1
|
Yang J. Partial Cell Fate Transitions to Promote Cardiac Regeneration. Cells 2024; 13:2002. [PMID: 39682750 DOI: 10.3390/cells13232002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Heart disease, including myocardial infarction (MI), remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective regenerative therapies. Direct reprogramming of cardiomyocyte-like cells from resident fibroblasts offers a promising avenue for myocardial regeneration, but its efficiency and consistency in generating functional cardiomyocytes remain limited. Alternatively, reprogramming induced cardiac progenitor cells (iCPCs) could generate essential cardiac lineages, but existing methods often involve complex procedures. These limitations underscore the need for advanced mechanistic insights and refined reprogramming strategies to improve reparative outcomes in the heart. Partial cellular fate transitions, while still a relatively less well-defined area and primarily explored in longevity and neurobiology, hold remarkable promise for cardiac repair. It enables the reprogramming or rejuvenation of resident cardiac cells into a stem or progenitor-like state with enhanced cardiogenic potential, generating the reparative lineages necessary for comprehensive myocardial recovery while reducing safety risks. As an emerging strategy, partial cellular fate transitions play a pivotal role in reversing myocardial infarction damage and offer substantial potential for therapeutic innovation. This review will summarize current advances in these areas, including recent findings involving two transcription factors that critically regulate stemness and cardiogenesis. It will also explore considerations for further refining these approaches to enhance their therapeutic potential and safety.
Collapse
Affiliation(s)
- Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
2
|
Pathan SS, Pugazenthi A, Dixon BREA, Wensel TG, Rosengart TK, Mathison M. Activation of a GPCR, ORL1 Receptor: A Novel Therapy to Prevent Heart Failure Progression. J Cardiovasc Dev Dis 2024; 11:355. [PMID: 39590198 PMCID: PMC11595155 DOI: 10.3390/jcdd11110355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
The number of ischemic heart failure (HF) patients is growing dramatically worldwide. However, there are at present no preventive treatments for HF. Our previous study showed that Gata4 overexpression improved cardiac function after myocardial infarction in rat hearts. We also found that Gata4 overexpression significantly increased the expression of a Pnoc gene, an endogenous ligand for the cell membrane receptor ORL1. We hypothesized that the activation of the ORL1 receptor would suppress HF in a rat ischemic heart model. Adult Sprague Dawley rats (8 weeks old, six males and six females) underwent left anterior descending coronary artery ligation. Three weeks later, normal saline or MCOPPB (ORL1 activator, 2.5 mg/kg/day) intraperitoneal injection was started, and continued 5 days a week for 3 months. Echocardiography was performed six times: pre-operative, 3 days after coronary artery ligation, pre-MCOPPB or saline injection, and 1, 2, and 3 months after saline or MCOPPB injection started. Animals were euthanized after 3 months' follow-up and the hearts were harvested for histological analysis. The ORL1 activator, MCOPPB, significantly improved cardiac function after myocardial infarction in rats (ejection fraction, MCOPPB vs. saline at euthanasia, 67 ± 3% vs. 43 ± 2%, p < 0.001). MCOPPB also decreased fibrosis and induced angiogenesis. Thus, the ORL1 activator, MCOPPB, may be a novel treatment for preventing HF progression.
Collapse
Affiliation(s)
- Saliha S. Pathan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Beverly R. E. A. Dixon
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Theodore G. Wensel
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA;
| | - Todd K. Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| |
Collapse
|
3
|
Pathan S, Pugazenthi A, Dixon BR, Wensel TG, Rosengart TK, Mathison M. Activation of a GPCR, ORL1 receptor: A novel therapy to prevent heart failure progression. RESEARCH SQUARE 2024:rs.3.rs-4578315. [PMID: 39070633 PMCID: PMC11275996 DOI: 10.21203/rs.3.rs-4578315/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Purpose The number of ischemic heart failure (HF) patients is growing dramatically worldwide. However, there are at present no preventive treatments for HF. Our previous study showed that Gata4 overexpression improved cardiac function after myocardial infarction in the rat heart. We also found that Gata4 overexpression significantly increased a Pnoc gene expression, an endogenous ligand for cell membrane receptor, ORL1. We hypothesized that an activation of ORL1 receptor would suppress HF in a rat ischemic heart model. Method Adult Sprague Dawley rats (8 weeks old, 6 males and 6 females) underwent left anterior descending coronary artery ligation. Three weeks later, normal saline or MCOPPB (ORL1 activator, 2.5mg/kg/day) intraperitoneal injection was started, and continued 5 days a week, for 3 months. Echocardiography was performed six times, pre-operative, 3 days after coronary artery ligation, pre-MCOPPB or saline injection, and 1, 2, and 3 months after saline or MCOPPB injection started. Animals were euthanized after 3 months follow up and the heart was harvested for histological analysis. Results ORL1 activator, MCOPPB, significantly improved cardiac function after myocardial infarction in rat (Ejection fraction, MCOPPB vs saline at euthanasia, 67 ± 3 vs 43 ± 2, p < 0.001). MCOPPB also decreased fibrosis and induced angiogenesis. Conclusion ORL1 activator, MCOPPB, may be a novel treatment for preventing HF progression.
Collapse
|
4
|
Javanshir E, Ebrahimi ZJ, Mirzohreh ST, Ghaffari S, Banisefid E, Alamdari NM, Roshanravan N. Disparity of gene expression in coronary artery disease: insights from MEIS1, HIRA, and Myocardin. Mol Biol Rep 2024; 51:712. [PMID: 38824221 DOI: 10.1007/s11033-024-09657-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.
Collapse
Affiliation(s)
- Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Banisefid
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Roshanravan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Islam P, Schaly S, Abosalha AK, Boyajian J, Thareja R, Ahmad W, Shum-Tim D, Prakash S. Nanotechnology in development of next generation of stent and related medical devices: Current and future aspects. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1941. [PMID: 38528392 DOI: 10.1002/wnan.1941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/08/2023] [Accepted: 01/03/2024] [Indexed: 03/27/2024]
Abstract
Coronary stents have saved millions of lives in the last three decades by treating atherosclerosis especially, by preventing plaque protrusion and subsequent aneurysms. They attenuate the vascular SMC proliferation and promote reconstruction of the endothelial bed to ensure superior revascularization. With the evolution of modern stent types, nanotechnology has become an integral part of stent technology. Nanocoating and nanosurface fabrication on metallic and polymeric stents have improved their drug loading capacity as well as other mechanical, physico-chemical, and biological properties. Nanofeatures can mimic the natural nanofeatures of vascular tissue and control drug-delivery. This review will highlight the role of nanotechnology in addressing the challenges of coronary stents and the recent advancements in the field of related medical devices. Different generations of stents carrying nanoparticle-based formulations like liposomes, lipid-polymer hybrid NPs, polymeric micelles, and dendrimers are discussed highlighting their roles in local drug delivery and anti-restenotic properties. Drug nanoparticles like Paclitaxel embedded in metal stents are discussed as a feature of first-generation drug-eluting stents. Customized precision stents ensure safe delivery of nanoparticle-mediated genes or concerted transfer of gene, drug, and/or bioactive molecules like antibodies, gene mimics via nanofabricated stents. Nanotechnology can aid such therapies for drug delivery successfully due to its easy scale-up possibilities. However, limitations of this technology such as their potential cytotoxic effects associated with nanoparticle delivery that can trigger hypersensitivity reactions have also been discussed in this review. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Ahmed Kh Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Jacqueline Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Waqar Ahmad
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Dominique Shum-Tim
- Division of Cardiac Surgery, Royal Victoria Hospital, McGill University Health Centre, McGill University, Faculty of Medicine and Health Sciences, Montreal, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Fang J, Yang Q, Maas RGC, Buono M, Meijlink B, Lotgerink Bruinenberg D, Benavente ED, Mokry M, van Mil A, Qian L, Goumans MJ, Schiffelers R, Lei Z, Sluijter JPG. Vitamin C facilitates direct cardiac reprogramming by inhibiting reactive oxygen species. Stem Cell Res Ther 2024; 15:19. [PMID: 38229180 PMCID: PMC10792814 DOI: 10.1186/s13287-023-03615-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND After myocardial infarction, the lost myocardium is replaced by fibrotic tissue, eventually progressively leading to myocardial dysfunction. Direct reprogramming of fibroblasts into cardiomyocytes via the forced overexpression of cardiac transcription factors Gata4, Mef2c, and Tbx5 (GMT) offers a promising strategy for cardiac repair. The limited reprogramming efficiency of this approach, however, remains a significant challenge. METHODS We screened seven factors capable of improving direct cardiac reprogramming of both mice and human fibroblasts by evaluating small molecules known to be involved in cardiomyocyte differentiation or promoting human-induced pluripotent stem cell reprogramming. RESULTS We found that vitamin C (VitC) significantly increased cardiac reprogramming efficiency when added to GMT-overexpressing fibroblasts from human and mice in 2D and 3D model. We observed a significant increase in reactive oxygen species (ROS) generation in human and mice fibroblasts upon Doxy induction, and ROS generation was subsequently reduced upon VitC treatment, associated with increased reprogramming efficiency. However, upon treatment with dehydroascorbic acid, a structural analog of VitC but lacking antioxidant properties, no difference in reprogramming efficiency was observed, suggesting that the effect of VitC in enhancing cardiac reprogramming is partly dependent of its antioxidant properties. CONCLUSIONS Our findings demonstrate that VitC supplementation significantly enhances the efficiency of cardiac reprogramming, partially by suppressing ROS production in the presence of GMT.
