated with congenital heart disease exhibits reduced binding to JMJD3 and impairs cardiomyogenesis. Thus, we have identified an epigenetic mechanism wherein canonical TGF-β pathway activation impairs cardiac gene programming, in part by interfering with GATA4-JMJD3 interactions.
Collapse
Affiliation(s)
- Andrew S Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Etienne Danis
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuanbiao Zhao
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yingqiong Cao
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rushita A Bagchi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brianna J Klein
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hongyan Xu
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Tatiana G Kutateladze
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Peter M Buttrick
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
57
|
Wang L, Ma H, Huang P, Xie Y, Near D, Wang H, Xu J, Yang Y, Xu Y, Garbutt T, Zhou Y, Liu Z, Yin C, Bressan M, Taylor JM, Liu J, Qian L. Down-regulation of Beclin1 promotes direct cardiac reprogramming. Sci Transl Med 2020; 12:eaay7856. [PMID: 33087505 PMCID: PMC8188650 DOI: 10.1126/scitranslmed.aay7856] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 05/07/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
Abstract
Direct reprogramming of fibroblasts to alternative cell fates by forced expression of transcription factors offers a platform to explore fundamental molecular events governing cell fate identity. The discovery and study of induced cardiomyocytes (iCMs) not only provides alternative therapeutic strategies for heart disease but also sheds lights on basic biology underlying CM fate determination. The iCM field has primarily focused on early transcriptome and epigenome repatterning, whereas little is known about how reprogramming iCMs remodel, erase, and exit the initial fibroblast lineage to acquire final cell identity. Here, we show that autophagy-related 5 (Atg5)-dependent autophagy, an evolutionarily conserved self-digestion process, was induced and required for iCM reprogramming. Unexpectedly, the autophagic factor Beclin1 (Becn1) was found to suppress iCM induction in an autophagy-independent manner. Depletion of Becn1 resulted in improved iCM induction from both murine and human fibroblasts. In a mouse genetic model, Becn1 haploinsufficiency further enhanced reprogramming factor-mediated heart function recovery and scar size reduction after myocardial infarction. Mechanistically, loss of Becn1 up-regulated Lef1 and down-regulated Wnt inhibitors, leading to activation of the canonical Wnt/β-catenin signaling pathway. In addition, Becn1 physically interacts with other classical class III phosphatidylinositol 3-kinase (PI3K III) complex components, the knockdown of which phenocopied Becn1 depletion in cardiac reprogramming. Collectively, our study revealed an inductive role of Atg5-dependent autophagy as well as a previously unrecognized autophagy-independent inhibitory function of Becn1 in iCM reprogramming.
Collapse
Affiliation(s)
- Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Peisen Huang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David Near
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yangxi Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tiffany Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chaoying Yin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael Bressan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
58
|
Jiang L, Liang J, Huang W, Wu Z, Paul C, Wang Y. Strategies and Challenges to Improve Cellular Programming-Based Approaches for Heart Regeneration Therapy. Int J Mol Sci 2020; 21:E7662. [PMID: 33081233 PMCID: PMC7589611 DOI: 10.3390/ijms21207662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
59
|
Zhou Y, Tian Q, Zheng C, Yang J, Fan J, Shentu Y. Myocardial infarction-induced anxiety-like behavior is associated with epigenetic alterations in the hippocampus of rat. Brain Res Bull 2020; 164:172-183. [PMID: 32871241 DOI: 10.1016/j.brainresbull.2020.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 01/01/2023]
Abstract
Epidemiological and experimental animal studies indicate that there is a high risk for the incidence of neuropsychiatric disorders suffering from cardiovascular diseases such as myocardial infarction (MI). However, the potential mechanism of this association remains largely unknown. This study sought to evaluate whether epigenetic alterations in the hippocampus is associated with MI-induced anxiety-like behavior in rats. MI was induced by occlusion of the left anterior descending artery in adult female rats. Anxiety-like behavior was examined by elevated plus maze, light-dark box, and open field test. Relative gene and protein levels expression in the hippocampus were tested by qRT-PCR and western blotting, respectively. We found that MI rats exhibited anxiety-like behavior compared with those in controls, and there is a positive correlation between MI and anxiety-like behavior. We also found that MI decreased KDM6B while increased SIRT1 expression in the hippocampus of MI rats relative to those in controls. In addition, MI not only increased levels of IL-1β, bax, and cleaved-caspase 3, but also increased Iba-1 and GFAP expression in the hippocampus, as compared to those in controls, suggesting a promotion of neuro-inflammation and apoptosis in hippocampus. Co-immunoprecipitation assay illustrated that H3K27me3 functioned by counteracting with YAP activation in the hippocampus of MI rats relative to those in controls. Together, these results suggest a potential role of hippocampal epigenetic signaling in MI-induced anxiety-like behavior in rats, and pharmacological targeting KDM6B or SIRT1 could be a strategy to ameliorate anxiety-like behavior induced by MI.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qiuyun Tian
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Chenfei Zheng
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jinge Yang
- Department of Medical Technology, Jiangxi Medical College, Shangrao, Jiangxi, 334709, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
60
|
Bmi1 inhibitor PTC-209 promotes Chemically-induced Direct Cardiac Reprogramming of cardiac fibroblasts into cardiomyocytes. Sci Rep 2020; 10:7129. [PMID: 32346096 PMCID: PMC7189257 DOI: 10.1038/s41598-020-63992-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
The development of therapeutic approaches based on direct cardiac reprogramming of fibroblasts into induced-cardiomyocytes (iCM) has emerged as an attractive strategy to repair the injured myocardium. The identification of the mechanisms driving lineage conversion represents a crucial step toward the development of new and more efficient regenerative strategies. To this aim, here we show that pre-treatment with the Bmi1 inhibitor PTC-209 is sufficient to increase the efficiency of Chemical-induced Direct Cardiac Reprogramming both in mouse embryonic fibroblasts and adult cardiac fibroblasts. PTC-209 induces an overall increase of spontaneously beating iCM at end-stage of reprogramming, expressing high levels of late cardiac markers Troponin T and myosin muscle light chain-2v. The inhibition of Bmi1 expression occurring upon PTC-209 pre-treatment was maintained throughout the reprogramming protocol, contributing to a significant gene expression de-regulation. RNA profiling revealed that, upon Bmi1 inhibition a significant down-regulation of genes associated with immune and inflammatory signalling pathways occurred, with repression of different genes involved in interleukin, cytokine and chemokine pathways. Accordingly, we observed the down-regulation of both JAK/STAT3 and MAPK/ERK1-2 pathway activation, highlighting the crucial role of these pathways as a barrier for cardiac reprogramming. These findings have significant implications for the development of new cardiac regenerative therapies.
