1
|
Almeida M, Inácio JM, Vital CM, Rodrigues MR, Araújo BC, Belo JA. Cell Reprogramming, Transdifferentiation, and Dedifferentiation Approaches for Heart Repair. Int J Mol Sci 2025; 26:3063. [PMID: 40243729 PMCID: PMC11988544 DOI: 10.3390/ijms26073063] [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: 03/01/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death globally, with myocardial infarction (MI) being a major contributor. The current therapeutic approaches are limited in effectively regenerating damaged cardiac tissue. Up-to-date strategies for heart regeneration/reconstitution aim at cardiac remodeling through repairing the damaged tissue with an external cell source or by stimulating the existing cells to proliferate and repopulate the compromised area. Cell reprogramming is addressed to this challenge as a promising solution, converting fibroblasts and other cell types into functional cardiomyocytes, either by reverting cells to a pluripotent state or by directly switching cell lineage. Several strategies such as gene editing and the application of miRNA and small molecules have been explored for their potential to enhance cardiac regeneration. Those strategies take advantage of cell plasticity by introducing reprogramming factors that regress cell maturity in vitro, allowing for their later differentiation and thus endorsing cell transplantation, or promote in situ cell proliferation, leveraged by scaffolds embedded with pro-regenerative factors promoting efficient heart restoration. Despite notable advancements, important challenges persist, including low reprogramming efficiency, cell maturation limitations, and safety concerns in clinical applications. Nonetheless, integrating these innovative approaches offers a promising alternative for restoring cardiac function and reducing the dependency on full heart transplants.
Collapse
Affiliation(s)
| | - José M. Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| | | | | | | | - José A. Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| |
Collapse
|
2
|
Santos F, Correia M, Dias R, Bola B, Noberini R, Ferreira RS, Trigo D, Domingues P, Teixeira J, Bonaldi T, Oliveira PJ, Bär C, de Jesus BB, Nóbrega‐Pereira S. Age-associated metabolic and epigenetic barriers during direct reprogramming of mouse fibroblasts into induced cardiomyocytes. Aging Cell 2025; 24:e14371. [PMID: 39540462 PMCID: PMC11822649 DOI: 10.1111/acel.14371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
Heart disease is the leading cause of mortality in developed countries, and novel regenerative procedures are warranted. Direct cardiac conversion (DCC) of adult fibroblasts can create induced cardiomyocytes (iCMs) for gene and cell-based heart therapy, and in addition to holding great promise, still lacks effectiveness as metabolic and age-associated barriers remain elusive. Here, by employing MGT (Mef2c, Gata4, Tbx5) transduction of mouse embryonic fibroblasts (MEFs) and adult (dermal and cardiac) fibroblasts from animals of different ages, we provide evidence that the direct reprogramming of fibroblasts into iCMs decreases with age. Analyses of histone posttranslational modifications and ChIP-qPCR revealed age-dependent alterations in the epigenetic landscape of DCC. Moreover, DCC is accompanied by profound mitochondrial metabolic adaptations, including a lower abundance of anabolic metabolites, network remodeling, and reliance on mitochondrial respiration. In vitro metabolic modulation and dietary manipulation in vivo improve DCC efficiency and are accompanied by significant alterations in histone marks and mitochondrial homeostasis. Importantly, adult-derived iCMs exhibit increased accumulation of oxidative stress in the mitochondria and activation of mitophagy or dietary lipids; they improve DCC and revert mitochondrial oxidative damage. Our study provides evidence that metaboloepigenetics plays a direct role in cell fate transitions driving DCC, highlighting the potential use of metabolic modulation to improve cardiac regenerative strategies.
Collapse
Affiliation(s)
- Francisco Santos
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Magda Correia
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Rafaela Dias
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Bárbara Bola
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Roberta Noberini
- Department of Experimental OncologyEuropean Institute of Oncology (IEO), IRCCSMilanItaly
| | - Rita S. Ferreira
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Diogo Trigo
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Pedro Domingues
- Mass Spectrometry Center, Department of ChemistryUniversity of AveiroAveiroPortugal
- LAQV/REQUIMTEUniversity of AveiroAveiroPortugal
| | - José Teixeira
- CNC‐UC, Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB, Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCantanhedePortugal
| | - Tiziana Bonaldi
- Department of Experimental OncologyEuropean Institute of Oncology (IEO), IRCCSMilanItaly
- Department of Oncology and Hematology‐OncologyUniversity of MilanoMilanItaly
| | - Paulo J. Oliveira
- CNC‐UC, Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB, Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCantanhedePortugal
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS)Hannover Medical School (MHH)HannoverGermany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM)HannoverGermany
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Sandrina Nóbrega‐Pereira
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| |
Collapse
|
3
|
Piven OO, Vaičiulevičiūtė R, Bernotiene E, Dobrzyn P. Cardiomyocyte engineering: The meeting point of transcription factors, signaling networks, metabolism and function. Acta Physiol (Oxf) 2025; 241:e14271. [PMID: 39801134 DOI: 10.1111/apha.14271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/22/2024] [Accepted: 01/01/2025] [Indexed: 05/02/2025]
Abstract
Direct cardiac reprogramming or transdifferentiation is a relatively new and promising area in regenerative therapy, cardiovascular disease modeling, and drug discovery. Effective reprogramming of fibroblasts is limited by their plasticity, that is, their ability to reprogram, and depends on solving several levels of tasks: inducing cardiomyocyte-like cells and obtaining functionally and metabolically mature cardiomyocytes. Currently, in addition to the use of more classical approaches such as overexpression of exogenous transcription factors, activation of endogenous cardiac transcription factors via controlled nucleases, such as CRISPR, represents another interesting way to obtain cardiomyocytes. Therefore, special attention is given to the potential of synthetic biology, in particular the CRISPR system, for the targeted conversion of only certain subpopulations of fibroblasts into cardiomyocytes. However, obtaining functionally and metabolically mature cardiomyocytes remains a challenge despite the range of recently developed approaches. In this review, we summarized current knowledge on the function and diversity of human cardiac fibroblasts and alternative cell sources for in vitro human cardiomyocyte models. We examined in detail the transcription factors that initiate cardiomyogenic reprogramming and their interactions. Additionally, we critically analyzed the strategies used for the metabolic and physiological maturation of induced cardiomyocytes.