Collapse
Affiliation(s)
- Juntao Fang
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Qiangbing Yang
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Renée G C Maas
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Michele Buono
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bram Meijlink
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dyonne Lotgerink Bruinenberg
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ernest Diez Benavente
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michal Mokry
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alain van Mil
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Zhiyong Lei
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands.
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, The Netherlands.
| | - Joost P G Sluijter
- Experimental Cardiology laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Circulatory Health Laboratory, UMC Utrecht, Regenerative Medicine Center Utrecht, University Utrecht, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
7
|
He X, Dutta S, Liang J, Paul C, Huang W, Xu M, Chang V, Ao I, Wang Y. Direct cellular reprogramming techniques for cardiovascular regenerative therapeutics. Can J Physiol Pharmacol 2024; 102:1-13. [PMID: 37903419 DOI: 10.1139/cjpp-2023-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Cardiovascular diseases remain a leading cause of hospitalization affecting approximately 38 million people worldwide. While pharmacological and revascularization techniques can improve the patient's survival and quality of life, they cannot help reversing myocardial infarction injury and heart failure. Direct reprogramming of somatic cells to cardiomyocyte and cardiac progenitor cells offers a new approach to cellular reprogramming and paves the way for translational regenerative medicine. Direct reprogramming can bypass the pluripotent stage with the potential advantage of non-immunogenic cell products, reduced carcinogenic risk, and no requirement for embryonic tissue. The process of directly reprogramming cardiac cells was first achieved through the overexpression of transcription factors such as GATA4, MEF2C, and TBX5. However, over the past decade, significant work has been focused on enhancing direct reprogramming using a mixture of transcription factors, microRNAs, and small molecules to achieve cardiac cell fate. This review discusses the evolution of direct reprogramming, recent progress in achieving efficient cardiac cell fate conversion, and describes the reprogramming mechanisms at a molecular level. We also explore various viral and non-viral delivery methods currently being used to aid in the delivery of reprogramming factors to improve efficiency. However, further studies will be needed to overcome molecular and epigenetic barriers to successfully achieve translational cardiac regenerative therapeutics.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Suchandrima Dutta
- Department of Internal MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Meifeng Xu
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Vivian Chang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Ian Ao
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology & Laboratory MedicineCollege of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
8
|
Xie Y, Van Handel B, Qian L, Ardehali R. Recent advances and future prospects in direct cardiac reprogramming. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1148-1158. [PMID: 39196156 DOI: 10.1038/s44161-023-00377-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/09/2023] [Indexed: 08/29/2024]
Abstract
Cardiovascular disease remains a leading cause of death worldwide despite important advances in modern medical and surgical therapies. As human adult cardiomyocytes have limited regenerative ability, cardiomyocytes lost after myocardial infarction are replaced by fibrotic scar tissue, leading to cardiac dysfunction and heart failure. To replace lost cardiomyocytes, a promising approach is direct cardiac reprogramming, in which cardiac fibroblasts are transdifferentiated into induced cardiomyocyte-like cells (iCMs). Here we review cardiac reprogramming cocktails (including transcription factors, microRNAs and small molecules) that mediate iCM generation. We also highlight mechanistic studies exploring the barriers to and facilitators of this process. We then review recent progress in iCM reprogramming, with a focus on single-cell '-omics' research. Finally, we discuss obstacles to clinical application.
Collapse
Affiliation(s)
- Yifang Xie
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ben Van Handel
- Department of Orthopedic Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Reza Ardehali
- Section of Cardiology, Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA.
- The Texas Heart Institute, Houston, TX, USA.
| |
Collapse
|
9
|
Zhou H, Yang J, Srinath C, Zeng A, Wu I, Leon EC, Qureshi TN, Reid CA, Nettesheim ER, Xu E, Duclos Z, Mohamed TMA, Farshidfar F, Fejes A, Liu J, Jones S, Feathers C, Chung TW, Jing F, Prince WS, Lin J, Yu P, Srivastava D, Hoey T, Ivey KN, Lombardi LM. Improved Cardiac Function in Postischemic Rats Using an Optimized Cardiac Reprogramming Cocktail Delivered in a Single Novel Adeno-Associated Virus. Circulation 2023; 148:1099-1112. [PMID: 37602409 DOI: 10.1161/circulationaha.122.061542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/20/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Cardiac reprogramming is a technique to directly convert nonmyocytes into myocardial cells using genes or small molecules. This intervention provides functional benefit to the rodent heart when delivered at the time of myocardial infarction or activated transgenically up to 4 weeks after myocardial infarction. Yet, several hurdles have prevented the advancement of cardiac reprogramming for clinical use. METHODS Through a combination of screening and rational design, we identified a cardiac reprogramming cocktail that can be encoded in a single adeno-associated virus. We also created a novel adeno-associated virus capsid that can transduce cardiac fibroblasts more efficiently than available parental serotypes by mutating posttranslationally modified capsid residues. Because a constitutive promoter was needed to drive high expression of these cell fate-altering reprogramming factors, we included binding sites to a cardiomyocyte-restricted microRNA within the 3' untranslated region of the expression cassette that limits expression to nonmyocytes. After optimizing this expression cassette to reprogram human cardiac fibroblasts into induced cardiomyocyte-like cells in vitro, we also tested the ability of this capsid/cassette combination to confer functional benefit in acute mouse myocardial infarction and chronic rat myocardial infarction models. RESULTS We demonstrated sustained, dose-dependent improvement in cardiac function when treating a rat model 2 weeks after myocardial infarction, showing that cardiac reprogramming, when delivered in a single, clinically relevant adeno-associated virus vector, can support functional improvement in the postremodeled heart. This benefit was not observed with GFP (green fluorescent protein) or a hepatocyte reprogramming cocktail and was achieved even in the presence of immunosuppression, supporting myocyte formation as the underlying mechanism. CONCLUSIONS Collectively, these results advance the application of cardiac reprogramming gene therapy as a viable therapeutic approach to treat chronic heart failure resulting from ischemic injury.
Collapse
Affiliation(s)
- Huanyu Zhou
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Jin Yang
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Chetan Srinath
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Aliya Zeng
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Iris Wu
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Elena C Leon
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Tawny Neal Qureshi
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Christopher A Reid
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Emily R Nettesheim
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Emma Xu
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Zoe Duclos
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Tamer M A Mohamed
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Farshad Farshidfar
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Anthony Fejes
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Jun Liu
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Samantha Jones
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Charles Feathers
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Tae Won Chung
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Frank Jing
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - William S Prince
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - JianMin Lin
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Pengzhi Yu
- Gladstone Institutes, San Francisco, CA (P.Y., D.S.)
| | - Deepak Srivastava
- Gladstone Institutes, San Francisco, CA (P.Y., D.S.)
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA (D.S.)
- Division of Cardiology, Department of Pediatrics, School of Medicine (D.S.), University of California San Francisco
- Department of Biochemistry and Biophysics (D.S.), University of California San Francisco
| | - Timothy Hoey
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Kathryn N Ivey
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| | - Laura M Lombardi
- Tenaya Therapeutics, Inc, South San Francisco, CA (H.Z., J.Y., C.S., A.Z., I.W., E.C.L., T.N.Q., C.A.R., E.R.N., E.X., Z.D., T.M.A.M., F.F., A.F., J. Liu, S.J., C.F., T.W.C., F.J., W.S.P., J. Lin, T.H., K.N.I., L.M.L.)
| |
Collapse
|
10
|
Yuan X, Braun T. Amending the injured heart by in vivo reprogramming. Curr Opin Genet Dev 2023; 82:102098. [PMID: 37595409 DOI: 10.1016/j.gde.2023.102098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/20/2023]
Abstract
Ischemic heart injury causes death of cardiomyocyte (CM), formation of a fibrotic scar, and often adverse cardiac remodeling, resulting in chronic heart failure. Therapeutic interventions have lowered myocardial damage and improved heart function, but pharmacological treatment of heart failure has only shown limited progress in recent years. Over the past two decades, different approaches have been pursued to regenerate the heart, by transplantation of newly generated CMs derived from pluripotent stem cells, generation of new CMs by reprogramming of cardiac fibroblasts, or by activating proliferation of preexisting CMs. Here, we summarize recent progress in the development of strategies for in situ generation of new CMs, review recent advances in understanding the underlying mechanisms, and discuss the challenges and future directions of the field.