Collapse
|
61
|
Kang X, Zhao Y, Van Arsdell G, Nelson SF, Touma M. Ppp1r1b-lncRNA inhibits PRC2 at myogenic regulatory genes to promote cardiac and skeletal muscle development in mouse and human. RNA (NEW YORK, N.Y.) 2020; 26:481-491. [PMID: 31953255 PMCID: PMC7075267 DOI: 10.1261/rna.073692.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/13/2020] [Indexed: 06/10/2023]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators and play important roles in cardiac development and congenital heart disease. In a previous study, we identified a novel lncRNA, Ppp1r1b, with expression highly correlated with myogenesis. However, the molecular mechanism that underlies Ppp1r1b-lncRNA function in myogenic regulation is unknown. By silencing Ppp1r1b-lncRNA, mouse C2C12 and human skeletal myoblasts failed to develop fully differentiated myotubes. Myogenic differentiation was also impaired in PPP1R1B-lncRNA deficient human-induced pluripotent stem cell-derived cardiomyocytes (hiPSCs-CMs). The expression of myogenic transcription factors, including MyoD, Myogenin, and Tbx5, as well as sarcomere proteins, was significantly suppressed in Ppp1r1b-lncRNA inhibited myoblast cells and neonatal mouse heart. Histone modification analysis revealed increased H3K27 tri-methylation at MyoD1 and Myogenin promoters in GapmeR treated C2C12 cells. Furthermore, Ppp1r1b-lncRNA was found to bind to Ezh2, and chromatin isolation by RNA purification (ChIRP) assay revealed enriched interaction of Ppp1r1b-lncRNA with Myod1 and Tbx5 promoters, suggesting that Ppp1r1b-lncRNA induces transcription of myogenic transcription factors by interacting with the polycomb repressive complex 2 (PRC2) at the chromatin interface. Correspondingly, the silencing of Ppp1r1b-lncRNA increased EZH2 binding at promoter regions of myogenic transcription factors. Therefore, our results suggest that Ppp1r1b-lncRNA promotes myogenic differentiation through competing for PRC2 binding with chromatin of myogenic master regulators during heart and skeletal muscle development.
Collapse
Affiliation(s)
- Xuedong Kang
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Yan Zhao
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Glen Van Arsdell
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Cardiothoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Stanley F Nelson
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Human Genetics, Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Marlin Touma
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Neonatal/Congenital Heart Laboratory, Cardiovascular Research Laboratory, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Human Genetics, Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Institute of Precision Health, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- The Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Children's Discovery and Innovation Institute, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Eli and Edythe Broad Stem Cell Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
62
|
Wang J, Jiang X, Zhao L, Zuo S, Chen X, Zhang L, Lin Z, Zhao X, Qin Y, Zhou X, Yu XY. Lineage reprogramming of fibroblasts into induced cardiac progenitor cells by CRISPR/Cas9-based transcriptional activators. Acta Pharm Sin B 2020; 10:313-326. [PMID: 32082976 PMCID: PMC7016296 DOI: 10.1016/j.apsb.2019.09.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022] Open
Abstract
Overexpression of exogenous lineage-determining factors succeeds in directly reprogramming fibroblasts to various cell types. Several studies have reported reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs). CRISPR/Cas9-mediated gene activation is a potential approach for cellular reprogramming due to its high precision and multiplexing capacity. Here we show lineage reprogramming to iCPCs through a dead Cas9 (dCas9)-based transcription activation system. Targeted and robust activation of endogenous cardiac factors, including GATA4, HAND2, MEF2C and TBX5 (G, H, M and T; GHMT), can reprogram human fibroblasts toward iCPCs. The iCPCs show potentials to differentiate into cardiomyocytes, smooth muscle cells and endothelial cells in vitro. Addition of MEIS1 to GHMT induces cell cycle arrest in G2/M and facilitates cardiac reprogramming. Lineage reprogramming of human fibroblasts into iCPCs provides a promising cellular resource for disease modeling, drug discovery and individualized cardiac cell therapy.
Collapse
|
63
|
Isoform Specific Effects of Mef2C during Direct Cardiac Reprogramming. Cells 2020; 9:cells9020268. [PMID: 31979018 PMCID: PMC7072587 DOI: 10.3390/cells9020268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 01/14/2023] Open
Abstract
Direct conversion of cardiac fibroblasts into induced cardiomyocytes (iCMs) by forced expression of defined factors holds great potential for regenerative medicine by offering an alternative strategy for treatment of heart disease. Successful iCM conversion can be achieved by minimally using three transcription factors, Mef2c (M), Gata4(G), and Tbx5 (T). Despite increasing interest in iCM mechanistic studies using MGT(polycistronic construct with optimal expression of M,G and T), the reprogramming efficiency varies among different laboratories. Two main Mef2c isoforms (isoform2, Mi2 and isoform4, Mi4) are present in heart and are used separately by different labs, for iCM reprogramming. It is currently unknown if differently spliced isoform of Mef2c contributes to varied reprogramming efficiency. Here, we used Mi2 and Mi4 together with Gata4 and Tbx5 in separate vectors or polycistronic vector, to convert fibroblasts to iCMs. We found that Mi2 can induce higher reprogramming efficiency than Mi4 in MEFs. Addition of Hand2 to MGT retroviral cocktail or polycistronic Mi2-GT retroviruses further enhanced the iCM conversion. Overall, this study demonstrated the isoform specific effects of Mef2c, during iCM reprogramming, clarified some discrepancy about varied efficiency among labs and might lead to future research into the role of alternative splicing and the consequent variants in cell fate determination.
Collapse
|
64
|
Blin G, Liand M, Mauduit C, Chehade H, Benahmed M, Simeoni U, Siddeek B. Maternal Exposure to High-Fat Diet Induces Long-Term Derepressive Chromatin Marks in the Heart. Nutrients 2020; 12:E181. [PMID: 31936461 PMCID: PMC7019950 DOI: 10.3390/nu12010181] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Heart diseases are a leading cause of death. While the link between early exposure to nutritional excess and heart disease risk is clear, the molecular mechanisms involved are poorly understood. In the developmental programming field, increasing evidence is pointing out the critical role of epigenetic mechanisms. Among them, polycomb repressive complex 2 (PRC2) and DNA methylation play a critical role in heart development and pathogenesis. In this context, we aimed at evaluating the role of these epigenetic marks in the long-term cardiac alterations induced by early dietary challenge. Using a model of rats exposed to maternal high-fat diet during gestation and lactation, we evaluated cardiac alterations at adulthood. Expression levels of PRC2 components, its histone marks di- and trimethylated histone H3 (H3K27me2/3), associated histone mark (ubiquitinated histone H2A, H2AK119ub1) and target genes were measured by Western blot. Global DNA methylation level and DNA methyl transferase 3B (DNMT3B) protein levels were measured. Maternal high-fat diet decreased H3K27me3, H2Ak119ub1 and DNA methylation levels, down-regulated the enhancer of zeste homolog 2 (EZH2), and DNMT3B expression. The levels of the target genes, isl lim homeobox 1 (Isl1), six homeobox 1 (Six1) and mads box transcription enhancer factor 2, polypeptide C (Mef2c), involved in cardiac pathogenesis were up regulated. Overall, our data suggest that the programming of cardiac alterations by maternal exposure to high-fat diet involves the derepression of pro-fibrotic and pro-hypertrophic genes through the induction of EZH2 and DNMT3B deficiency.
Collapse
Affiliation(s)
- Guillaume Blin
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Marjorie Liand
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Claire Mauduit
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Hassib Chehade
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Mohamed Benahmed
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Umberto Simeoni
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Benazir Siddeek
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| |
Collapse
|
65
|
Abstract
Cardiovascular disease is the leading cause of death worldwide. Cardiomyocytes have limited regenerative capacity; consequently, regenerative therapies are in high demand. There are currently several potential strategies for heart regeneration, with one approach involving in situ generation of new cardiomyocytes from endogenous cell sources. Direct cardiac reprogramming has emerged as a novel therapeutic approach to regenerating the damaged heart by directly converting endogenous cardiac fibroblasts into cardiomyocyte-like cells. Following our first report of direct cardiac reprogramming, significant advances have elucidated the molecular mechanisms associated with cardiac reprogramming. These advances have also improved cardiac-reprogramming efficiency by enabling direct in vivo cardiac reprogramming. Moreover, progress has been made in cardiac reprogramming of human fibroblasts. Although basic research has supported substantial progress in this field, numerous challenges remain in terms of clinical application. Here, we review the current state of cardiac reprogramming as a new technology for understanding and treating cardiovascular diseases.