Collapse
Affiliation(s)
- Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Raminta Vaičiulevičiūtė
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Faculty of Fundamental Sciences, VilniusTech University, Vilnius, Lithuania
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
4
|
Yang J. Emerging Insights into Sall4's Role in Cardiac Regenerative Medicine. Cells 2025; 14:154. [PMID: 39936946 PMCID: PMC11817359 DOI: 10.3390/cells14030154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Sall4 as a pivotal transcription factor has been extensively studied across diverse biological processes, including stem cell biology, embryonic development, hematopoiesis, tissue stem/progenitor maintenance, and the progression of various cancers. Recent research highlights Sall4's emerging roles in modulating cardiac progenitors and cellular reprogramming, linking its functions to early heart development and regenerative medicine. These findings provide new insights into the critical functions of Sall4 in cardiobiology. This review explores Sall4's complex molecular mechanisms and their implications for advancing cardiac regenerative medicine.
Collapse
Affiliation(s)
- Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
5
|
Ambroise R, Takasugi P, Liu J, Qian L. Direct Cardiac Reprogramming in the Age of Computational Biology. J Cardiovasc Dev Dis 2024; 11:273. [PMID: 39330331 PMCID: PMC11432431 DOI: 10.3390/jcdd11090273] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Heart disease continues to be one of the most fatal conditions worldwide. This is in part due to the maladaptive remodeling process by which ischemic cardiac tissue is replaced with a fibrotic scar. Direct cardiac reprogramming presents a unique solution for restoring injured cardiac tissue through the direct conversion of fibroblasts into induced cardiomyocytes, bypassing the transition through a pluripotent state. Since its inception in 2010, direct cardiac reprogramming using the transcription factors Gata4, Mef2c, and Tbx5 has revolutionized the field of cardiac regenerative medicine. Just over a decade later, the field has rapidly evolved through the expansion of identified molecular and genetic factors that can be used to optimize reprogramming efficiency. The integration of computational tools into the study of direct cardiac reprogramming has been critical to this progress. Advancements in transcriptomics, epigenetics, proteomics, genome editing, and machine learning have not only enhanced our understanding of the underlying mechanisms driving this cell fate transition, but have also driven innovations that push direct cardiac reprogramming closer to clinical application. This review article explores how these computational advancements have impacted and continue to shape the field of direct cardiac reprogramming.
Collapse
Affiliation(s)
- Rachelle Ambroise
- Department of Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- 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
| | - Paige Takasugi
- 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
| | - Jiandong Liu
- 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
| | - 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
| |
Collapse
|
6
|
Liu L, Lei I, Tian S, Gao W, Guo Y, Li Z, Sabry Z, Tang P, Chen YE, Wang Z. 14-3-3 binding motif phosphorylation disrupts Hdac4-organized condensates to stimulate cardiac reprogramming. Cell Rep 2024; 43:114054. [PMID: 38578832 PMCID: PMC11081035 DOI: 10.1016/j.celrep.2024.114054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/15/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
Cell fate conversion is associated with extensive post-translational modifications (PTMs) and architectural changes of sub-organelles, yet how these events are interconnected remains unknown. We report here the identification of a phosphorylation code in 14-3-3 binding motifs (PC14-3-3) that greatly stimulates induced cardiomyocyte (iCM) formation from fibroblasts. PC14-3-3 is identified in pivotal functional proteins for iCM reprogramming, including transcription factors and chromatin modifiers. Akt1 kinase and protein phosphatase 2A are the key writer and key eraser of the PC14-3-3 code, respectively. PC14-3-3 activation induces iCM formation with the presence of only Tbx5. In contrast, PC14-3-3 inhibition by mutagenesis or inhibitor-mediated code removal abolishes reprogramming. We discover that key PC14-3-3-embedded factors, such as histone deacetylase 4 (Hdac4), Mef2c, and Foxo1, form Hdac4-organized inhibitory nuclear condensates. PC14-3-3 activation disrupts Hdac4 condensates to promote cardiac gene expression. Our study suggests that sub-organelle dynamics regulated by a PTM code could be a general mechanism for stimulating cell reprogramming.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenbin Gao
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Yijing Guo
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhaokai Li
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Ziad Sabry
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Paul Tang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
García-Loredo JA, Santoyo-Suarez MG, Rodríguez-Nuñez O, Benitez Chao DF, Garza-Treviño EN, Zapata-Morin PA, Padilla-Rivas GR, Islas JF. Is the Cis-Element CACCC-Box a Master Regulatory Element during Cardiovascular Disease? A Bioinformatics Approach from the Perspective of the Krüppel-like Family of Transcription Factors. Life (Basel) 2024; 14:493. [PMID: 38672763 PMCID: PMC11051458 DOI: 10.3390/life14040493] [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: 03/14/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The CACCC-box motif emerges as a pivotal cis-regulatory element implicated in diverse developmental processes and diseases, particularly cardiovascular diseases (CVDs). This study centers on the intricate interplay between the CACCC-box and its binding proteins such as: the Krüppel-Like Family (KLF) of transcription factors as primary effectors in the context of CVDs. Our analysis was through a bioinformatics approach, which revealed significant transcriptional activity among KLF subgroup 2, exhibiting the highest number of interactions focusing on the established roles: pluripotency, cancer, and cardiovascular development and diseases. Our analysis reveals KLF's interactions with GATA4, MEF2C, NKX2.5 and other ~90 potential genes that participate in the regulation of the hypertrophic environment (or CVDs' Environment). Also, the GO analysis showed that genes containing the motif CACCC were enriched for multiple CVDs; in combination with STRING analysis, these results pointed to a link between KLFs and these diseases. The analysis further identifies other potential CACCC-box binding factors, such as SP family members, WT1, VEZF1, and -SALL4, which are implicated in cardiac contraction, remodeling, and inflammation processes.
Collapse
Affiliation(s)
- Juan Andrés García-Loredo
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
- Laboratorio de Micología y Fitopatología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo León, Mexico;
| | - Michelle G. Santoyo-Suarez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Oscar Rodríguez-Nuñez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Diego Francisco Benitez Chao
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Elsa N. Garza-Treviño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Patricio Adrián Zapata-Morin
- Laboratorio de Micología y Fitopatología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo León, Mexico;
| | - Gerardo R. Padilla-Rivas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| | - Jose Francisco Islas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Nuevo León, Mexico; (J.A.G.-L.); (M.G.S.-S.); (O.R.-N.); (D.F.B.C.); (E.N.G.-T.); (G.R.P.-R.)