Collapse
Affiliation(s)
- Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Brlecic PE, Bonham CA, Rosengart TK, Mathison M. Direct cardiac reprogramming: A new technology for cardiac repair. J Mol Cell Cardiol 2023; 178:51-58. [PMID: 36965701 PMCID: PMC10124164 DOI: 10.1016/j.yjmcc.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide, with myocardial infarctions being amongst the deadliest manifestations. Reduced blood flow to the heart can result in the death of cardiac tissue, leaving affected patients susceptible to further complications and recurrent disease. Further, contemporary management typically involves a pharmacopeia to manage the metabolic conditions contributing to atherosclerotic and hypertensive heart disease, rather than regeneration of the damaged myocardium. With modern healthcare extending lifespan, a larger demographic will be at risk for heart disease, driving the need for novel therapeutics that surpass those currently available in efficacy. Transdifferentiation and cellular reprogramming have been looked to as potential methods for the treatment of diseases throughout the body. Specifically targeting the fibrotic cells in cardiac scar tissue as a source to be reprogrammed into induced cardiomyocytes remains an appealing option. This review aims to highlight the history of and advances in cardiac reprogramming and describe its translational potential as a treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Paige E Brlecic
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Clark A Bonham
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Yamada Y, Sadahiro T, Ieda M. Development of direct cardiac reprogramming for clinical applications. J Mol Cell Cardiol 2023; 178:1-8. [PMID: 36918145 DOI: 10.1016/j.yjmcc.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
The incidence of cardiovascular diseases is increasing worldwide, and cardiac regenerative therapy has great potential as a new treatment strategy, especially for ischemic heart disease. Direct cardiac reprogramming is a promising new cardiac regenerative therapy that uses defined factors to induce transdifferentiation of endogenous cardiac fibroblasts (CFs) into induced cardiomyocyte-like cells (iCMs). In vivo reprogramming is expected to restore lost cardiac function without necessitating cardiac transplantation by converting endogenous CFs that exist abundantly in cardiac tissues directly into iCMs. Indeed, we and other groups have demonstrated that in vivo cardiac reprogramming improves cardiac contractile function and reduces scar area after acute myocardial infarction (MI). Recently, we demonstrated that in vivo cardiac reprogramming is an innovative cardiac regenerative therapy that not only regenerates the myocardium, but also reverses fibrosis by inducing the quiescence of pro-fibrotic fibroblasts, thereby improving heart failure in chronic MI. In this review, we summarize the recent progresses in in vivo cardiac reprogramming, and discuss its prospects for future clinical applications and the challenges of direct human reprogramming, which has been a longstanding issue.
Collapse
Affiliation(s)
- Yu Yamada
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki 305-8575, Japan
| | - Taketaro Sadahiro
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki 305-8575, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki 305-8575, Japan.
| |
Collapse
|
13
|
Jt S, M H, Wam B, Ac B, Sa N. Adenoviral vectors for cardiovascular gene therapy applications: a clinical and industry perspective. J Mol Med (Berl) 2022; 100:875-901. [PMID: 35606652 PMCID: PMC9126699 DOI: 10.1007/s00109-022-02208-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
Abstract Despite the development of novel pharmacological treatments, cardiovascular disease morbidity and mortality remain high indicating an unmet clinical need. Viral gene therapy enables targeted delivery of therapeutic transgenes and represents an attractive platform for tackling acquired and inherited cardiovascular diseases in the future. Current cardiovascular gene therapy trials in humans mainly focus on improving cardiac angiogenesis and function. Encouragingly, local delivery of therapeutic transgenes utilising first-generation human adenovirus serotype (HAd)-5 is safe in the short term and has shown some efficacy in drug refractory angina pectoris and heart failure with reduced ejection fraction. Despite this success, systemic delivery of therapeutic HAd-5 vectors targeting cardiovascular tissues and internal organs is limited by negligible gene transfer to target cells, elimination by the immune system, liver sequestration, off-target effects, and episomal degradation. To circumvent these barriers, cardiovascular gene therapy research has focused on determining the safety and efficacy of rare alternative serotypes and/or genetically engineered adenoviral capsid protein-modified vectors following local or systemic delivery. Pre-clinical studies have identified several vectors including HAd-11, HAd-35, and HAd-20–42-42 as promising platforms for local and systemic targeting of vascular endothelial and smooth muscle cells. In the past, clinical gene therapy trials were often restricted by limited scale-up capabilities of gene therapy medicinal products (GTMPs) and lack of regulatory guidance. However, significant improvement of industrial GTMP scale-up and purification, development of novel producer cell lines, and issuing of GTMP regulatory guidance by national regulatory health agencies have addressed many of these challenges, creating a more robust framework for future adenoviral-based cardiovascular gene therapy. In addition, this has enabled the mass roll out of adenovirus vector-based COVID-19 vaccines. Key messages First-generation HAd-5 vectors are widely used in cardiovascular gene therapy. HAd-5-based gene therapy was shown to lead to cardiac angiogenesis and improved function. Novel HAd vectors may represent promising transgene carriers for systemic delivery. Novel methods allow industrial scale-up of rare/genetically altered Ad serotypes. National regulatory health agencies have issued guidance on GMP for GTMPs.
Collapse
Affiliation(s)
- Schwartze Jt
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Havenga M
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bakker Wam
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bradshaw Ac
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nicklin Sa
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
14
|
Nam YJ. Translational perspectives on cardiac reprogramming. Semin Cell Dev Biol 2022; 122:14-20. [PMID: 34210578 PMCID: PMC8712611 DOI: 10.1016/j.semcdb.2021.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/21/2021] [Accepted: 06/23/2021] [Indexed: 02/03/2023]
Abstract
Loss of cardiac muscle after cardiac injury is replaced by cardiac fibrosis, due to very limited regenerative capacity of the heart. Although initially beneficial, persistent cardiac fibrosis leads to pump failure and conduction abnormalities, common modes of death following cardiac injury. Thus, directly reprogramming cardiac fibroblasts into induced cardiomyocyte-like cells (iCMs) by forced expression of cardiogenic factors (referred to as cardiac reprogramming) is particularly attractive in that it targets cardiac fibroblasts, a major source of cardiac fibrosis, to induce new cardiac muscle. Over the last decade, remarkable progresses have been made on cardiac reprogramming, particularly focusing on how to enhance conversion of fibroblasts to iCMs in vitro. However, it still remains elusive whether this new regenerative approach can be translated into clinical practice. This review discusses progresses and challenges of cardiac reprogramming in the translational context.
Collapse
Affiliation(s)
- Young-Jae Nam
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA,To whom correspondence may be addressed: Young-Jae Nam, M.D., Ph.D., Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville , Tennessee 37232, USA, Phone: 615-936-5436,
| |
Collapse
|
15
|
Zhou Y, Zhang J. Remuscularization of Ventricular Infarcts Using the Existing Cardiac Cells. ADVANCED TECHNOLOGIES IN CARDIOVASCULAR BIOENGINEERING 2022:51-78. [DOI: 10.1007/978-3-030-86140-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Singh VP, Pinnamaneni JP, Pugazenthi A, Sanagasetti D, Mathison M, Martin JF, Yang J, Rosengart TK. Hippo Pathway Effector Tead1 Induces Cardiac Fibroblast to Cardiomyocyte Reprogramming. J Am Heart Assoc 2021; 10:e022659. [PMID: 34889103 PMCID: PMC9075224 DOI: 10.1161/jaha.121.022659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/09/2021] [Indexed: 01/14/2023]
Abstract
Background The conversion of fibroblasts into induced cardiomyocytes may regenerate myocardial tissue from cardiac scar through in situ cell transdifferentiation. The efficiency transdifferentiation is low, especially for human cells. We explored the leveraging of Hippo pathway intermediates to enhance induced cardiomyocyte generation. Methods and Results We screened Hippo effectors Yap (yes-associated protein), Taz (transcriptional activator binding domain), and Tead1 (TEA domain transcription factor 1; Td) for their reprogramming efficacy with cardio-differentiating factors Gata4, Mef2C, and Tbx5 (GMT). Td induced nearly 3-fold increased expression of cardiomyocyte marker cTnT (cardiac troponin T) by mouse embryonic and adult rat fibroblasts versus GMT administration alone (P<0.0001), while Yap and Taz failed to enhance cTnT expression. Serial substitution demonstrated that Td replacement of TBX5 induced the greatest cTnT expression enhancement and sarcomere organization in rat fibroblasts treated with all GMT substitutions (GMTd versus GMT: 17±1.2% versus 5.4±0.3%, P<0.0001). Cell contractility (beating) was seen in 6% of GMTd-treated cells by 4 weeks after treatment, whereas no beating GMT-treated cells were observed. Human cardiac fibroblasts likewise demonstrated increased cTnT expression with GMTd versus GMT treatment (7.5±0.3% versus 3.0±0.3%, P<0.01). Mechanistically, GMTd administration increased expression of the trimethylated lysine 4 of histone 3 (H3K4me3) mark at the promoter regions of cardio-differentiation genes and mitochondrial biogenesis regulator genes in rat and human fibroblast, compared with GMT. Conclusions These data suggest that the Hippo pathway intermediate Tead1 is an important regulator of cardiac reprogramming that increases the efficiency of maturate induced cardiomyocytes generation and may be a vital component of human cardiodifferentiation strategies.