Collapse
|
66
|
Spotlight on epigenetic reprogramming in cardiac regeneration. Semin Cell Dev Biol 2020; 97:26-37. [PMID: 31002867 DOI: 10.1016/j.semcdb.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
67
|
Dal-Pra S, Hodgkinson CP, Dzau VJ. Induced cardiomyocyte maturation: Cardiac transcription factors are necessary but not sufficient. PLoS One 2019; 14:e0223842. [PMID: 31622977 PMCID: PMC6797484 DOI: 10.1371/journal.pone.0223842] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/30/2019] [Indexed: 01/06/2023] Open
Abstract
The process by which fibroblasts are directly reprogrammed into cardiomyocytes involves two stages; initiation and maturation. Initiation represents the initial expression of factors that induce fibroblasts to transdifferentiate into cardiomyocytes. Following initiation, the cell undergoes a period of maturation before becoming a mature cardiomyocyte. We wanted to understand the role of cardiac development transcription factors in the maturation process. We directly reprogram fibroblasts into cardiomyocytes by a combination of miRNAs (miR combo). The ability of miR combo to induce cardiomyocyte-specific genes in fibroblasts was lost following the knockdown of the cardiac transcription factors Gata4, Mef2C, Tbx5 and Hand2 (GMTH). To further clarify the role of GMTH in miR combo reprogramming we utilized a modified CRISPR-Cas9 approach to activate endogenous GMTH genes. Importantly, both miR combo and the modified CRISPR-Cas9 approach induced comparable levels of GMTH expression. While miR combo was able to reprogram fibroblasts into cardiomyocyte-like cells, the modified CRISPR-Cas9 approach could not. Indeed, we found that cardiomyocyte maturation only occurred with very high levels of GMT expression. Taken together, our data indicates that while endogenous cardiac transcription factors are insufficient to reprogram fibroblasts into mature cardiomyocytes, endogenous cardiac transcription factors are necessary for expression of maturation genes.
Collapse
Affiliation(s)
- Sophie Dal-Pra
- Mandel Center for Hypertension and Atherosclerosis Research, Duke University, Durham, North Carolina, United States of America
- Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Conrad P. Hodgkinson
- Mandel Center for Hypertension and Atherosclerosis Research, Duke University, Durham, North Carolina, United States of America
- Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Victor J. Dzau
- Mandel Center for Hypertension and Atherosclerosis Research, Duke University, Durham, North Carolina, United States of America
- Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
68
|
Abstract
Supplemental Digital Content is available in the text. If unifying principles could be revealed for how the same genome encodes different eukaryotic cells and for how genetic variability and environmental input are integrated to impact cardiovascular health, grand challenges in basic cell biology and translational medicine may succumb to experimental dissection. A rich body of work in model systems has implicated chromatin-modifying enzymes, DNA methylation, noncoding RNAs, and other transcriptome-shaping factors in adult health and in the development, progression, and mitigation of cardiovascular disease. Meanwhile, deployment of epigenomic tools, powered by next-generation sequencing technologies in cardiovascular models and human populations, has enabled description of epigenomic landscapes underpinning cellular function in the cardiovascular system. This essay aims to unpack the conceptual framework in which epigenomes are studied and to stimulate discussion on how principles of chromatin function may inform investigations of cardiovascular disease and the development of new therapies.
Collapse
Affiliation(s)
- Manuel Rosa-Garrido
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Douglas J Chapski
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles
| | - Thomas M Vondriska
- From the Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California, Los Angeles.
| |
Collapse
|
69
|
Epigenetics and Mechanobiology in Heart Development and Congenital Heart Disease. Diseases 2019; 7:diseases7030052. [PMID: 31480510 PMCID: PMC6787645 DOI: 10.3390/diseases7030052] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
: Congenital heart disease (CHD) is the most common birth defect worldwide and the number one killer of live-born infants in the United States. Heart development occurs early in embryogenesis and involves complex interactions between multiple cell populations, limiting the understanding and consequent treatment of CHD. Furthermore, genome sequencing has largely failed to predict or yield therapeutics for CHD. In addition to the underlying genome, epigenetics and mechanobiology both drive heart development. A growing body of evidence implicates the aberrant regulation of these two extra-genomic systems in the pathogenesis of CHD. In this review, we describe the stages of human heart development and the heart defects known to manifest at each stage. Next, we discuss the distinct and overlapping roles of epigenetics and mechanobiology in normal development and in the pathogenesis of CHD. Finally, we highlight recent advances in the identification of novel epigenetic biomarkers and environmental risk factors that may be useful for improved diagnosis and further elucidation of CHD etiology.
Collapse
|
70
|
Ameliorating the Fibrotic Remodeling of the Heart through Direct Cardiac Reprogramming. Cells 2019; 8:cells8070679. [PMID: 31277520 PMCID: PMC6679082 DOI: 10.3390/cells8070679] [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: 05/05/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 12/20/2022] Open
Abstract
Coronary artery disease is the most common form of cardiovascular diseases, resulting in the loss of cardiomyocytes (CM) at the site of ischemic injury. To compensate for the loss of CMs, cardiac fibroblasts quickly respond to injury and initiate cardiac remodeling in an injured heart. In the remodeling process, cardiac fibroblasts proliferate and differentiate into myofibroblasts, which secrete extracellular matrix to support the intact structure of the heart, and eventually differentiate into matrifibrocytes to form chronic scar tissue. Discovery of direct cardiac reprogramming offers a promising therapeutic strategy to prevent/attenuate this pathologic remodeling and replace the cardiac fibrotic scar with myocardium in situ. Since the first discovery in 2010, many progresses have been made to improve the efficiency and efficacy of reprogramming by understanding the mechanisms and signaling pathways that are activated during direct cardiac reprogramming. Here, we overview the development and recent progresses of direct cardiac reprogramming and discuss future directions in order to translate this promising technology into an effective therapeutic paradigm to reverse cardiac pathological remodeling in an injured heart.