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Liu L, Lei I, Tian S, Gao W, Guo Y, Li Z, Sabry Z, Tang P, Chen YE, Wang Z. 14-3-3 binding motif phosphorylation disrupts Hdac4 organized condensates to stimulate cardiac reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567913. [PMID: 38045244 PMCID: PMC10690191 DOI: 10.1101/2023.11.20.567913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Cell fate conversion is associated with extensive epigenetic and post translational modifications (PTMs) and architectural changes of sub-organelles and organelles, yet how these events are interconnected remains unknown. We report here the identification of a phosphorylation code in 14-3-3 binding motifs (PC14-3-3) that greatly stimulates induced cardiomyocyte (iCM) formation from fibroblasts. PC14-3-3 was identified in pivotal functional proteins for iCM reprogramming, including transcription factors and epigenetic factors. Akt1 kinase and PP2A phosphatase were a key writer and eraser of the PC14-3-3 code, respectively. PC14-3-3 activation induces iCM formation with the presence of only Tbx5. In contrast, PC14-3-3 inhibition by mutagenesis or inhibitor-mediated code removal abolished reprogramming. We discovered that key PC14-3-3 embedded factors, such as Hdac4, Mef2c, Nrip1, and Foxo1, formed Hdac4 organized inhibitory nuclear condensates. Notably, PC14-3-3 activation disrupted Hdac4 condensates to promote cardiac gene expression. Our study suggests that sub-organelle dynamics regulated by a post-translational modification code could be a general mechanism for stimulating cell reprogramming and organ regeneration. Highlights A PC14-3-3 (phosphorylation code in 14-3-3 binding motifs) is identified in pivotal functional proteins, such as HDAC4 and Mef2c, that stimulates iCM formation.Akt1 kinase and PP2A phosphatase are a key writer and a key eraser of the PC14-3-3 code, respectively, and PC14-3-3 code activation can replace Mef2c and Gata4 in cardiac reprogramming.PC14-3-3 activation disrupts Hdac4 organized condensates which results in releasing multiple 14-3-3 motif embedded proteins from the condensates to stimulate cardiac reprogramming.Sub-organelle dynamics and function regulated by a post-translational modification code could be a general mechanism in stimulating cell reprogramming and organ regeneration. Graphic abstract
Collapse
|
12
|
Su F, Xiao R, Chen R, Yang T, Wang D, Xu X, Hou X, Guan Q, Feng M. WIPF1 promotes gastric cancer progression by regulating PI3K/Akt signaling in a myocardin-dependent manner. iScience 2023; 26:108273. [PMID: 38026208 PMCID: PMC10654612 DOI: 10.1016/j.isci.2023.108273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/04/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Wiskott-Aldrich syndrome protein-interacting protein family member 1 (WIPF1) is associated with malignant tumor progression. However, molecular links between WIPF1 and gastric cancer (GC) remain elusive. The expression of WIPF1 was detected in GC tissues and cells. WIPF1 was overexpressed in GC tissues and cells and high expression of WIPF1 was an independent risk factor for a poor prognosis in patients with GC. Further experiments indicated that WIPF1 promoted the proliferation, invasion, and migration of GC cells in vivo and in vitro. WIPF1-regulated genes were closely related to cell proliferation and migration in GC, and silencing WIPF1 significantly repressed PI3K/AKT signaling pathway activation. WIPF1 was activated by myocardin (MYOCD) translation. Rescue experiments confirmed that MYOCD promotes the proliferation, invasion, and migration of GC cells in a WIPF1-dependent manner and activates the PI3K/AKT signaling pathway. MYOCD may transactivate WIPF1 and facilitate GC cell growth and metastasis by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Fei Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ruowen Xiao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Rui Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Tianning Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Danwen Wang
- Center for Clinical Medicine of Peritoneal Cancer of Wuhan, Wuhan, Hubei 430060, P.R. China
- Clinical Cancer Study Center of Hubei Province, Wuhan, Hubei 430060, P.R. China
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xinni Xu
- Scientific Development and Planing Department, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiaoming Hou
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Quanlin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Maohui Feng
- Center for Clinical Medicine of Peritoneal Cancer of Wuhan, Wuhan, Hubei 430060, P.R. China
- Clinical Cancer Study Center of Hubei Province, Wuhan, Hubei 430060, P.R. China
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
13
|
Xie Y, Wang Q, Yang Y, Near D, Wang H, Colon M, Nguyen C, Slattery C, Keepers B, Farber G, Wang TW, Lee SH, Shih YYI, Liu J, Qian L. Translational landscape of direct cardiac reprogramming reveals a role of Ybx1 in repressing cardiac fate acquisition. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1060-1077. [PMID: 38524149 PMCID: PMC10959502 DOI: 10.1038/s44161-023-00344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/06/2023] [Indexed: 03/26/2024]
Abstract
Direct reprogramming of fibroblasts into induced cardiomyocytes holds great promise for heart regeneration. Although considerable progress has been made in understanding the transcriptional and epigenetic mechanisms of iCM reprogramming, its translational regulation remains largely unexplored. Here, we characterized the translational landscape of iCM reprogramming through integrative ribosome and transcriptomic profiling, and found extensive translatome repatterning during this process. Loss of function screening for translational regulators uncovered Ybx1 as a critical barrier to iCM induction. In a mouse model of myocardial infarction, removing Ybx1 enhanced in vivo reprogramming, resulting in improved heart function and reduced scar size. Mechanistically, Ybx1 depletion de-repressed the translation of its direct targets SRF and Baf60c, both of which mediated the effect of Ybx1 depletion on iCM generation. Furthermore, removal of Ybx1 allowed single factor Tbx5-mediated iCM conversion. In summary, this study revealed a new layer of regulatory mechanism that controls cardiac reprogramming at the translational level.