Collapse
Affiliation(s)
- Vivek P. Singh
- Department of SurgeryBaylor College of MedicineHoustonTX
| | | | | | | | | | - James F. Martin
- Department of Molecular Physiology and BiophysicsBaylor College of MedicineHoustonTX
| | - Jianchang Yang
- Department of SurgeryBaylor College of MedicineHoustonTX
| | | |
Collapse
|
17
|
Liu L, Guo Y, Li Z, Wang Z. Improving Cardiac Reprogramming for Heart Regeneration in Translational Medicine. Cells 2021; 10:cells10123297. [PMID: 34943805 PMCID: PMC8699771 DOI: 10.3390/cells10123297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
Direct reprogramming of fibroblasts into CM-like cells has emerged as an attractive strategy to generate induced CMs (iCMs) in heart regeneration. However, low conversion rate, poor purity, and the lack of precise conversion of iCMs are still present as significant challenges. In this review, we summarize the recent development in understanding the molecular mechanisms of cardiac reprogramming with various strategies to achieve more efficient iCMs. reprogramming. Specifically, we focus on the identified critical roles of transcriptional regulation, epigenetic modification, signaling pathways from the cellular microenvironment, and cell cycling regulation in cardiac reprogramming. We also discuss the progress in delivery system optimization and cardiac reprogramming in human cells related to preclinical applications. We anticipate that this will translate cardiac reprogramming-based heart therapy into clinical applications. In addition to optimizing the cardiogenesis related transcriptional regulation and signaling pathways, an important strategy is to modulate the pathological microenvironment associated with heart injury, including inflammation, pro-fibrotic signaling pathways, and the mechanical properties of the damaged myocardium. We are optimistic that cardiac reprogramming will provide a powerful therapy in heart regenerative medicine.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA; (L.L.); (Y.G.); (Z.L.)
| | - Yijing Guo
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA; (L.L.); (Y.G.); (Z.L.)
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zhaokai Li
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA; (L.L.); (Y.G.); (Z.L.)
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA; (L.L.); (Y.G.); (Z.L.)
- Correspondence:
| |
Collapse
|
18
|
Fibroblast transition to an endothelial "trans" state improves cell reprogramming efficiency. Sci Rep 2021; 11:22605. [PMID: 34799643 PMCID: PMC8604927 DOI: 10.1038/s41598-021-02056-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/02/2021] [Indexed: 01/04/2023] Open
Abstract
Fibroblast reprogramming offers the potential for myocardial regeneration via in situ cell transdifferentiation. We explored a novel strategy leveraging endothelial cell plasticity to enhance reprogramming efficiency. Rat cardiac endothelial cells and fibroblasts were treated with Gata4, Mef2c, and Tbx5 (GMT) to assess the cardio-differentiation potential of these cells. The endothelial cell transdifferentiation factor ETV2 was transiently over-expressed in fibroblasts followed by GMT treatment to assess “trans-endothelial” cardio-differentiation. Endothelial cells treated with GMT generated more cTnT+ cells than did cardiac fibroblasts (13% ± 2% vs 4% ± 0.5%, p < 0.01). Cardiac fibroblasts treated with ETV2 demonstrated increased endothelial cell markers, and when then treated with GMT yielded greater prevalence of cells expressing cardiomyocyte markers including cTnT than did fibroblasts treated with GMT or ETV2 (10.3% ± 0.2% vs 1.7% ± 0.06% and 0.6 ± 0.03, p < 0.01). Rat cardiac fibroblasts treated with GMT + ETV2 demonstrated calcium transients upon electrical stimulation and contractility synchronous with surrounding neonatal cardiomyocytes, whereas cells treated with GMT or ETV2 alone failed to contract in co-culture experiments. Human cardiac fibroblasts treated with ETV2 and then GMT likewise demonstrated greater prevalence of cTnT expression than did cells treated with GMT alone (2.8-fold increase, p < 0.05). Cardiac fibroblast transitioning through a trans-endothelial state appears to enhance cardio-differentiation by enhancing fibroblast plasticity.
Collapse
|
19
|
Ryan CT, Patel V, Rosengart TK. Clinical potential of angiogenic therapy and cellular reprogramming. ACTA ACUST UNITED AC 2021; 6:108-115. [PMID: 34746874 PMCID: PMC8570572 DOI: 10.1016/j.xjon.2020.12.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Christopher T Ryan
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Vivek Patel
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Todd K Rosengart
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| |
Collapse
|
20
|
Cell Transdifferentiation and Reprogramming in Disease Modeling: Insights into the Neuronal and Cardiac Disease Models and Current Translational Strategies. Cells 2021; 10:cells10102558. [PMID: 34685537 PMCID: PMC8533873 DOI: 10.3390/cells10102558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Cell transdifferentiation and reprogramming approaches in recent times have enabled the manipulation of cell fate by enrolling exogenous/artificial controls. The chemical/small molecule and regulatory components of transcription machinery serve as potential tools to execute cell transdifferentiation and have thereby uncovered new avenues for disease modeling and drug discovery. At the advanced stage, one can believe these methods can pave the way to develop efficient and sensitive gene therapy and regenerative medicine approaches. As we are beginning to learn about the utility of cell transdifferentiation and reprogramming, speculations about its applications in translational therapeutics are being largely anticipated. Although clinicians and researchers are endeavoring to scale these processes, we lack a comprehensive understanding of their mechanism(s), and the promises these offer for targeted and personalized therapeutics are scarce. In the present report, we endeavored to provide a detailed review of the original concept, methods and modalities enrolled in the field of cellular transdifferentiation and reprogramming. A special focus is given to the neuronal and cardiac systems/diseases towards scaling their utility in disease modeling and drug discovery.
Collapse
|
21
|
Testa G, Di Benedetto G, Passaro F. Advanced Technologies to Target Cardiac Cell Fate Plasticity for Heart Regeneration. Int J Mol Sci 2021; 22:ijms22179517. [PMID: 34502423 PMCID: PMC8431232 DOI: 10.3390/ijms22179517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
The adult human heart can only adapt to heart diseases by starting a myocardial remodeling process to compensate for the loss of functional cardiomyocytes, which ultimately develop into heart failure. In recent decades, the evolution of new strategies to regenerate the injured myocardium based on cellular reprogramming represents a revolutionary new paradigm for cardiac repair by targeting some key signaling molecules governing cardiac cell fate plasticity. While the indirect reprogramming routes require an in vitro engineered 3D tissue to be transplanted in vivo, the direct cardiac reprogramming would allow the administration of reprogramming factors directly in situ, thus holding great potential as in vivo treatment for clinical applications. In this framework, cellular reprogramming in partnership with nanotechnologies and bioengineering will offer new perspectives in the field of cardiovascular research for disease modeling, drug screening, and tissue engineering applications. In this review, we will summarize the recent progress in developing innovative therapeutic strategies based on manipulating cardiac cell fate plasticity in combination with bioengineering and nanotechnology-based approaches for targeting the failing heart.
Collapse
Affiliation(s)
- Gianluca Testa
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
- Interdepartmental Center for Nanotechnology Research—NanoBem, University of Molise, 86100 Campobasso, Italy
| | - Giorgia Di Benedetto
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80138 Naples, Italy;
| | - Fabiana Passaro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80138 Naples, Italy;
- Correspondence:
| |
Collapse
|
22
|
Sadahiro T, Ieda M. In vivo reprogramming as a new approach to cardiac regenerative therapy. Semin Cell Dev Biol 2021; 122:21-27. [PMID: 34210577 DOI: 10.1016/j.semcdb.2021.06.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are a common cause of death worldwide. Adult cardiomyocytes have limited regenerative capacity after injury, and there is growing interest in cardiac regeneration as a new therapeutic strategy. There are several limitations of induced pluripotent stem cell-based transplantation therapy with respect to efficiency and risks of tumorigenesis. Direct reprogramming enables the conversion of terminally differentiated cells into target cell types using defined factors. In most cardiac diseases, activated fibroblasts proliferate in the damaged heart and contribute to the progression of heart failure. In vivo cardiac reprogramming, in which resident cardiac fibroblasts are converted into cardiomyocytes in situ, is expected to become a new cardiac regenerative therapy. Indeed, we and other groups have demonstrated that in vivo reprogramming improves cardiac function and reduces fibrosis after myocardial infarction. In this review, we summarize recent discoveries and developments related to in vivo reprogramming. In addition, issues that need to be resolved for clinical application are described.