Collapse
|
71
|
Yu JSL, Palano G, Lim C, Moggio A, Drowley L, Plowright AT, Bohlooly‐Y M, Rosen BS, Hansson EM, Wang Q, Yusa K. CRISPR-Knockout Screen Identifies Dmap1 as a Regulator of Chemically Induced Reprogramming and Differentiation of Cardiac Progenitors. Stem Cells 2019; 37:958-972. [PMID: 30932271 PMCID: PMC6767549 DOI: 10.1002/stem.3012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/11/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Direct in vivo reprogramming of cardiac fibroblasts into myocytes is an attractive therapeutic intervention in resolving myogenic deterioration. Current transgene-dependent approaches can restore cardiac function, but dependence on retroviral delivery and persistent retention of transgenic sequences are significant therapeutic hurdles. Chemical reprogramming has been established as a legitimate method to generate functional cell types, including those of the cardiac lineage. Here, we have extended this approach to generate progenitor cells that can differentiate into endothelial cells and cardiomyocytes using a single inhibitor protocol. Depletion of terminally differentiated cells and enrichment for proliferative cells result in a second expandable progenitor population that can robustly give rise to myofibroblasts and smooth muscle. Deployment of a genome-wide knockout screen with clustered regularly interspaced short palindromic repeats-guide RNA library to identify novel mediators that regulate the reprogramming revealed the involvement of DNA methyltransferase 1-associated protein 1 (Dmap1). Loss of Dmap1 reduced promoter methylation, increased the expression of Nkx2-5, and enhanced the retention of self-renewal, although further differentiation is inhibited because of the sustained expression of Cdh1. Our results hence establish Dmap1 as a modulator of cardiac reprogramming and myocytic induction. Stem Cells 2019;37:958-972.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- CRISPR-Cas Systems
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cellular Reprogramming/drug effects
- Cellular Reprogramming/genetics
- Dioxoles/pharmacology
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Editing/methods
- Homeobox Protein Nkx-2.5/genetics
- Homeobox Protein Nkx-2.5/metabolism
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Primary Cell Culture
- Pyrazoles/pharmacology
- Pyridines/pharmacology
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Jason S. L. Yu
- Stem Cell Genetics, Wellcome Sanger InstituteHinxton, CambridgeUnited Kingdom
- Department of Cell BiologyThe Francis Crick InstituteLondonUnited Kingdom
| | - Giorgia Palano
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Cindy Lim
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Aldo Moggio
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Lauren Drowley
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Alleyn T. Plowright
- Medicinal Chemistry, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | | | - Barry S. Rosen
- Discovery Sciences, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Emil M. Hansson
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of MedicineKarolinska InstitutetHuddingeSweden
| | - Qing‐Dong Wang
- Bioscience Heart Failure, Cardiovascular, Renal and Metabolism, IMED Biotech UnitAstraZenecaGothenburgSweden
| | - Kosuke Yusa
- Stem Cell Genetics, Wellcome Sanger InstituteHinxton, CambridgeUnited Kingdom
- Stem Cell GeneticsInstitute for Frontier Life and Medical Sciences, Kyoto UniversityKyotoJapan
| |
Collapse
|
72
|
Sadahiro T. Cardiac regeneration with pluripotent stem cell-derived cardiomyocytes and direct cardiac reprogramming. Regen Ther 2019; 11:95-100. [PMID: 31304202 PMCID: PMC6606831 DOI: 10.1016/j.reth.2019.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/20/2019] [Accepted: 06/13/2019] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease is the leading cause of death globally. Cardiomyocytes (CMs) have poor regenerative capacity, and pharmacological therapies have limited efficacy in severe heart failure. Currently, there are several promising strategies for cardiac regeneration. The most promising approach to remuscularize failing hearts is cell transplantation therapy using newly generated CMs from exogenous sources, such as pluripotent stem cells. Alternatively, approaches to generate new CMs from endogenous cell sources in situ may also repair the injured heart and improve cardiac function. Direct cardiac reprogramming has emerged as a novel therapeutic approach to regenerate injured hearts by directly converting endogenous cardiac fibroblasts into CM-like cells. Through cell transplantation and direct cardiac reprogramming, new CMs can be generated and scar tissue reduced to improve cardiac function; therefore, cardiac regeneration may serve as a powerful strategy for treatment of severe heart failure. While substantial progress has been made in these two strategies for cardiac regeneration over the past several years, challenges remain for clinical translation. This review provide an overview of previous reports and current challenges in this field.
Collapse
Key Words
- BMP, bone morphogenic protein
- CFs, cardiac fibroblasts
- CMs, cardiomyocytes
- CPCs, cardiac progenitor cells
- Cardiomyocytes
- Direct reprogramming
- ESCs, embryonic stem cells
- Fibroblasts
- GHMT, GMT plus Hand2
- GMT, Gata4
- MI, myocardial infarction
- Mef2c, and Tbx5
- PSCs, pluripotent stem cells
- Pluripotent stem cells
- Regeneration
- SeV-GMT, Sendai virus vector expressing GMT
- iCMs, induced cardiomyocyte-like cells
- iPSCs, induced pluripotent stem cells
- miRs, microRNAs
Collapse
Affiliation(s)
- Taketaro Sadahiro
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennoudai, Tsukuba City, Ibaraki, 305-8575, Japan
| |
Collapse
|
73
|
Sauls K, Greco TM, Wang L, Zou M, Villasmil M, Qian L, Cristea IM, Conlon FL. Initiating Events in Direct Cardiomyocyte Reprogramming. Cell Rep 2019; 22:1913-1922. [PMID: 29444441 DOI: 10.1016/j.celrep.2018.01.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 11/30/2017] [Accepted: 01/17/2018] [Indexed: 01/14/2023] Open
Abstract
Direct reprogramming of fibroblasts into cardiomyocyte-like cells (iCM) holds great potential for heart regeneration and disease modeling and may lead to future therapeutic applications. Currently, application of this technology is limited by our lack of understanding of the molecular mechanisms that drive direct iCM reprogramming. Using a quantitative mass spectrometry-based proteomic approach, we identified the temporal global changes in protein abundance that occur during initial phases of iCM reprogramming. Collectively, our results show systematic and temporally distinct alterations in levels of specific functional classes of proteins during the initiating steps of reprogramming including extracellular matrix proteins, translation factors, and chromatin-binding proteins. We have constructed protein relational networks associated with the initial transition of a fibroblast into an iCM. These findings demonstrate the presence of an orchestrated series of temporal steps associated with dynamic changes in protein abundance in a defined group of protein pathways during the initiating events of direct reprogramming.
Collapse
Affiliation(s)
- Kimberly Sauls
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Li Wang
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Pathology and Laboratory Medicine, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Meng Zou
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA
| | - Michelle Villasmil
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA; Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Qian
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Pathology and Laboratory Medicine, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Frank L Conlon
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA; Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Integrative Program for Biological and Genome Sciences, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
74
|
Keepers B, Liu J, Qian L. What's in a cardiomyocyte - And how do we make one through reprogramming? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118464. [PMID: 30922868 DOI: 10.1016/j.bbamcr.2019.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/10/2019] [Accepted: 03/21/2019] [Indexed: 12/19/2022]
Abstract
Substantial progress is being made in the field cardiac reprogramming, and those in the field are hopeful that the technology will be formulated for therapeutic use. Beyond the excitement around generating a revolutionary new approach for treating ischemic heart diseases, cardiac reprogramming has delivered provocative findings that challenge common notions of cell fate and cell identity. Have we really made de novo cardiomyocytes? To answer this question, the essential characteristics of this unique and important cell type must first be defined. In this review, we walk through the history of scientific inquiry into cardiomyocytes, and then we examine the core features of cardiomyocytes as detailed in modern definitions. Informed by this, we turn to cardiac reprogramming to analyze the various screening approaches and ultimate factor combinations used in each study. We follow this with a dissection of the evidence used to support the authors' claims of successfully creating cardiomyocytes, and we end by discussing what is known about the molecular mechanisms of cardiac reprogramming. Through this analysis, we find interesting differences between the study designs and their results, but it becomes clear that the field at large is generating cells that closely match the textbook definition cardiomyocyte. However, the differences noted between the results of each study are largely unexplained, reflecting the need for further research in both cardiac reprogramming and in native cardiomyocyte biology. Knowledge gained from future research will help move the field towards better reprogramming techniques and technologies.
Collapse
Affiliation(s)
- Benjamin Keepers
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- McAllister Heart Institute, Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
75
|
Epigenetics, cardiovascular disease, and cellular reprogramming. J Mol Cell Cardiol 2019; 128:129-133. [PMID: 30690032 DOI: 10.1016/j.yjmcc.2019.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/22/2019] [Indexed: 12/29/2022]
Abstract
Under the seeming disorder of "junk" sequences the last decade has seen developments in our understanding of non-coding RNA's (ncRNAs). It's a complex revised order and nowhere is this more relevant than in the developing heart whereby old rules have been set aside to make room for new ones. The development of the mammalian heart has been studied at the genetic and cellular level for several decades because these areas were considered ideal control points. As such, detailed mechanisms governing cell lineages are well described. Emerging evidence suggests a complex new order regulated by epigenetic mechanisms mark cardiac cell lineage. Indeed, molecular cardiologists are in the process of shedding light on the roles played by ncRNAs, nucleic acid methylation and histone/chromatin modifications in specific pathologies of the heart. The aim of this article is to discuss some of the recent advances in the field of cardiovascular epigenetics that are related to direct cell reprogramming and repair. As such, we explore ncRNAs as nodes regulating signaling networks and attempt to make sense of regulatory disorder by reinforcing the importance of epigenetic components in the developmental program.