Collapse
Affiliation(s)
- Yifang Xie
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Qiaozi Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Yuchen Yang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - David Near
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Haofei Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Marazzano Colon
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Christopher Nguyen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Conor Slattery
- EIRNA Bio Ltd, BioInnovation Centre, Food Science and Technology Building, College Road, Cork, Ireland, T12 DP07
| | - Benjamin Keepers
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599
| | - Gregory Farber
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Tzu-Wen Wang
- Departments of Neurology, University of North Carolina, Chapel Hill, NC 27599
| | - Sung-Ho Lee
- Departments of Neurology, University of North Carolina, Chapel Hill, NC 27599
| | - Yen-Yu Ian Shih
- Departments of Neurology, University of North Carolina, Chapel Hill, NC 27599
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
14
|
Romero-Tejeda M, Fonoudi H, Weddle CJ, DeKeyser JM, Lenny B, Fetterman KA, Magdy T, Sapkota Y, Epting CL, Burridge PW. A novel transcription factor combination for direct reprogramming to a spontaneously contracting human cardiomyocyte-like state. J Mol Cell Cardiol 2023; 182:30-43. [PMID: 37421991 PMCID: PMC10495191 DOI: 10.1016/j.yjmcc.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/06/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
The reprogramming of somatic cells to a spontaneously contracting cardiomyocyte-like state using defined transcription factors has proven successful in mouse fibroblasts. However, this process has been less successful in human cells, thus limiting the potential clinical applicability of this technology in regenerative medicine. We hypothesized that this issue is due to a lack of cross-species concordance between the required transcription factor combinations for mouse and human cells. To address this issue, we identified novel transcription factor candidates to induce cell conversion between human fibroblasts and cardiomyocytes, using the network-based algorithm Mogrify. We developed an automated, high-throughput method for screening transcription factor, small molecule, and growth factor combinations, utilizing acoustic liquid handling and high-content kinetic imaging cytometry. Using this high-throughput platform, we screened the effect of 4960 unique transcription factor combinations on direct conversion of 24 patient-specific primary human cardiac fibroblast samples to cardiomyocytes. Our screen revealed the combination of MYOCD, SMAD6, and TBX20 (MST) as the most successful direct reprogramming combination, which consistently produced up to 40% TNNT2+ cells in just 25 days. Addition of FGF2 and XAV939 to the MST cocktail resulted in reprogrammed cells with spontaneous contraction and cardiomyocyte-like calcium transients. Gene expression profiling of the reprogrammed cells also revealed the expression of cardiomyocyte associated genes. Together, these findings indicate that cardiac direct reprogramming in human cells can be achieved at similar levels to those attained in mouse fibroblasts. This progress represents a step forward towards the clinical application of the cardiac direct reprogramming approach.
Collapse
Affiliation(s)
- Marisol Romero-Tejeda
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Carly J Weddle
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jean-Marc DeKeyser
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Brian Lenny
- Department of Epidemiology and Cancer Control, St. Jude Children's Hospital, Memphis, TN, USA
| | - K Ashley Fetterman
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Hospital, Memphis, TN, USA
| | - Conrad L Epting
- Departments of Pediatrics and Pathology, Northwestern University and Ann & Robert H.Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
15
|
Romero-Tejeda M, Fonoudi H, Weddle CJ, DeKeyser JM, Lenny B, Fetterman KA, Magdy T, Sapkota Y, Epting C, Burridge PW. A Novel Transcription Factor Combination for Direct Reprogramming to a Spontaneously Contracting Human Cardiomyocyte-like State. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532629. [PMID: 36993577 PMCID: PMC10055062 DOI: 10.1101/2023.03.14.532629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
UNLABELLED The reprogramming of somatic cells to a spontaneously contracting cardiomyocyte-like state using defined transcription factors has proven successful in mouse fibroblasts. However, this process has been less successful in human cells, thus limiting the potential clinical applicability of this technology in regenerative medicine. We hypothesized that this issue is due to a lack of cross-species concordance between the required transcription factor combinations for mouse and human cells. To address this issue, we identified novel transcription factor candidates to induce cell conversion between human fibroblasts and cardiomyocytes, using the network-based algorithm Mogrify. We developed an automated, high-throughput method for screening transcription factor, small molecule, and growth factor combinations, utilizing acoustic liquid handling and high-content kinetic imaging cytometry. Using this high-throughput platform, we screened the effect of 4,960 unique transcription factor combinations on direct conversion of 24 patient-specific primary human cardiac fibroblast samples to cardiomyocytes. Our screen revealed the combination of MYOCD , SMAD6 , and TBX20 (MST) as the most successful direct reprogramming combination, which consistently produced up to 40% TNNT2 + cells in just 25 days. Addition of FGF2 and XAV939 to the MST cocktail resulted in reprogrammed cells with spontaneous contraction and cardiomyocyte-like calcium transients. Gene expression profiling of the reprogrammed cells also revealed the expression of cardiomyocyte associated genes. Together, these findings indicate that cardiac direct reprogramming in human cells can be achieved at similar levels to those attained in mouse fibroblasts. This progress represents a step forward towards the clinical application of the cardiac direct reprogramming approach. HIGHLIGHTS Using network-based algorithm Mogrify, acoustic liquid handling, and high-content kinetic imaging cytometry we screened the effect of 4,960 unique transcription factor combinations. Using 24 patient-specific human fibroblast samples we identified the combination of MYOCD , SMAD6 , and TBX20 (MST) as the most successful direct reprogramming combination. MST cocktail results in reprogrammed cells with spontaneous contraction, cardiomyocyte-like calcium transients, and expression of cardiomyocyte associated genes.