Collapse
Affiliation(s)
- Taketaro Sadahiro
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki 305-8575, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba City, Ibaraki 305-8575, Japan.
| |
Collapse
|
23
|
Nishida H, Ota T. Commentary: Cardiac cellular “time travel” to when it was bouncing. JTCVS OPEN 2021; 6:118-119. [PMID: 36003554 PMCID: PMC9390163 DOI: 10.1016/j.xjon.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 10/27/2022]
|
24
|
Van Handel B, Wang L, Ardehali R. Environmental factors influence somatic cell reprogramming to cardiomyocyte-like cells. Semin Cell Dev Biol 2021; 122:44-49. [PMID: 34083115 DOI: 10.1016/j.semcdb.2021.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 12/11/2022]
Abstract
Direct cardiac reprogramming, which refers to somatic cell (i.e. fibroblast) fate conversion to cardiomyocyte-like cell without transitioning through an intermediate pluripotent state, provides a novel therapeutic strategy for heart regeneration by converting resident cardiac fibroblasts to cardiomyocytes in situ. However, several limitations need to be addressed prior to clinical translation of this technology. They include low efficiency of reprogramming, heterogeneity of starting fibroblasts, functional immaturity of induced cardiomyocytes (iCMs), virus immunogenicity and toxicity, incomplete understanding of changes in the epigenetic landscape as fibroblasts undergo reprogramming, and the environmental factors that influence fate conversion. Several studies have demonstrated that a combination of enforced expression of cardiac transcription factors along with certain cytokines and growth factors in the presence of favorable environmental cues (including extracellular matrix, topography, and mechanical properties) enhance the efficiency and quality of direct reprogramming. This paper reviews the literature on the influence of the microenvironment on direct cardiac reprogramming in vitro and in vivo.
Collapse
Affiliation(s)
- Ben Van Handel
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Orthopedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, 90033, USA
| | - Lingjun Wang
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Ghanta RK, Aghlara-Fotovat S, Pugazenthi A, Ryan CT, Singh VP, Mathison M, Jarvis MI, Mukherjee S, Hernandez A, Veiseh O. Immune-modulatory alginate protects mesenchymal stem cells for sustained delivery of reparative factors to ischemic myocardium. Biomater Sci 2021; 8:5061-5070. [PMID: 32797143 DOI: 10.1039/d0bm00855a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Paracrine factors secreted by mesenchymal stem cells (MSCs) have been previously shown to improve cardiac function following acute myocardial infarction (MI). However, cell therapy activates the innate immune response, leading to the rapid elimination of transplanted cells and only short-term therapeutic delivery. Herein, we describe a new strategy to deliver sustained paracrine-mediated MSC therapy to ischemic myocardium. Using an immune evasive, small molecule modified alginate, we encapsulated rat MSC cells in a core-shell hydrogel capsule and implanted them in the pericardial sac of post-MI rats. Encapsulated cells allowed diffusion of reparative paracrine factors at levels similar to non-encapsulated cells in vitro. Encapsulation enabled sustained cell survival with localization over the heart for 2 weeks. The effect of the experimental group on ventricular function and fibrosis was compared with blank (cell free) capsules and unencapsulated MSCs injected into infarcted myocardium. MSC capsules improved post-MI ventricular function ∼2.5× greater than MSC injection. After 4 weeks, post-MI fibrosis was reduced ∼2/3 with MSC capsules, but unchanged with MSC injection. MSC encapsulation with alginate core-shell capsules sustains cell survival and potentiates efficacy of therapy.
Collapse
Affiliation(s)
- Ravi K Ghanta
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | | | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Christopher T Ryan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Vivek P Singh
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Andrea Hernandez
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
26
|
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
|
27
|
The Future of Direct Cardiac Reprogramming: Any GMT Cocktail Variety? Int J Mol Sci 2020; 21:ijms21217950. [PMID: 33114756 PMCID: PMC7663133 DOI: 10.3390/ijms21217950] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings.
Collapse
|
28
|
Fathi E, Farahzadi R, Vietor I, Javanmardi S. Cardiac differentiation of bone-marrow-resident c-kit+ stem cells by L-carnitine increases through secretion of VEGF, IL6, IGF-1, and TGF-β as clinical agents in cardiac regeneration. J Biosci 2020. [DOI: 10.1007/s12038-020-00063-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Abstract
PURPOSE OF REVIEW The high global incidence of heart disease drives the need for methods of mending damaged hearts. Direct reprogramming of cardiac fibroblasts into cardiomyocyte-like cells (called iCMs) has been successful in the creation of new muscle cells, in the repair of hearts post-myocardial injury, and therefore has great promise for the clinic. The purpose of this paper is to review and highlight the approaches for and underlying molecular mechanisms of direct cardiac reprogramming. RECENT FINDINGS Single-cell genomics and mechanistic studies have elucidated the stepwise transition of fibroblasts to iCMs as well as the molecular roadblocks that hinder reprogramming. Cardiac fibroblasts are able to be directly reprogrammed, in vitro and in vivo, into induced cardiomyocyte-like cells by the ectopic expression of a combination of transcription factors, microRNAs or small molecules. Recent works have illustrated methods that improve the efficiency of iCM generation and delivery of reprogramming cocktails as well as have revealed the molecular networks governing the reprogramming process. Current studies have also begun to identify and address the additional hurdles in human iCM reprogramming.
Collapse
Affiliation(s)
- Gregory Farber
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.
- , Chapel Hill, NC, USA.
| |
Collapse
|
30
|
Singh VP, Pinnamaneni JP, Pugazenthi A, Sanagasetti D, Mathison M, Wang K, Yang J, Rosengart TK. Enhanced Generation of Induced Cardiomyocytes Using a Small-Molecule Cocktail to Overcome Barriers to Cardiac Cellular Reprogramming. J Am Heart Assoc 2020; 9:e015686. [PMID: 32500803 PMCID: PMC7429035 DOI: 10.1161/jaha.119.015686] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Given known inefficiencies in reprogramming of fibroblasts into mature induced cardiomyocytes (iCMs), we sought to identify small molecules that would overcome these barriers to cardiac cell transdifferentiation. Methods and Results We screened alternative combinations of compounds known to impact cell reprogramming using morphologic and functional cell differentiation assays in vitro. After screening 6 putative reprogramming factors, we found that a combination of the histone deacetylase inhibitor sodium butyrate, the WNT inhibitor ICG‐001, and the cardiac growth regulator retinoic acid (RA) maximally enhanced iCM generation from primary rat cardiac fibroblasts when combined with administration of the cardiodifferentiating transcription factors Gata4, Mef2C, and Tbx5 (GMT) compared with GMT administration alone (23±1.5% versus 3.3±0.2%; P<0.0001). Expression of the cardiac markers cardiac troponin T, Myh6, and Nkx2.5 was upregulated as early as 10 days after GMT–sodium butyrate, ICG‐001, and RA treatment. Human iCM generation was likewise enhanced when administration of the human cardiac reprogramming factors GMT, Hand2, and Myocardin plus miR‐590 was combined with sodium butyrate, ICG‐001, and RA compared with GMT, Hand2, and Myocardin plus miR‐590 treatment alone (25±1.3% versus 5.7±0.4%; P<0.0001). Rat and human iCMs also more frequently demonstrated spontaneous beating in coculture with neonatal cardiomyocytes with the addition of sodium butyrate, ICG‐001, and RA to transcription factor cocktails compared with transcription factor treatment alone. Conclusions The combined administration of histone deacetylase and WNT inhibitors with RA enhances rat and human iCM generation induced by transcription factor administration alone. These findings suggest opportunities for improved translational approaches for cardiac regeneration.