Collapse
|
76
|
Chang Y, Lee E, Kim J, Kwon YW, Kwon Y, Kim J. Efficient in vivo direct conversion of fibroblasts into cardiomyocytes using a nanoparticle-based gene carrier. Biomaterials 2018; 192:500-509. [PMID: 30513475 DOI: 10.1016/j.biomaterials.2018.11.034] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/16/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
The reprogramming of induced cardiomyocytes (iCMs) has shown potential in regenerative medicine. However, in vivo reprogramming of iCMs is significantly inefficient, and novel gene delivery systems are required to more efficiently and safely induce in vivo reprogramming of iCMs for therapeutic applications in heart injury. In this study, we show that cationic gold nanoparticles (AuNPs) loaded with Gata4, Mef2c, and Tbx5 function as nanocarriers for cardiac reprogramming. The AuNP/GMT/PEI nanocomplexes show high reprogramming efficiency in human and mouse somatic cells with low cytotoxicity and direct conversion into iCMs without integrating factors into the genome. Importantly, AuNP/GMT/PEI nanocomplexes led to efficient in vivo conversion into cardiomyocytes after myocardial infarction (MI), resulting in the effective recovery of cardiac function and scar area. Taken together, these results show that the AuNP/GMT/PEI nanocarrier can be used to develop effective therapeutics for heart regeneration in cardiac disease patients.
Collapse
Affiliation(s)
- Yujung Chang
- Department of Biomedical Engineering (BK21plus), Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Euiyeon Lee
- Department of Biomedical Engineering (BK21plus), Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Junyeop Kim
- Department of Biomedical Engineering (BK21plus), Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Yoo-Wook Kwon
- Biomedical Research Institute, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Youngeun Kwon
- Department of Biomedical Engineering (BK21plus), Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea.
| | - Jongpil Kim
- Department of Biomedical Engineering (BK21plus), Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea; Department of Chemistry, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul, 04620, Republic of Korea.
| |
Collapse
|
77
|
Klose K, Gossen M, Stamm C. Turning fibroblasts into cardiomyocytes: technological review of cardiac transdifferentiation strategies. FASEB J 2018; 33:49-70. [DOI: 10.1096/fj.201800712r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Kristin Klose
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Berlin Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT) Berlin Germany
- Charité–Universitätsmedizin Berlin Berlin Germany
| | - Manfred Gossen
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Berlin Germany
- Helmholtz‐Zentrum Geesthacht (HZG)Institute of Biomaterial Science Teltow Germany
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Berlin Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT) Berlin Germany
- Charité–Universitätsmedizin Berlin Berlin Germany
- German Centre for Cardiovascular Research (DZHK)Partner Site Berlin Berlin Germany
- Department of Cardiothoracic and Vascular SurgeryDeutsches Herzzentrum Berlin (DHZB) Berlin Germany
| |
Collapse
|
78
|
Direct Cardiac Reprogramming: A Novel Approach for Heart Regeneration. Int J Mol Sci 2018; 19:ijms19092629. [PMID: 30189626 PMCID: PMC6165160 DOI: 10.3390/ijms19092629] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/01/2018] [Accepted: 09/01/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiac diseases are among the most common causes of death globally. Cardiac muscle has limited proliferative capacity, and regenerative therapies are highly in demand as a new treatment strategy. Although pluripotent reprogramming has been developed, it has obstacles, such as a potential risk of tumor formation, poor survival of the transplanted cells, and high cost. We previously reported that fibroblasts can be directly reprogrammed to cardiomyocytes by overexpressing a combination of three cardiac-specific transcription factors (Gata4, Mef2c, Tbx5 (together, GMT)). We and other groups have promoted cardiac reprogramming by the addition of certain miRNAs, cytokines, and epigenetic factors, and unraveled new molecular mechanisms of cardiac reprogramming. More recently, we discovered that Sendai virus (SeV) vector expressing GMT could efficiently and rapidly reprogram fibroblasts into integration-free cardiomyocytes in vitro via robust transgene expression. Gene delivery of SeV-GMT also improves cardiac function and reduces fibrosis after myocardial infarction in mice. Through direct cardiac reprogramming, new cardiomyocytes can be generated and scar tissue reduced to restore cardiac function, and, thus, direct cardiac reprogramming may serve as a powerful strategy for cardiac regeneration. Here, we provide an overview of the previous reports and current challenges in this field.
Collapse
|
79
|
Zhou Y, Wang L, Liu Z, Alimohamadi S, Yin C, Liu J, Qian L. Comparative Gene Expression Analyses Reveal Distinct Molecular Signatures between Differentially Reprogrammed Cardiomyocytes. Cell Rep 2018; 20:3014-3024. [PMID: 28954220 DOI: 10.1016/j.celrep.2017.09.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 06/20/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
Cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) or directly reprogrammed from non-myocytes (induced cardiomyocytes [iCMs]) are promising sources for heart regeneration or disease modeling. However, the similarities and differences between iPSC-CMs and iCMs are still unknown. Here, we performed transcriptome analyses of beating iPSC-CMs and iCMs generated from cardiac fibroblasts (CFs) of the same origin. Although both iPSC-CMs and iCMs establish CM-like molecular features globally, iPSC-CMs exhibit a relatively hyperdynamic epigenetic status, whereas iCMs exhibit a maturation status that more closely resembles that of adult CMs. Based on gene expression of metabolic enzymes, iPSC-CMs primarily employ glycolysis, whereas iCMs utilize fatty acid oxidation as the main pathway. Importantly, iPSC-CMs and iCMs exhibit different cell-cycle status, alteration of which influenced their maturation. Therefore, our study provides a foundation for understanding the pros and cons of different reprogramming approaches.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sahar Alimohamadi
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chaoying Yin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
80
|
Direct Reprograming to Regenerate Myocardium and Repair Its Pacemaker and Conduction System. MEDICINES 2018; 5:medicines5020048. [PMID: 29867004 PMCID: PMC6023490 DOI: 10.3390/medicines5020048] [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: 05/07/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 01/14/2023]
Abstract
The regenerative medicine field has been revolutionized by the direct conversion of one cell type to another by ectopic expression of lineage-specific transcription factors. The direct reprogramming of fibroblasts to induced cardiac myocytes (iCMs) by core cardiac transcription factors (Gata4, Mef2c, Tbx5) both in vitro and in vivo has paved the way in cardiac regeneration and repair. Several independent research groups have successfully reported the direct reprogramming of fibroblasts in injured myocardium to cardiac myocytes employing a variety of approaches that rely on transcription factors, small molecules, and micro RNAs (miRNAs). Recently, this technology has been considered for local repair of the pacemaker and the cardiac conduction system. To address this, we will first discuss the direct reprograming advancements in the setting of working myocardium regeneration, and then elaborate on how this technology can be applied to repair the cardiac pacemaker and the conduction system.