Collapse
|
16
|
Cao Y, Dong Z, Yang D, Ma X, Wang X. LSD1 regulates the expressions of core cardiogenic transcription factors and cardiac genes in oxygen and glucose deprivation injured mice fibroblasts in vitro. Exp Cell Res 2022; 418:113228. [PMID: 35688282 DOI: 10.1016/j.yexcr.2022.113228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
Cardiac reprogramming has emerged as a novel therapeutic approach to regenerating the damaged heart by directly converting endogenous cardiac fibroblasts (CFs) into induced cardiomyocytes (iCMs). Cardiac reprogramming requires the activation of the cardiogenic transcriptional program in concert with the repression of the fibroblastic transcriptional program. Lysine-specific demethylase 1 (LSD1) plays an instrumental role in many physiological processes such as cell growth, differentiation and metabolism. The epigenetic modifications of histones are essential for the accurate expression of genes in cardiomyocytes and the normal functioning of the heart. However, the effect of LSD1 in regulating the cardiogenic transcriptional program under myocardial ischemia/reperfusion (I/R) injury remains unclear. Thus, mice I/R injury was induced by 4 and 24 h reperfusion after 1-h occlusion of the left anterior descending coronary artery. The primary CFs and CMs were exposed under oxygen and glucose deprivation (OGD) to mimic I/R injury. The expression of LSD1 significantly decreased in I/R injured heart tissue and OGD-injured primary CFs and CM, and methylated histone presented a notable increase in OGD-injured primary CFs. Overexpression of LSD1 inhibited the injury of primary CFs induced by OGD, but showed limited inhibition on injured primary CMs. Under the OGD condition, LSD1 overexpression significantly increased cell viability, decreased cell apoptosis and reactive oxygen species (ROS) production of primary CFs. The expression of core cardiogenic transcription factors and cardiac genes were significantly decreased in OGD injured primary CFs, whereas LSD1 overexpression reversed the decrease of transcription factors and cardiac genes under the OGD condition. In conclusion, the overexpression of LSD1 has a protective role in I/R injury by inhibiting the histone methylation of primary CFs and regulates the expressions of core cardiogenic transcription factors and cardiac genes, which can prove to be a potential approach for direct cardiac reprogramming.
Collapse
Affiliation(s)
- Yiqiu Cao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China; Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, People's Republic of China; Department of Cardiovascular Surgery, People's Liberation Army General Hospital of Southern Theater Command, Guangzhou, 510170, People's Republic of China
| | - Zhu Dong
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, People's Republic of China
| | - Dongpeng Yang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China; Department of Cardiovascular Surgery, Guangzhou Red Cross Hospital, Jinan University, 510235, People's Republic of China
| | - Ximiao Ma
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China; Department of Cardiothoracic Surgery, Haikou People's Hospital, Haikou, 570208, People's Republic of China
| | - Xiaowu Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, People's Republic of China; Department of Cardiovascular Surgery, People's Liberation Army General Hospital of Southern Theater Command, Guangzhou, 510170, People's Republic of China.
| |
Collapse
|
17
|
Metabolic Determinants in Cardiomyocyte Function and Heart Regenerative Strategies. Metabolites 2022; 12:metabo12060500. [PMID: 35736435 PMCID: PMC9227827 DOI: 10.3390/metabo12060500] [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: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart disease is the leading cause of mortality in developed countries. The associated pathology is characterized by a loss of cardiomyocytes that leads, eventually, to heart failure. In this context, several cardiac regenerative strategies have been developed, but they still lack clinical effectiveness. The mammalian neonatal heart is capable of substantial regeneration following injury, but this capacity is lost at postnatal stages when cardiomyocytes become terminally differentiated and transit to the fetal metabolic switch. Cardiomyocytes are metabolically versatile cells capable of using an array of fuel sources, and the metabolism of cardiomyocytes suffers extended reprogramming after injury. Apart from energetic sources, metabolites are emerging regulators of epigenetic programs driving cell pluripotency and differentiation. Thus, understanding the metabolic determinants that regulate cardiomyocyte maturation and function is key for unlocking future metabolic interventions for cardiac regeneration. In this review, we will discuss the emerging role of metabolism and nutrient signaling in cardiomyocyte function and repair, as well as whether exploiting this axis could potentiate current cellular regenerative strategies for the mammalian heart.
Collapse
|
18
|
Afouda BA. Towards Understanding the Gene-Specific Roles of GATA Factors in Heart Development: Does GATA4 Lead the Way? Int J Mol Sci 2022; 23:5255. [PMID: 35563646 PMCID: PMC9099915 DOI: 10.3390/ijms23095255] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023] Open
Abstract
Transcription factors play crucial roles in the regulation of heart induction, formation, growth and morphogenesis. Zinc finger GATA transcription factors are among the critical regulators of these processes. GATA4, 5 and 6 genes are expressed in a partially overlapping manner in developing hearts, and GATA4 and 6 continue their expression in adult cardiac myocytes. Using different experimental models, GATA4, 5 and 6 were shown to work together not only to ensure specification of cardiac cells but also during subsequent heart development. The complex involvement of these related gene family members in those processes is demonstrated through the redundancy among them and crossregulation of each other. Our recent identification at the genome-wide level of genes specifically regulated by each of the three family members and our earlier discovery that gata4 and gata6 function upstream, while gata5 functions downstream of noncanonical Wnt signalling during cardiac differentiation, clearly demonstrate the functional differences among the cardiogenic GATA factors. Such suspected functional differences are worth exploring more widely. It appears that in the past few years, significant advances have indeed been made in providing a deeper understanding of the mechanisms by which each of these molecules function during heart development. In this review, I will therefore discuss current evidence of the role of individual cardiogenic GATA factors in the process of heart development and emphasize the emerging central role of GATA4.
Collapse
Affiliation(s)
- Boni A Afouda
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| |
Collapse
|
19
|
Xie Y, Liu J, Qian L. Direct cardiac reprogramming comes of age: Recent advance and remaining challenges. Semin Cell Dev Biol 2022; 122:37-43. [PMID: 34304993 PMCID: PMC8782931 DOI: 10.1016/j.semcdb.2021.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 02/03/2023]
Abstract
The adult human heart has limited regenerative capacity. As such, the massive cardiomyocyte loss due to myocardial infarction leads to scar formation and adverse cardiac remodeling, which ultimately results in chronic heart failure. Direct cardiac reprogramming that converts cardiac fibroblast into functional cardiomyocyte-like cells (also called iCMs) holds great promise for heart regeneration. Cardiac reprogramming has been achieved both in vitro and in vivo by using a variety of cocktails that comprise transcription factors, microRNAs, or small molecules. During the past several years, great progress has been made in improving reprogramming efficiency and understanding the underlying molecular mechanisms. Here, we summarize the direct cardiac reprogramming methods, review the current advances in understanding the molecular mechanisms of cardiac reprogramming, and highlight the novel insights gained from single-cell omics studies. Finally, we discuss the remaining challenges and future directions for the field.