Collapse
Affiliation(s)
- Vivek P Singh
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | | | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Deepthi Sanagasetti
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Kai Wang
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Jianchang Yang
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| |
Collapse
|
31
|
Yan C, Quan XJ, Feng YM. Nanomedicine for Gene Delivery for the Treatment of Cardiovascular Diseases. Curr Gene Ther 2020; 19:20-30. [PMID: 30280665 PMCID: PMC6751340 DOI: 10.2174/1566523218666181003125308] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/21/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Abstract
Background: Myocardial infarction (MI) is the most severe ischemic heart disease and di-rectly leads to heart failure till death. Target molecules have been identified in the event of MI including increasing angiogenesis, promoting cardiomyocyte survival, improving heart function and restraining inflammation and myocyte activation and subsequent fibrosis. All of which are substantial in cardiomy-ocyte protection and preservation of cardiac function. Methodology: To modulate target molecule expression, virus and non-virus-mediated gene transfer have been investigated. Despite successful in animal models of MI, virus-mediated gene transfer is hampered by poor targeting efficiency, low packaging capacity for large DNA sequences, immunogenicity induced by virus and random integration into the human genome. Discussion: Nanoparticles could be synthesized and equipped on purpose for large-scale production. They are relatively small in size and do not incorporate into the genome. They could carry DNA and drug within the same transfer. All of these properties make them an alternative strategy for gene transfer. In the review, we first introduce the pathological progression of MI. After concise discussion on the current status of virus-mediated gene therapy in treating MI, we overview the history and development of nanoparticle-based gene delivery system. We point out the limitations and future perspective in the field of nanoparticle vehicle. Conclusion: Ultimately, we hope that this review could help to better understand how far we are with nanoparticle-facilitated gene transfer strategy and what obstacles we need to solve for utilization of na-nomedicine in the treatment of MI.
Collapse
Affiliation(s)
- Cen Yan
- Beijing Key Laboratory of Diabetes Prevention and Research, Endocrinology Center, Lu He Hospital, Capital Medical University, Beijing 101149, China
| | - Xiao-Jiang Quan
- Laboratory of Brain Development, Institut du Cerveau et de la Moelle Epiniere- ICM, Hospital Pitie-Salpetriere, 75013 Paris, France
| | - Ying-Mei Feng
- Beijing Key Laboratory of Diabetes Prevention and Research, Endocrinology Center, Lu He Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
32
|
Song SY, Kim H, Yoo J, Kwon SP, Park BW, Kim JJ, Ban K, Char K, Park HJ, Kim BS. Prevascularized, multiple-layered cell sheets of direct cardiac reprogrammed cells for cardiac repair. Biomater Sci 2020; 8:4508-4520. [DOI: 10.1039/d0bm00701c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We developed cardiac-reprogrammed cell sheets via cardiac-mimetic cell culture system with biodegradable PLGA membrane. The prevascularized, multiple-layered cell sheets prevented heart failure after myocardial infarction.
Collapse
Affiliation(s)
- Seuk Young Song
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Hyeok Kim
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Jin Yoo
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Bong Woo Park
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Jin-ju Kim
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences
- City University of Hong Kong
- Kowloon Tong
- Hong Kong
| | - Kookheon Char
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
| | - Hun-Jun Park
- Department of Medical Life Science
- College of Medicine
- The Catholic University of Korea
- Seoul
- Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul
- Republic of Korea
- Institute of Chemical Processes
| |
Collapse
|
33
|
Li B, Hu Y, Li X, Jin G, Chen X, Chen G, Chen Y, Huang S, Liao W, Liao Y, Teng Z, Bin J. Sirt1 Antisense Long Noncoding RNA Promotes Cardiomyocyte Proliferation by Enhancing the Stability of Sirt1. J Am Heart Assoc 2019; 7:e009700. [PMID: 30608184 PMCID: PMC6404207 DOI: 10.1161/jaha.118.009700] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background Antisense long noncoding RNAs (lncRNAs) are single‐stranded RNAs that overlapped gene‐coding regions on the opposite DNA strand and play as critical regulators in cardiovascular diseases. The high conservation and stability may be good advantages for antisense lncRNAs. However, the roles of antisense lncRNAs in cardiomyocyte proliferation and cardiac regeneration are still unknown. Methods and Results In this study, we found that Silent information regulator factor 2 related enzyme 1 (Sirt1) antisense lncRNA expression was significantly increased during heart development. By gain and loss function of Sirt1 antisense lncRNA using adenovirus and locked nucleic acid, respectively, we demonstrated that Sirt1 antisense lncRNA promoted cardiomyocyte proliferation in vitro and in vivo, and the suppression of Sirt1 antisense lncRNA inhibited cardiomyocyte proliferation. Moreover, overexpression of Sirt1 antisense lncRNA enhanced cardiomyocyte proliferation, attenuated cardiomyocyte apoptosis, improved cardiac function, and decreased mortality rate after myocardial infarction. Furthermore, Sirt1 antisense lncRNA can bind the Sirt1 3′‐untranslated region, enhancing the stability of Sirt1 and increasing Sirt1 abundance at both the mRNA and protein levels. Finally, we found that Sirt1 was involved in Sirt1 antisense lncRNA‐induced cardiomyocyte proliferation. Conclusions The present study identified Sirt1 antisense lncRNA as a novel regulator of cardiomyocyte proliferation and cardiac regeneration by interacting and stabilizing Sirt1 mRNA, which may serve as an effective gene target for preventing myocardial infarction.
Collapse
Affiliation(s)
- Bing Li
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Yinlan Hu
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Xinzhong Li
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Guoqing Jin
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Xiaoqiang Chen
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Guojun Chen
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Yanmei Chen
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Senlin Huang
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Wangjun Liao
- 2 Department of Oncology Nanfang Hospital Southern Medical University Guangzhou China
| | - Yulin Liao
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Zhonghua Teng
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Jianping Bin
- 1 State Key Laboratory of Organ Failure Research Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| |
Collapse
|
34
|
Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac Progenitor Cells from Stem Cells: Learning from Genetics and Biomaterials. Cells 2019; 8:E1536. [PMID: 31795206 PMCID: PMC6952950 DOI: 10.3390/cells8121536] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac Progenitor Cells (CPCs) show great potential as a cell resource for restoring cardiac function in patients affected by heart disease or heart failure. CPCs are proliferative and committed to cardiac fate, capable of generating cells of all the cardiac lineages. These cells offer a significant shift in paradigm over the use of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes owing to the latter's inability to recapitulate mature features of a native myocardium, limiting their translational applications. The iPSCs and direct reprogramming of somatic cells have been attempted to produce CPCs and, in this process, a variety of chemical and/or genetic factors have been evaluated for their ability to generate, expand, and maintain CPCs in vitro. However, the precise stoichiometry and spatiotemporal activity of these factors and the genetic interplay during embryonic CPC development remain challenging to reproduce in culture, in terms of efficiency, numbers, and translational potential. Recent advances in biomaterials to mimic the native cardiac microenvironment have shown promise to influence CPC regenerative functions, while being capable of integrating with host tissue. This review highlights recent developments and limitations in the generation and use of CPCs from stem cells, and the trends that influence the direction of research to promote better application of CPCs.
Collapse
Affiliation(s)
- Sara Barreto
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | | | - Teresa Schiatti
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Ying Yang
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Vinoj George
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| |
Collapse
|
35
|
Paul S, Zhang X, He JQ. Homeobox gene Meis1 modulates cardiovascular regeneration. Semin Cell Dev Biol 2019; 100:52-61. [PMID: 31623926 DOI: 10.1016/j.semcdb.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022]
Abstract
Regeneration of cardiomyocytes, endothelial cells and vascular smooth muscle cells (three major lineages of cardiac tissues) following myocardial infarction is the critical step to recover the function of the damaged heart. Myeloid ecotropic viral integration site 1 (Meis1) was first discovered in leukemic mice in 1995 and its biological function has been extensively studied in leukemia, hematopoiesis, the embryonic pattering of body axis, eye development and various genetic diseases, such as restless leg syndrome. It was found that Meis1 is highly associated with Hox genes and their cofactors to exert its regulatory effects on multiple intracellular signaling pathways. Recently with the advent of bioinformatics, biochemical methods and advanced genetic engineering tools, new function of Meis1 has been found to be involved in the cell cycle regulation of cardiomyocytes and endothelial cells. For example, inhibition of Meis1 expression increases the proliferative capacity of neonatal mouse cardiomyocytes, whereas overexpression of Meis1 results in the reduction in the length of cardiomyocyte proliferative window. Interestingly, downregulation of one of the circular RNAs, which acts downstream of Meis1 in the cardiomyocytes, promotes angiogenesis and restores the myocardial blood supply, thus reinforcing better regeneration of the damaged heart. It appears that Meis1 may play double roles in modulating proliferation and regeneration of cardiomyocytes and endothelial cells post-myocardial infarction. In this review, we propose to summarize the major findings of Meis1 in modulating fetal development and adult abnormalities, especially focusing on the recent discoveries of Meis1 in controlling the fate of cardiomyocytes and endothelial cells.