Collapse
|
81
|
Hodgkinson CP, Pratt RE, Kirste I, Dal-Pra S, Cooke JP, Dzau VJ. Cardiomyocyte Maturation Requires TLR3 Activated Nuclear Factor Kappa B. Stem Cells 2018; 36:1198-1209. [PMID: 29676038 DOI: 10.1002/stem.2833] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/15/2018] [Accepted: 03/30/2018] [Indexed: 12/18/2022]
Abstract
The process by which committed precursors mature into cardiomyocytes is poorly understood. We found that TLR3 inhibition blocked cardiomyocyte maturation; precursor cells committed to the cardiomyocyte lineage failed to express maturation genes and sarcomeres did not develop. Using various approaches, we found that the effects of TLR3 upon cardiomyocyte maturation were dependent upon the RelA subunit of nuclear factor kappa B (NFκB). Importantly, under conditions that promote the development of mature cardiomyocytes NFκB became significantly enriched at the promoters of cardiomyocyte maturation genes. Furthermore, activation of the TLR3-NFκB pathway enhanced cardiomyocyte maturation. This study, therefore, demonstrates that the TLR3-NFκB pathway is necessary for the maturation of committed precursors into mature cardiomyocytes. Stem Cells 2018;36:1198-1209.
Collapse
Affiliation(s)
- Conrad P Hodgkinson
- Department of Medicine, Division of Cardiology, Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North California, USA
| | - Richard E Pratt
- Department of Medicine, Division of Cardiology, Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North California, USA
| | - Imke Kirste
- Department of Medicine, Division of Cardiology, Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North California, USA
| | - Sophie Dal-Pra
- Department of Medicine, Division of Cardiology, Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North California, USA
| | - John P Cooke
- Houston Methodist Research Institute, Department of Cardiovascular Sciences, Houston, Texas, USA
| | - Victor J Dzau
- Department of Medicine, Division of Cardiology, Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North California, USA
| |
Collapse
|
82
|
A Loss of Function Screen of Epigenetic Modifiers and Splicing Factors during Early Stage of Cardiac Reprogramming. Stem Cells Int 2018; 2018:3814747. [PMID: 29743891 PMCID: PMC5878887 DOI: 10.1155/2018/3814747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
Direct reprogramming of cardiac fibroblasts (CFs) to induced cardiomyocytes (iCMs) is a newly emerged promising approach for cardiac regeneration, disease modeling, and drug discovery. However, its potential has been drastically limited due to the low reprogramming efficiency and largely unknown underlying molecular mechanisms. We have previously screened and identified epigenetic factors related to histone modification during iCM reprogramming. Here, we used shRNAs targeting an additional battery of epigenetic factors involved in chromatin remodeling and RNA splicing factors to further identify inhibitors and facilitators of direct cardiac reprogramming. Knockdown of RNA splicing factors Sf3a1 or Sf3b1 significantly reduced the percentage and total number of cardiac marker positive iCMs accompanied with generally repressed gene expression. Removal of another RNA splicing factor Zrsr2 promoted the acquisition of CM molecular features in CFs and mouse embryonic fibroblasts (MEFs) at both protein and mRNA levels. Moreover, a consistent increase of reprogramming efficiency was observed in CFs and MEFs treated with shRNAs targeting Bcor (component of BCOR complex superfamily) or Stag2 (component of cohesin complex). Our work thus reveals several additional epigenetic and splicing factors that are either inhibitory to or required for iCM reprogramming and highlights the importance of epigenetic regulation and RNA splicing process during cell fate conversion.
Collapse
|
83
|
Direct Cardiac Reprogramming: Progress and Promise. Stem Cells Int 2018; 2018:1435746. [PMID: 29731772 PMCID: PMC5872587 DOI: 10.1155/2018/1435746] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/15/2017] [Accepted: 01/09/2018] [Indexed: 01/14/2023] Open
Abstract
The human adult heart lacks a robust endogenous repair mechanism to fully restore cardiac function after insult; thus, the ability to regenerate and repair the injured myocardium remains a top priority in treating heart failure. The ability to efficiently generate a large number of functioning cardiomyocytes capable of functional integration within the injured heart has been difficult. However, the ability to directly convert fibroblasts into cardiomyocyte-like cells both in vitro and in vivo offers great promise in overcoming this problem. In this review, we describe the insights and progress that have been gained from the investigation of direct cardiac reprogramming. We focus on the use of key transcription factors and cardiogenic genes as well as on the use of other biological molecules such as small molecules, cytokines, noncoding RNAs, and epigenetic modifiers to improve the efficiency of cardiac reprogramming. Finally, we discuss the development of safer reprogramming approaches for future clinical application.
Collapse
|
84
|
Nanotechnology-Based Cardiac Targeting and Direct Cardiac Reprogramming: The Betrothed. Stem Cells Int 2017; 2017:4940397. [PMID: 29375623 PMCID: PMC5742458 DOI: 10.1155/2017/4940397] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/18/2017] [Accepted: 10/18/2017] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases represent the first cause of morbidity in Western countries, and chronic heart failure features a significant health care burden in developed countries. Efforts in the attempt of finding new possible strategies for the treatment of CHF yielded several approaches based on the use of stem cells. The discovery of direct cardiac reprogramming has unveiled a new approach to heart regeneration, allowing, at least in principle, the conversion of one differentiated cell type into another without proceeding through a pluripotent intermediate. First developed for cancer treatment, nanotechnology-based approaches have opened new perspectives in many fields of medical research, including cardiovascular research. Nanotechnology could allow the delivery of molecules with specific biological activity at a sustained and controlled rate in heart tissue, in a cell-specific manner. Potentially, all the mediators and structural molecules involved in the fibrotic process could be selectively targeted by nanocarriers, but to date, only few experiences have been made in cardiac research. This review highlights the most prominent concepts that characterize both the field of cardiac reprogramming and a nanomedicine-based approach to cardiovascular diseases, hypothesizing a possible synergy between these two very promising fields of research in the treatment of heart failure.
Collapse
|
85
|
Abstract
Somatic cells can be reprogrammed to pluripotent stem cells or transdifferentiate to another lineage cell type. Much efforts have been made to unravel the epigenetic mechanisms underlying the cell fate conversion. Histone modifications as the major epigenetic regulator are implicated in various aspects of reprogramming and transdifferentiation. Here, we discuss the roles of histone modifications on reprogramming and transdifferentiation and hopefully provide new insights into induction and promotion of the cell fate conversion by modulating histone modifications.
Collapse
|
86
|
Liu Z, Wang L, Welch JD, Ma H, Zhou Y, Vaseghi HR, Yu S, Wall JB, Alimohamadi S, Zheng M, Yin C, Shen W, Prins JF, Liu J, Qian L. Single-cell transcriptomics reconstructs fate conversion from fibroblast to cardiomyocyte. Nature 2017; 551:100-104. [PMID: 29072293 PMCID: PMC5954984 DOI: 10.1038/nature24454] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/22/2017] [Indexed: 12/18/2022]
Abstract
Direct lineage conversion offers a new strategy for tissue regeneration and disease modelling. Despite recent success in directly reprogramming fibroblasts into various cell types, the precise changes that occur as fibroblasts progressively convert to the target cell fates remain unclear. The inherent heterogeneity and asynchronous nature of the reprogramming process renders it difficult to study this process using bulk genomic techniques. Here we used single-cell RNA sequencing to overcome this limitation and analysed global transcriptome changes at early stages during the reprogramming of mouse fibroblasts into induced cardiomyocytes (iCMs). Using unsupervised dimensionality reduction and clustering algorithms, we identified molecularly distinct subpopulations of cells during reprogramming. We also constructed routes of iCM formation, and delineated the relationship between cell proliferation and iCM induction. Further analysis of global gene expression changes during reprogramming revealed unexpected downregulation of factors involved in mRNA processing and splicing. Detailed functional analysis of the top candidate splicing factor, Ptbp1, revealed that it is a critical barrier for the acquisition of cardiomyocyte-specific splicing patterns in fibroblasts. Concomitantly, Ptbp1 depletion promoted cardiac transcriptome acquisition and increased iCM reprogramming efficiency. Additional quantitative analysis of our dataset revealed a strong correlation between the expression of each reprogramming factor and the progress of individual cells through the reprogramming process, and led to the discovery of new surface markers for the enrichment of iCMs. In summary, our single-cell transcriptomics approaches enabled us to reconstruct the reprogramming trajectory and to uncover intermediate cell populations, gene pathways and regulators involved in iCM induction.