Collapse
|
20
|
Garry GA, Bassel-Duby R, Olson EN. Direct reprogramming as a route to cardiac repair. Semin Cell Dev Biol 2022; 122:3-13. [PMID: 34246567 PMCID: PMC8738780 DOI: 10.1016/j.semcdb.2021.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/14/2021] [Indexed: 02/03/2023]
Abstract
Ischemic heart disease is the leading cause of morbidity, mortality, and healthcare expenditure worldwide due to an inability of the heart to regenerate following injury. Thus, novel heart failure therapies aimed at promoting cardiomyocyte regeneration are desperately needed. In recent years, direct reprogramming of resident cardiac fibroblasts to induced cardiac-like myocytes (iCMs) has emerged as a promising therapeutic strategy to repurpose the fibrotic response of the injured heart toward a functional myocardium. Direct cardiac reprogramming was initially achieved through the overexpression of the transcription factors (TFs) Gata4, Mef2c, and Tbx5 (GMT). However, this combination of TFs and other subsequent cocktails demonstrated limited success in reprogramming adult human and mouse fibroblasts, constraining the clinical translation of this therapy. Over the past decade, significant effort has been dedicated to optimizing reprogramming cocktails comprised of cardiac TFs, epigenetic factors, microRNAs, or small molecules to yield efficient cardiac cell fate conversion. Yet, efficient reprogramming of adult human fibroblasts remains a significant challenge. Underlying mechanisms identified to accelerate this process have been centered on epigenetic remodeling at cardiac gene regulatory regions. Further studies to achieve a refined understanding and directed means of overcoming epigenetic barriers are merited to more rapidly translate these promising therapies to the clinic.
Collapse
Affiliation(s)
- Glynnis A. Garry
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Eric N. Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX,Correspondence: Eric N. Olson, Ph.D. 5323 Harry Hines Boulevard, Dallas, Texas, 75390-9148, Tel: 214-648-1187,
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Khazaei S, Soleimani M, Tafti SHA, Aghdam RM, Hojati Z. Improvement of Heart Function After Transplantation of Encapsulated Stem Cells Induced with miR-1/Myocd in Myocardial Infarction Model of Rat. Cell Transplant 2021; 30:9636897211048786. [PMID: 34606735 PMCID: PMC8493326 DOI: 10.1177/09636897211048786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular disease is one of the most common causes of death worldwide. Mesenchymal stem cells (MSCs) are one of the most common sources in cell-based therapies in heart regeneration. There are several methods to differentiate MSCs into cardiac-like cells, such as gene induction. Moreover, using a three-dimensional (3D) culture, such as hydrogels increases efficiency of differentiation. In the current study, mouse adipose-derived MSCs were co-transduced with lentiviruses containing microRNA-1 (miR-1) and Myocardin (Myocd). Then, expression of cardiac markers, such as NK2 homeobox 5(Nkx2-5), GATA binding protein 4 (Gata4), and troponin T type 2 (Tnnt2) was investigated, at both gene and protein levels in two-dimensional (2D) culture and chitosan/collagen hydrogel (CS/CO) as a 3D culture. Additionally, after induction of myocardial infarction (MI) in rats, a patch containing the encapsulated induced cardiomyocytes (iCM/P) was implanted to MI zone. Subsequently, 30 days after MI induction, echocardiography, immunohistochemistry staining, and histological examination were performed to evaluate cardiac function. The results of quantitative real -time polymerase chain reaction (qRT-PCR) and immunocytochemistry showed that co-induction of miR-1 and Myocd in MSCs followed by 3D culture of transduced cells increased expression of cardiac markers. Besides, results of in vivo study implicated that heart function was improved in MI model of rats in iCM/P-treated group. The results suggested that miR-1/Myocd induction combined with encapsulation of transduced cells in CS/CO hydrogel increased efficiency of MSCs differentiation into iCMs and could improve heart function in MI model of rats after implantation.
Collapse
Affiliation(s)
- Samaneh Khazaei
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, Isfahan University, Isfahan, Iran
| | - Masoud Soleimani
- Tissue Engineering and Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Tissue Engineering and Nanomedicine Research Center, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zohreh Hojati
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, Isfahan University, Isfahan, Iran
| |
Collapse
|
24
|
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
|
25
|
Naumova N, Iop L. Bioengineering the Cardiac Conduction System: Advances in Cellular, Gene, and Tissue Engineering for Heart Rhythm Regeneration. Front Bioeng Biotechnol 2021; 9:673477. [PMID: 34409019 PMCID: PMC8365186 DOI: 10.3389/fbioe.2021.673477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/24/2021] [Indexed: 01/01/2023] Open
Abstract
Heart rhythm disturbances caused by different etiologies may affect pediatric and adult patients with life-threatening consequences. When pharmacological therapy is ineffective in treating the disturbances, the implantation of electronic devices to control and/or restore normal heart pacing is a unique clinical management option. Although these artificial devices are life-saving, they display many limitations; not least, they do not have any capability to adapt to somatic growth or respond to neuroautonomic physiological changes. A biological pacemaker could offer a new clinical solution for restoring heart rhythms in the conditions of disorder in the cardiac conduction system. Several experimental approaches, such as cell-based, gene-based approaches, and the combination of both, for the generation of biological pacemakers are currently established and widely studied. Pacemaker bioengineering is also emerging as a technology to regenerate nodal tissues. This review analyzes and summarizes the strategies applied so far for the development of biological pacemakers, and discusses current translational challenges toward the first-in-human clinical application.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
26
|
Kaur K, Hadas Y, Kurian AA, Żak MM, Yoo J, Mahmood A, Girard H, Komargodski R, Io T, Santini MP, Sultana N, Kabir Sharkar MT, Magadum A, Fargnoli A, Yoon S, Chepurko E, Chepurko V, Eliyahu E, Pinto D, Lebeche D, Kovacic JC, Hajjar RJ, Rafii S, Zangi L. Direct Reprogramming Induces Vascular Regeneration Post Muscle Ischemic Injury. Mol Ther 2021; 29:3042-3058. [PMID: 34332145 PMCID: PMC8531157 DOI: 10.1016/j.ymthe.2021.07.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 07/02/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022] Open
Abstract
Reprogramming non-cardiomyocytes (non-CMs) into cardiomyocyte (CM)-like cells is a promising strategy for cardiac regeneration in conditions such as ischemic heart disease. Here, we used a modified mRNA (modRNA) gene delivery platform to deliver a cocktail, termed 7G-modRNA, of four cardiac-reprogramming genes—Gata4 (G), Mef2c (M), Tbx5 (T), and Hand2 (H)—together with three reprogramming-helper genes—dominant-negative (DN)-TGFβ, DN-Wnt8a, and acid ceramidase (AC)—to induce CM-like cells. We showed that 7G-modRNA reprogrammed 57% of CM-like cells in vitro. Through a lineage-tracing model, we determined that delivering the 7G-modRNA cocktail at the time of myocardial infarction reprogrammed ∼25% of CM-like cells in the scar area and significantly improved cardiac function, scar size, long-term survival, and capillary density. Mechanistically, we determined that while 7G-modRNA cannot create de novo beating CMs in vitro or in vivo, it can significantly upregulate pro-angiogenic mesenchymal stromal cells markers and transcription factors. We also demonstrated that our 7G-modRNA cocktail leads to neovascularization in ischemic-limb injury, indicating CM-like cells importance in other organs besides the heart. modRNA is currently being used around the globe for vaccination against COVID-19, and this study proves this is a safe, highly efficient gene delivery approach with therapeutic potential to treat ischemic diseases.