Collapse
Affiliation(s)
- Swagatika Paul
- Department of Biomedical Sciences & Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xiaonan Zhang
- Beijing Yulong Shengshi Biotechnology, Haidian District, Beijing, 100085, China
| | - Jia-Qiang He
- Department of Biomedical Sciences & Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
36
|
Song SY, Yoo J, Go S, Hong J, Sohn HS, Lee JR, Kang M, Jeong GJ, Ryu S, Kim SHL, Hwang NS, Char K, Kim BS. Cardiac-mimetic cell-culture system for direct cardiac reprogramming. Theranostics 2019; 9:6734-6744. [PMID: 31660065 PMCID: PMC6815967 DOI: 10.7150/thno.35574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Rationale: Cardiovascular diseases often cause substantial heart damage and even heart failure due to the limited regenerative capacity of adult cardiomyocytes. The direct cardiac reprogramming of fibroblasts could be a promising therapeutic option for these patients. Although exogenous transcriptional factors can induce direct cardiac reprogramming, the reprogramming efficiency is too low to be used clinically. Herein, we introduce a cardiac-mimetic cell-culture system that resembles the microenvironment in the heart and provides interactions with cardiomyocytes and electrical cues to the cultured fibroblasts for direct cardiac reprogramming. Methods: Nano-thin and nano-porous membranes and heart like electric stimulus were used in the cardiac-mimetic cell-culture system. The human neonatal dermal fibroblasts containing cardiac transcription factors were plated on the membrane and cultured with the murine cardiomyocyte in the presence of the electric stimulus. The reprogramming efficiency was evaluated by qRT-PCR and immunocytochemistry. Results: Nano-thin and nano-porous membranes in the culture system facilitated interactions between fibroblasts and cardiomyocytes in coculture. The cellular interactions and electric stimulation supplied by the culture system dramatically enhanced the cardiac reprogramming efficiency of cardiac-specific transcriptional factor-transfected fibroblasts. Conclusion: The cardiac-mimetic culture system may serve as an effective tool for producing a feasible number of reprogrammed cardiomyocytes from fibroblasts.
Collapse
|
37
|
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: 18] [Impact Index Per Article: 3.0] [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
|
38
|
Ensuring expression of four core cardiogenic transcription factors enhances cardiac reprogramming. Sci Rep 2019; 9:6362. [PMID: 31019236 PMCID: PMC6482135 DOI: 10.1038/s41598-019-42945-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/12/2019] [Indexed: 01/12/2023] Open
Abstract
Previous studies have shown that forced expression of core cardiogenic transcription factors can directly reprogram fibroblasts to induced cardiomyocyte-like cells (iCMs). This cardiac reprogramming approach suggests a potential strategy for cardiomyocyte regeneration. However, a major challenge of this approach remains the low conversion rate. Here, we showed that ensuring expression of four cardiogenic transcription factors (i.e. Gata4 (G), Hand2 (H), Mef2c (M), and Tbx5 (T)) in individual fibroblasts is an initial bottleneck for cardiac reprogramming. Following co-transduction of three or four retroviral vectors encoding individual cardiogenic transcription factors, only a minor subpopulation of cells indeed expressed all three (GMT) or four (GHMT) factors. By selectively analyzing subpopulations of cells expressing various combinations of reprogramming factors, we found that co-expression of GMT in individual fibroblasts is sufficient to induce sarcomeric proteins. However, only a small fraction of those cells expressing GMT were able to develop organized sarcomeric structures and contractility. In contrast, ensuring expression of GHMT markedly enhanced the development of contractile cardiac structures and functions in fibroblasts, although its incremental effect on sarcomeric protein induction was relatively small. Our findings provide new insights into the mechanistic basis of inefficient cardiac reprogramming and can help to devise efficient reprogramming strategies.
Collapse
|
39
|
Ebrahimi B. Cardiac progenitor reprogramming for heart regeneration. CELL REGENERATION 2019; 7:1-6. [PMID: 30671223 PMCID: PMC6326243 DOI: 10.1016/j.cr.2018.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/21/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
Abstract
Myocardial infarction leads to the loss of a huge number of cardiomyocytes and the reparatory response to this phenomenon is scar tissue formation, which impairs heart function. Direct reprogramming technology offers an alternative strategy for the generation of functional cardiomyocytes not only in vitro, but also in vivo in the site of injury. Results have demonstrated cardiac tissue regeneration and improvement in heart function after myocardial infarction following local injection of vectors encoding reprogramming transcription factors or miRNAs. This shows the great potential of cardiac reprogramming technology for heart regeneration. However, in addition to cardiomyocytes, other cell types, including endothelial cells and smooth muscle cells are also required to be generated in the damaged area in order to achieve complete cardiac tissue regeneration. To this aim induced proliferative/expandable cardiovascular progenitor cells (iCPCs) appear to be an appropriate cell source, which is capable of differentiation into three cardiovascular lineages both in vitro and in vivo. In this regard, this study goes over in vitro and in vivo cardiac reprogramming technology and specifically deals with cardiac progenitor reprogramming and its potential for heart regeneration.
Collapse
Key Words
- CASD, cell-activation and signaling-directed
- Cellular reprogramming
- ECs, endothelial cells
- FGF, fibroblast growth factor
- GMT, Gata4, Mef2c, and Tbx5
- Heart regeneration
- Myocardial infarction
- PI3K/AKT, phosphoinositol 3-kinase pathway
- SMCs, smooth muscle cells
- TF, transcription factor
- Transdifferentiation
- VEGF, vascular endothelial growth factor
- iCMs, induced cardiomyocytes
- iCPCs, induced cardiac progenitor cells
- iCSs, induced cardiospheres
- iPSC, induced pluripotent stem cell
- p38 MAPK, p38 mitogen-activated protein kinase pathway
Collapse
Affiliation(s)
- Behnam Ebrahimi
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
40
|
Werner JH, Rosenberg JH, Um JY, Moulton MJ, Agrawal DK. Molecular discoveries and treatment strategies by direct reprogramming in cardiac regeneration. Transl Res 2019; 203:73-87. [PMID: 30142308 PMCID: PMC6289806 DOI: 10.1016/j.trsl.2018.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/27/2018] [Accepted: 07/25/2018] [Indexed: 12/14/2022]
Abstract
Cardiac tissue has minimal endogenous regenerative capacity in response to injury. Treatment options are limited following tissue damage after events such as myocardial infarction. Current strategies are aimed primarily at injury prevention, but attention has been increasingly targeted toward the development of regenerative therapies. This review focuses on recent developments in the field of cardiac fibroblast reprogramming into induced cardiomyocytes. Early efforts to produce cardiac regeneration centered around induced pluripotent stem cells, but clinical translation has proved elusive. Currently, techniques are being developed to directly transdifferentiate cardiac fibroblasts into induced cardiomyocytes. Viral vector-driven expression of a combination of transcription factors including Gata4, Mef2c, and Tbx5 induced cardiomyocyte development in mice. Subsequent combinational modifications have extended these results to human cell lines and increased efficacy. The miRNAs including combinations of miR-1, miR-133, miR-208, and miR-499 can improve or independently drive regeneration of cardiomyocytes. Similar results could be obtained by combinations of small molecules with or without transcription factor or miRNA expression. The local tissue environment greatly impacts favorability for reprogramming. Modulation of signaling pathways, especially those mediated by VEGF and TGF-β, enhance differentiation to cardiomyocytes. Current reprogramming strategies are not ready for clinical application, but recent breakthroughs promise regenerative cardiac therapies in the near future.
Collapse
Affiliation(s)
- John H Werner
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - John H Rosenberg
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - John Y Um
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael J Moulton
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska.
| |
Collapse
|
41
|
Cell-Based Therapies for Cardiac Regeneration: A Comprehensive Review of Past and Ongoing Strategies. Int J Mol Sci 2018; 19:ijms19103194. [PMID: 30332812 PMCID: PMC6214096 DOI: 10.3390/ijms19103194] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
Despite considerable improvements in the treatment of cardiovascular diseases, heart failure (HF) still represents one of the leading causes of death worldwide. Poor prognosis is mostly due to the limited regenerative capacity of the adult human heart, which ultimately leads to left ventricular dysfunction. As a consequence, heart transplantation is virtually the only alternative for many patients. Therefore, novel regenerative approaches are extremely needed, and several attempts have been performed to improve HF patients’ clinical conditions by promoting the replacement of the lost cardiomyocytes and by activating cardiac repair. In particular, cell-based therapies have been shown to possess a great potential for cardiac regeneration. Different cell types have been extensively tested in clinical trials, demonstrating consistent safety results. However, heterogeneous efficacy data have been reported, probably because precise end-points still need to be clearly defined. Moreover, the principal mechanism responsible for these beneficial effects seems to be the paracrine release of antiapoptotic and immunomodulatory molecules from the injected cells. This review covers past and state-of-the-art strategies in cell-based heart regeneration, highlighting the advantages, challenges, and limitations of each approach.