Collapse
Affiliation(s)
- Ziqing Liu
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Li Wang
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Joshua D. Welch
- Department of Computer Science, University of North Carolina at Chapel Hill
| | - Hong Ma
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Yang Zhou
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Haley Ruth Vaseghi
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Shuo Yu
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Joseph Blake Wall
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Sahar Alimohamadi
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Michael Zheng
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Chaoying Yin
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Weining Shen
- Department of Statistics, University of California at Irvine
| | - Jan F. Prins
- Department of Computer Science, University of North Carolina at Chapel Hill
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
87
|
Liu L, Lei I, Wang Z. Improving cardiac reprogramming for heart regeneration. Curr Opin Organ Transplant 2017; 21:588-594. [PMID: 27755167 DOI: 10.1097/mot.0000000000000363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease is the leading cause of death in the world today, and the death rate has remained virtually unchanged in the last 20 years (American Heart Association). This severe life-threatening disease underscores a critical need for developing novel therapeutic strategies to effectively treat this devastating disease. Cell-based therapy represents an extremely promising approach. Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers an attractive novel strategy for in-situ heart regeneration. Major challenges of iCM reprogramming include the low conversion rate and heterogeneity of the iCMs. This review will summarize the major advancements in improving the iCM reprogramming efficiency and iCM maturation. RECENT FINDINGS Numerous studies have been published in the past 18 months to describe various strategies for achieving more efficient iCM reprogramming. These strategies are based on our understanding of the molecular mechanisms of cardiogenesis, which include transcriptional networks, signaling pathways and epigenetic cell fate change. SUMMARY Novel strategies for highly efficient iCM reprogramming will be required for applying iCM reprogramming to patients. Creative and combined methods based on our understanding of cardiogenesis will continue to contribute heavily in the advancement of iCM reprogramming. We are highly optimistic that iCM reprogramming-based heart therapy will restore the pumping function of damaged patient hearts.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
88
|
Abstract
Decades of studies have shown that epigenetic alterations play a significant role on cancer development both in vitro and in vivo. However, considering that many cancers harbor mutations at epigenetic modifier genes and that transcription factor-mediated gene regulations are tightly coupled with epigenetic modifications, the majority of epigenetic alterations in cancers could be the consequence of the dysfunction or dysregulation of epigenetic modifiers caused by genetic abnormalities. Therefore, it remains unclear whether bona fide epigenetic abnormalities have causal roles on cancer development. Reprogramming technologies enable us to actively alter epigenetic regulations while preserving genomic information. Taking advantage, recent studies have provided in vivo evidence for the significant impact of epigenetic abnormalities on the initiation, maintenance and progression of cancer cells.
Collapse
Affiliation(s)
- Kenji Ito
- Center for iPS Cell Research & Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yasuhiro Yamada
- Center for iPS Cell Research & Application, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
89
|
Kojima H, Ieda M. Discovery and progress of direct cardiac reprogramming. Cell Mol Life Sci 2017; 74:2203-2215. [PMID: 28197667 PMCID: PMC11107684 DOI: 10.1007/s00018-017-2466-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/27/2016] [Accepted: 01/16/2017] [Indexed: 12/17/2022]
Abstract
Cardiac disease remains a major cause of death worldwide. Direct cardiac reprogramming has emerged as a promising approach for cardiac regenerative therapy. After the discovery of MyoD, a master regulator for skeletal muscle, other single cardiac reprogramming factors (master regulators) have been sought. Discovery of cardiac reprogramming factors was inspired by the finding that multiple, but not single, transcription factors were needed to generate induced pluripotent stem cells (iPSCs) from fibroblasts. We first reported a combination of cardiac-specific transcription factors, Gata4, Mef2c, and Tbx5 (GMT), that could convert mouse fibroblasts into cardiomyocyte-like cells, which were designated as induced cardiomyocyte-like cells (iCMs). Following our first report of cardiac reprogramming, many researchers, including ourselves, demonstrated an improvement in cardiac reprogramming efficiency, in vivo direct cardiac reprogramming for heart regeneration, and cardiac reprogramming in human cells. However, cardiac reprogramming in human cells and adult fibroblasts remains inefficient, and further efforts are needed. We believe that future research elucidating epigenetic barriers and molecular mechanisms of direct cardiac reprogramming will improve the reprogramming efficiency, and that this new technology has great potential for clinical applications.
Collapse
Affiliation(s)
- Hidenori Kojima
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
- AMED-PRIME, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
90
|
Talkhabi M, Zonooz ER, Baharvand H. Boosters and barriers for direct cardiac reprogramming. Life Sci 2017; 178:70-86. [PMID: 28427897 DOI: 10.1016/j.lfs.2017.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/08/2017] [Accepted: 04/16/2017] [Indexed: 12/16/2022]
Abstract
Heart disease is currently the most significant cause of morbidity and mortality worldwide, which accounts for approximately 33% of all deaths. Recently, a promising and alchemy-like strategy has been developed called direct cardiac reprogramming, which directly converts somatic cells such as fibroblasts to cardiac lineage cells such as cardiomyocytes (CMs), termed induced CMs or iCMs. The first in vitro cardiac reprogramming study, mediated by cardiac transcription factors (TFs)-Gata4, Tbx5 and Mef2C-, was not enough efficient to produce an adequate number of fully reprogrammed, functional iCMs. As a result, numerous combinations of cardiac TFs exist for direct cardiac reprogramming of mouse and human fibroblasts. However, the efficiency of direct cardiac reprogramming remains low. Recently, a number of cellular and molecular mechanisms have been identified to increase the efficiency of direct cardiac reprogramming and the quality of iCMs. For example, microgrooved substrate, cardiogenic growth factors [VEGF, FGF, BMP4 and Activin A], and an appropriate stoichiometry of TFs boost the direct cardiac reprogramming. On the other hand, serum, TGFβ signaling, activators of epithelial to mesenchymal transition, and some epigenetic factors (Bmi1 and Ezh2) are barriers for direct cardiac reprogramming. Manipulating these mechanisms by the application of boosters and removing barriers can increase the efficiency of direct cardiac reprogramming and possibly make iCMs reliable for cell-based therapy or other potential applications. In this review, we summarize the latest trends in cardiac TF- or miRNA-based direct cardiac reprogramming and comprehensively discuses all molecular and cellular boosters and barriers affecting direct cardiac reprogramming.
Collapse
Affiliation(s)
- Mahmood Talkhabi
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Elmira Rezaei Zonooz
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
91
|
Liu Z, Chen O, Wall JBJ, Zheng M, Zhou Y, Wang L, Vaseghi HR, Qian L, Liu J. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci Rep 2017; 7:2193. [PMID: 28526819 PMCID: PMC5438344 DOI: 10.1038/s41598-017-02460-2] [Citation(s) in RCA: 438] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/12/2017] [Indexed: 11/09/2022] Open
Abstract
Cloning of multiple genes in a single vector has greatly facilitated both basic and translational studies that require co-expression of multiple factors or multi-units of complex protein. Many strategies have been adopted, among which 2A “self-cleaving” peptides have garnered increased interest for their polycistronic nature, small size and high “cleavage” efficiency. However, broad application of 2 A peptides is limited by the lack of systematic comparison of different 2As alone or in combination. Here we characterized the effect of varying gene position and 2As on the expression of proteins encoded in bi-, tri-, or quad-cistronic constructs. Using direct cardiac reprogramming as an example, we further determined the effect of varied 2As on the efficiency of fluorescent cell labeling and cell fate conversion. We found that the expression of fluorophores decreased as it was moved towards the end of the construct while reprogramming was most efficient with the fluorophore at the second position. Moreover, quad-cistronic TPE2A constructs resulted in more efficient reprogramming than 3P2A or PTE2A constructs. We expect that the bi-, tri-, and quad-cistronic vectors constructed here and our results on protein expression ratios from different 2A constructs could serve to guide future utilization of 2A peptides in basic research and clinical applications.