Collapse
Affiliation(s)
- Keerat Kaur
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Yoav Hadas
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Ann Anu Kurian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Magdalena M Żak
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Asharee Mahmood
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Hanna Girard
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Rinat Komargodski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Toshiro Io
- Research Department, Ono Pharmaceutical Co. Ltd., Osaka, Japan, 103-0023
| | - Maria Paola Santini
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Nishat Sultana
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Mohammad Tofael Kabir Sharkar
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Ajit Magadum
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Anthony Fargnoli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Seonghun Yoon
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Elena Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Vadim Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Multiscale Biology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Dalila Pinto
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Djamel Lebeche
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Roger J Hajjar
- Phospholamban Foundation, Amsterdam, The Netherlands 1775 ZH
| | - Shahin Rafii
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029.
| |
Collapse
|
27
|
Lai X, Lin P, Ye J, Liu W, Lin S, Lin Z. Reference Module-Based Analysis of Ovarian Cancer Transcriptome Identifies Important Modules and Potential Drugs. Biochem Genet 2021; 60:433-451. [PMID: 34173117 DOI: 10.1007/s10528-021-10101-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022]
Abstract
Ovarian cancer (OVC) is often diagnosed at the advanced stage resulting in a poor overall outcome for the patient. The disease mechanisms, prognosis, and treatment require imperative elucidation. A rank-based module-centric framework was proposed to analyze the key modules related to the development, prognosis, and treatment of OVC. The ovarian cancer cell line microarray dataset GSE43765 from the Gene Expression Omnibus database was used to construct the reference modules by weighted gene correlation network analysis. Twenty-three reference modules were tested for stability and functionally annotated. Furthermore, to demonstrate the utility of reference modules, two more OVC datasets were collected, and their gene expression profiles were projected to the reference modules to generate a module-level expression. An epithelial-mesenchymal transition module was activated in OVC compared to the normal epithelium, and a pluripotency module was activated in ovarian cancer stroma compared to ovarian cancer epithelium. Seven differentially expressed modules were identified in OVC compared to the normal ovarian epithelium, with five up-regulated, and two down-regulated. One module was identified to be predictive of patient overall survival. Four modules were enriched with SNP signals. Based on differentially expressed modules and hub genes, five candidate drugs were screened. The hub genes of those modules merit further investigation. We firstly propose the reference module-based analysis of OVC. The utility of the analysis framework can be extended to transcriptome data of other kinds of diseases.
Collapse
Affiliation(s)
- Xuedan Lai
- Department of Gynaecology and Obstetrics, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Peihong Lin
- Department of Gynaecology and Obstetrics, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Jianwen Ye
- Department of Gynaecology and Obstetrics, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Wei Liu
- Department of Bioinformatics, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, People's Republic of China
| | - Shiqiang Lin
- Department of Bioinformatics, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, People's Republic of China
| | - Zhou Lin
- Department of Gynaecology and Obstetrics, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China.
| |
Collapse
|
28
|
Abdel-Raouf KMA, Rezgui R, Stefanini C, Teo JCM, Christoforou N. Transdifferentiation of Human Fibroblasts into Skeletal Muscle Cells: Optimization and Assembly into Engineered Tissue Constructs through Biological Ligands. BIOLOGY 2021; 10:biology10060539. [PMID: 34208436 PMCID: PMC8235639 DOI: 10.3390/biology10060539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Engineered human skeletal muscle tissue is a platform tool that can help scientists and physicians better understand human physiology, pharmacology, and disease modeling. Over the past few years this area of research has been actively being pursued by many labs worldwide. Significant challenges remain, including accessing an adequate cell source, and achieving proper physiological-like architecture of the engineered tissue. To address cell resourcing we aimed at further optimizing a process called transdifferentiation which involves the direct conversion of fibroblasts into skeletal muscle cells. The opportunity here is that fibroblasts are readily available and can be expanded sufficiently to meet the needs of a tissue engineering approach. Additionally, we aimed to demonstrate the applicability of transdifferentiation in assembling tissue engineered skeletal muscle. We implemented a screening process of protein ligands in an effort to refine transdifferentiation, and identified that most proteins resulted in a deficit in transdifferentiation efficiency, although one resulted in robust expansion of cultured cells. We were also successful in assembling engineered constructs consisting of transdifferentiated cells. Future directives involve demonstrating that the engineered tissues are capable of contractile and functional activity, and pursuit of optimizing factors such as electrical and chemical exposure, towards achieving physiological parameters observed in human muscle. Abstract The development of robust skeletal muscle models has been challenging due to the partial recapitulation of human physiology and architecture. Reliable and innovative 3D skeletal muscle models recently described offer an alternative that more accurately captures the in vivo environment but require an abundant cell source. Direct reprogramming or transdifferentiation has been considered as an alternative. Recent reports have provided evidence for significant improvements in the efficiency of derivation of human skeletal myotubes from human fibroblasts. Herein we aimed at improving the transdifferentiation process of human fibroblasts (tHFs), in addition to the differentiation of murine skeletal myoblasts (C2C12), and the differentiation of primary human skeletal myoblasts (HSkM). Differentiating or transdifferentiating cells were exposed to single or combinations of biological ligands, including Follistatin, GDF8, FGF2, GDF11, GDF15, hGH, TMSB4X, BMP4, BMP7, IL6, and TNF-α. These were selected for their critical roles in myogenesis and regeneration. C2C12 and tHFs displayed significant differentiation deficits when exposed to FGF2, BMP4, BMP7, and TNF-α, while proliferation was significantly enhanced by FGF2. When exposed to combinations of ligands, we observed consistent deficit differentiation when TNF-α was included. Finally, our direct reprogramming technique allowed for the assembly of elongated, cross-striated, and aligned tHFs within tissue-engineered 3D skeletal muscle constructs. In conclusion, we describe an efficient system to transdifferentiate human fibroblasts into myogenic cells and a platform for the generation of tissue-engineered constructs. Future directions will involve the evaluation of the functional characteristics of these engineered tissues.