Collapse
|
42
|
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
|
43
|
Yoo SY, Jeong SN, Kang JI, Lee SW. Chimeric Adeno-Associated Virus-Mediated Cardiovascular Reprogramming for Ischemic Heart Disease. ACS OMEGA 2018; 3:5918-5925. [PMID: 30023931 PMCID: PMC6044635 DOI: 10.1021/acsomega.8b00904] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 05/28/2023]
Abstract
Here, we demonstrated chimeric adeno-associated virus (chimeric AAV), AAV-DJ-mediated cardiovascular reprogramming strategy to generate new cardiomyocytes and limit collagen deposition in cardiac fibroblasts by inducing synergism of chimeric AAV-expressing Gata4, Mef2c, Tbx5 (AAV-GMT)-mediated heart reprogramming and chimeric AAV-expressing thymosin β4 (AAV-Tβ4)-mediated heart regeneration. AAV-GMT promoted a gradual increase in expression of cardiac-specific genes, including Actc1, Gja1, Myh6, Ryr2, and cTnT, with a gradual decrease in expression of a fibrosis-specific gene, procollagen type I and here AAV-Tβ4 help to induce GMT expression, providing a chimeric AAV-mediated therapeutic cell reprogramming strategy for ischemic heart diseases.
Collapse
Affiliation(s)
- So Young Yoo
- BIO-IT
Foundry Technology Institute, Pusan National
University, Busan 46241, Republic of Korea
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Su-Nam Jeong
- BIO-IT
Foundry Technology Institute, Pusan National
University, Busan 46241, Republic of Korea
| | - Jeong-In Kang
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Control
and Instrumentation Engineering, Korea Maritime
and Ocean University, Busan 49112, Republic of Korea
| | - Seung-Wuk Lee
- Bioengineering,
University of California, Berkeley, Lawrence
Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
44
|
Xu J, Lian W, Li L, Huang Z. Generation of induced cardiac progenitor cells via somatic reprogramming. Oncotarget 2018; 8:29442-29457. [PMID: 28199972 PMCID: PMC5438743 DOI: 10.18632/oncotarget.15272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
It has been demonstrated that cardiac progenitor cells (CPCs) represent a more effective cell-based therapy for treatment of myocardial infarction. Unfortunately, their therapeutic application is limited by low yield of cell harvesting, declining quality and quantity during the ageing process, and the need for highly invasive heart biopsy. Therefore, there is an emerging interest in generating CPC-like stem cells from somatic cells via somatic reprogramming. This novel approach would provide an unlimited source of stem cells with cardiac differentiation potential. Here we would firstly discuss the different types of CPC and their importance in stem cell therapy for treatment of myocardial infarction; secondly, the necessity of generating induced CPC from somatic cells via somatic reprogramming; and finally the current progress of somatic reprogramming in cardiac cells, especially induced CPC generation.
Collapse
Affiliation(s)
- Jianyong Xu
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Wei Lian
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Lingyun Li
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Zhong Huang
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| |
Collapse
|
45
|
Patel V, Singh VP, Pinnamaneni JP, Sanagasetti D, Olive J, Mathison M, Cooney A, Flores ER, Crystal RG, Yang J, Rosengart TK. p63 Silencing induces reprogramming of cardiac fibroblasts into cardiomyocyte-like cells. J Thorac Cardiovasc Surg 2018; 156:556-565.e1. [PMID: 29716728 DOI: 10.1016/j.jtcvs.2018.03.162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Reprogramming of fibroblasts into induced cardiomyocytes represents a potential new therapy for heart failure. We hypothesized that inactivation of p63, a p53 gene family member, may help overcome human cell resistance to reprogramming. METHODS p63 Knockout (-/-) and knockdown murine embryonic fibroblasts (MEFs), p63-/- adult murine cardiac fibroblasts, and human cardiac fibroblasts were assessed for cardiomyocyte-specific feature changes, with or without treatment by the cardiac transcription factors Hand2-Myocardin (HM). RESULTS Flow cytometry revealed that a significantly greater number of p63-/- MEFs expressed the cardiac-specific marker cardiac troponin T (cTnT) in culture compared with wild-type (WT) cells (38% ± 11% vs 0.9% ± 0.9%, P < .05). HM treatment of p63-/- MEFs increased cTnT expression to 74% ± 3% of cells but did not induce cTnT expression in wild-type murine embryonic fibroblasts. shRNA-mediated p63 knockdown likewise yielded a 20-fold increase in cTnT microRNA expression compared with untreated MEFs. Adult murine cardiac fibroblasts demonstrated a 200-fold increase in cTnT gene expression after inducible p63 knockout and expressed sarcomeric α-actinin as well as cTnT. These p63-/- adult cardiac fibroblasts exhibited calcium transients and electrically stimulated contractions when co-cultured with neonatal rat cardiomyocytes and treated with HM. Increased expression of cTnT and other marker genes was also observed in p63 knockdown human cardiac fibroblasts procured from patients undergoing procedures for heart failure. CONCLUSIONS Downregulation of p63 facilitates direct cardiac cellular reprogramming and may help overcome the resistance of human cells to reprogramming.
Collapse
Affiliation(s)
- Vivekkumar Patel
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Vivek P Singh
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | | | - Deepthi Sanagasetti
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Jacqueline Olive
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Austin Cooney
- Department of Pediatrics, The University of Texas at Austin, Dell Medical School, Austin, Tex
| | - Elsa R Flores
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Fla
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY
| | - Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
46
|
Abstract
Stem cell therapy is a promising approach to the treatment of ischemic heart disease via replenishing cell loss after myocardial infarction. Both preclinical studies and clinical trials have indicated that cardiac function improved consistently, but very modestly after cell-based therapy. This mainly attributed to low cell survival rate, engraftment and functional integration, which became the major challenges to regenerative medicine. In recent years, several new cell types have been developed to regenerate cardiomyocytes and novel delivery approaches helped to increase local cell retention. New strategies, such as cell pretreatment, gene-based therapy, tissue engineering, extracellular vesicles application and immunologic regulation, have surged and brought about improved cell survival and functional integration leading to better therapeutic effects after cell transplantation. In this review, we summarize these new strategies targeting at challenges of cardiac regenerative medicine and discuss recent evidences that may hint their effectiveness in the future clinical settings.
Collapse
|
47
|
Rosengart TK, Patel V, Sellke FW. Cardiac stem cell trials and the new world of cellular reprogramming: Time to move on. J Thorac Cardiovasc Surg 2017; 155:1642-1646. [PMID: 29397153 DOI: 10.1016/j.jtcvs.2017.11.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/09/2017] [Accepted: 11/16/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| | - Vivek Patel
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Brown Medical School, Providence, RI
| |
Collapse
|
48
|
Osakabe R, Suzuki T, Ieda M. [Heart repair using direct cardiac reprogramming]. Nihon Yakurigaku Zasshi 2017; 150:276-281. [PMID: 29225289 DOI: 10.1254/fpj.150.276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
49
|
Ingason AB, Goldstone AB, Paulsen MJ, Thakore AD, Truong VN, Edwards BB, Eskandari A, Bollig T, Steele AN, Woo YJ. Angiogenesis precedes cardiomyocyte migration in regenerating mammalian hearts. J Thorac Cardiovasc Surg 2017; 155:1118-1127.e1. [PMID: 29452461 DOI: 10.1016/j.jtcvs.2017.08.127] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Although the mammalian heart's ability to fully regenerate is debated, its potential to extensively repair itself is gaining support. We hypothesized that heart regeneration relies on rapid angiogenesis to support myocardial regrowth and sought to characterize the timeline for angiogenesis and cell proliferation in regeneration. METHODS One-day-old CD-1 mice (P1, N = 60) underwent apical resection or sham surgery. Hearts were explanted at serial time points from 0 to 30 days postresection and analyzed with immunohistochemistry to visualize vessel ingrowth and cardiomyocyte migration into the resected region. Proliferating cells were labeled with 5-ethynyl-2'-deoxyuridine injections 12 hours before explant. 5-Ethynyl-2'-deoxyuridine-positive cells were counted in both the apex and remote areas of the heart. Masson's trichrome was used to assess fibrosis. RESULTS By 30 days postresection, hearts regenerated with minimal fibrosis. Compared with sham surgery, apical resection stimulated a significant increase in proliferation of preexisting cardiomyocytes between 3 and 11 days after injury. Capillary migration into the apical thrombus was detected as early as 2 days postresection, with development of mature arteries by 5 days postresection. New vessels became perfused by 5 days postresection as evidenced by lectin injection. Vessel density and diameter significantly increased within the resected area over 21 days, and vessel ingrowth always preceded cardiomyocyte migration, with coalignment of most migrating cardiomyocytes with ingrowing vessels. CONCLUSIONS Endothelial cells migrate into the apical thrombus early after resection, develop into functional arteries, and precede cardiomyocyte ingrowth during mammalian heart regeneration. This endogenous neonatal response emphasizes the importance of expeditious angiogenesis required for neomyogenesis.
Collapse
Affiliation(s)
- Arnar B Ingason
- Department of Medicine, University of Iceland, Reykjavík, Iceland; Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Akshara D Thakore
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Vi N Truong
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Bryan B Edwards
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Tanner Bollig
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
50
|
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: 146] [Impact Index Per Article: 18.3] [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
|