Collapse
Affiliation(s)
- Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Olivia Chen
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - J Blake Joseph Wall
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Michael Zheng
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Haley Ruth Vaseghi
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA. .,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA. .,McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
92
|
Kurotsu S, Suzuki T, Ieda M. Direct Reprogramming, Epigenetics, and Cardiac Regeneration. J Card Fail 2017; 23:552-557. [PMID: 28529134 DOI: 10.1016/j.cardfail.2017.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 01/14/2023]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has revolutionized regenerative medicine. Autologous iPSCs can be generated by introducing 4 stem cell-specific factors (Oct4, Sox2, Klf4, c-Myc) into fibroblasts. iPSCs can propagate indefinitely and differentiate into clinically important cell types, including cardiomyocytes, in vitro. The iPSC-derived cardiomyocytes represent a promising source of cells for cell-based therapeutic approaches for cardiac regeneration. However, there are several challenges in the clinical application of iPSCs: tumorigenicity of immature cells, poor survival of the transplanted myocardial cells, and cost and efficacy of this therapeutic approach. We developed a new alternate approach for cardiac regeneration, called direct cardiac reprogramming. Instead of using stem cell factors, we overexpressed combinations of cardiac cell-specific genes in fibroblasts to directly induce cardiomyocytes without mediating through iPSCs. The direct reprogramming approach may overcome the challenges faced in the applicability of iPSC-based cell therapy. After the development of direct cardiac reprogramming, great progress has been made in improving the efficiency of direct cardiac reprogramming and applying this technology to regenerative medicine. Here, we provide an overview of the recent progress made, epigenetics, and potential clinical applications of direct cardiac reprogramming.
Collapse
Affiliation(s)
- Shota Kurotsu
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Amed Prime, Tokyo, Japan; Division of Basic Biologic Sciences, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Takeshi Suzuki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Division of Basic Biologic Sciences, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Amed Prime, Tokyo, Japan.
| |
Collapse
|
93
|
Abstract
Myocardial infarction afflicts close to three quarters of a million Americans annually, resulting in reduced heart function, arrhythmia, and frequently death. Cardiomyocyte death reduces the heart’s pump capacity while the deposition of a non-conductive scar incurs the risk of arrhythmia. Direct cardiac reprogramming emerged as a novel technology to simultaneously reduce scar tissue and generate new cardiomyocytes to restore cardiac function. This technology converts endogenous cardiac fibroblasts directly into induced cardiomyocyte-like cells using a variety of cocktails including transcription factors, microRNAs, and small molecules. Although promising, direct cardiac reprogramming is still in its fledging phase, and numerous barriers have to be overcome prior to its clinical application. This review discusses current findings to optimize reprogramming efficiency, including reprogramming factor cocktails and stoichiometry, epigenetic barriers to cell fate reprogramming, incomplete conversion and residual fibroblast identity, requisite growth factors, and environmental cues. Finally, we address the current challenges and future directions for the field.
Collapse
|
94
|
Dal-Pra S, Hodgkinson CP, Mirotsou M, Kirste I, Dzau VJ. Demethylation of H3K27 Is Essential for the Induction of Direct Cardiac Reprogramming by miR Combo. Circ Res 2017; 120:1403-1413. [PMID: 28209718 DOI: 10.1161/circresaha.116.308741] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 01/10/2023]
Abstract
RATIONALE Direct reprogramming of cardiac fibroblasts to cardiomyocytes has recently emerged as a novel and promising approach to regenerate the injured myocardium. We have previously demonstrated the feasibility of this approach in vitro and in vivo using a combination of 4 microRNAs (miR-1, miR-133, miR-208, and miR-499) that we named miR combo. However, the mechanism of miR combo mediated direct cardiac reprogramming is currently unknown. OBJECTIVE Here, we investigated the possibility that miR combo initiated direct cardiac reprogramming through an epigenetic mechanism. METHODS AND RESULTS Using a quantitative polymerase chain reaction array, we found that histone methyltransferases and demethylases that regulate the trimethylation of H3K27 (H3K27me3), an epigenetic modification that marks transcriptional repression, were changed in miR combo-treated fibroblasts. Accordingly, global H3K27me3 levels were downregulated by miR combo treatment. In particular, the promoter region of cardiac transcription factors showed decreased H3K27me3 as revealed by chromatin immunoprecipitation coupled with quantitative polymerase chain reaction. Inhibition of H3K27 methyltransferases or of the PRC2 (Polycomb Repressive Complex 2) by pharmaceutical inhibition or siRNA reduced the levels of H3K27me3 and induced cardiogenic markers at the RNA and protein level, similarly to miR combo treatment. In contrast, knockdown of the H3K27 demethylases Kdm6A and Kdm6B restored the levels of H3K27me3 and blocked the induction of cardiac gene expression in miR combo-treated fibroblasts. CONCLUSIONS In summary, we demonstrated that removal of the repressive mark H3K27me3 is essential for the induction of cardiac reprogramming by miR combo. Our data not only highlight the importance of regulating the epigenetic landscape during cell fate conversion but also provide a framework to improve this technique.
Collapse
Affiliation(s)
- Sophie Dal-Pra
- From the Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Conrad P Hodgkinson
- From the Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Maria Mirotsou
- From the Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Imke Kirste
- From the Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Victor J Dzau
- From the Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC.
| |
Collapse
|
95
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
96
|
Liu L, Lei I, Karatas H, Li Y, Wang L, Gnatovskiy L, Dou Y, Wang S, Qian L, Wang Z. Targeting Mll1 H3K4 methyltransferase activity to guide cardiac lineage specific reprogramming of fibroblasts. Cell Discov 2016; 2:16036. [PMID: 27924221 PMCID: PMC5113048 DOI: 10.1038/celldisc.2016.36] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers a great opportunity for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate. To address this issue, we attempted to identify small molecules that could potentiate the reprogramming ability towards cardiac fate by removing inhibitory roadblocks. Using mouse embryonic fibroblasts as the starting cell source, we first screened 47 cardiac development related epigenetic and transcription factors, and identified an unexpected role of H3K4 methyltransferase Mll1 and related factor Men1 in inhibiting iCM reprogramming. We then applied small molecules (MM408 and MI503) of Mll1 pathway inhibitors and observed an improved efficiency in converting embryonic fibroblasts and cardiac fibroblasts into functional cardiomyocyte-like cells. We further observed that these inhibitors directly suppressed the expression of Mll1 target gene Ebf1 involved in adipocyte differentiation. Consequently, Mll1 inhibition significantly decreased the formation of adipocytes during iCM induction. Therefore, Mll1 inhibitors likely increased iCM efficiency by suppressing alternative lineage gene expression. Our studies show that targeting Mll1 dependent H3K4 methyltransferase activity provides specificity in the process of cardiac reprogramming. These findings shed new light on the molecular mechanisms underlying cardiac conversion of fibroblasts and provide novel targets and small molecules to improve iCM reprogramming for clinical applications.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI, USA; Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hacer Karatas
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yangbing Li
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid Gnatovskiy
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Yali Dou
- Department of Pathology, University of Michigan Medical School, 1301 Catherine , Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
97
|
Affiliation(s)
- Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|