Collapse
Affiliation(s)
- Khaled M. A. Abdel-Raouf
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Department of Biology, American University in Cairo, New Cairo 11835, Egypt
- Correspondence: (K.M.A.A.-R.); (N.C.)
| | - Rachid Rezgui
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Cesare Stefanini
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Jeremy C. M. Teo
- Department of Mechanical and Biomedical Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Nicolas Christoforou
- Pfizer Inc., Rare Disease Research Unit, 610 Main Street, Cambridge, MA 02139, USA
- Correspondence: (K.M.A.A.-R.); (N.C.)
| |
Collapse
|
29
|
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
|
30
|
Aberdeen H, Battles K, Taylor A, Garner-Donald J, Davis-Wilson A, Rogers BT, Cavalier C, Williams ED. The Aging Vasculature: Glucose Tolerance, Hypoglycemia and the Role of the Serum Response Factor. J Cardiovasc Dev Dis 2021; 8:58. [PMID: 34067715 PMCID: PMC8156687 DOI: 10.3390/jcdd8050058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
The fastest growing demographic in the U.S. at the present time is those aged 65 years and older. Accompanying advancing age are a myriad of physiological changes in which reserve capacity is diminished and homeostatic control attenuates. One facet of homeostatic control lost with advancing age is glucose tolerance. Nowhere is this more accentuated than in the high proportion of older Americans who are diabetic. Coupled with advancing age, diabetes predisposes affected subjects to the onset and progression of cardiovascular disease (CVD). In the treatment of type 2 diabetes, hypoglycemic episodes are a frequent clinical manifestation, which often result in more severe pathological outcomes compared to those observed in cases of insulin resistance, including premature appearance of biomarkers of senescence. Unfortunately, molecular mechanisms of hypoglycemia remain unclear and the subject of much debate. In this review, the molecular basis of the aging vasculature (endothelium) and how glycemic flux drives the appearance of cardiovascular lesions and injury are discussed. Further, we review the potential role of the serum response factor (SRF) in driving glycemic flux-related cellular signaling through its association with various proteins.
Collapse
Affiliation(s)
- Hazel Aberdeen
- Department of Biomedical Sciences, Baptist Health Sciences University, Memphis, TN 38103, USA; or
| | - Kaela Battles
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Ariana Taylor
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Jeranae Garner-Donald
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Ana Davis-Wilson
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Bryan T. Rogers
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Candice Cavalier
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Emmanuel D. Williams
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| |
Collapse
|
31
|
Garry GA, Bezprozvannaya S, Chen K, Zhou H, Hashimoto H, Morales MG, Liu N, Bassel-Duby R, Olson EN. The histone reader PHF7 cooperates with the SWI/SNF complex at cardiac super enhancers to promote direct reprogramming. Nat Cell Biol 2021; 23:467-475. [PMID: 33941892 DOI: 10.1038/s41556-021-00668-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Direct cardiac reprogramming of fibroblasts to cardiomyocytes presents an attractive therapeutic strategy to restore cardiac function following injury. Cardiac reprogramming was initially achieved through overexpression of the transcription factors Gata4, Mef2c and Tbx5; later, Hand2 and Akt1 were found to further enhance this process1-5. Yet, staunch epigenetic barriers severely limit the ability of these cocktails to reprogramme adult fibroblasts6,7. We undertook a screen of mammalian gene regulatory factors to discover novel regulators of cardiac reprogramming in adult fibroblasts and identified the histone reader PHF7 as the most potent activating factor8. Mechanistically, PHF7 localizes to cardiac super enhancers in fibroblasts, and through cooperation with the SWI/SNF complex, it increases chromatin accessibility and transcription factor binding at these sites. Furthermore, PHF7 recruits cardiac transcription factors to activate a positive transcriptional autoregulatory circuit in reprogramming. Importantly, PHF7 achieves efficient reprogramming in the absence of Gata4. Here, we highlight the underexplored necessity of cardiac epigenetic readers, such as PHF7, in harnessing chromatin remodelling and transcriptional complexes to overcome critical barriers to direct cardiac reprogramming.
Collapse
Affiliation(s)
- Glynnis A Garry
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenian Chen
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huanyu Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hisayuki Hashimoto
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,The Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
32
|
Lee JH, Chen Z, He S, Zhou JK, Tsai A, Truskey GA, Leong KW. Emulating Early Atherosclerosis in a Vascular Microphysiological System Using Branched Tissue-Engineered Blood Vessels. Adv Biol (Weinh) 2021; 5:e2000428. [PMID: 33852179 PMCID: PMC9951769 DOI: 10.1002/adbi.202000428] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/26/2021] [Indexed: 02/04/2023]
Abstract
Atherosclerosis begins with the accumulation of cholesterol-carrying lipoproteins on blood vessel walls and progresses to endothelial cell dysfunction, monocyte adhesion, and foam cell formation. Endothelialized tissue-engineered blood vessels (TEBVs) have previously been fabricated to recapitulate artery functionalities, including vasoconstriction, vasodilation, and endothelium activation. Here, the initiation of atherosclerosis is emulated by designing branched TEBVs (brTEBVs) of various geometries treated with enzyme-modified low-density-lipoprotein (eLDL) and TNF-α to induce endothelial cell dysfunction and adhesion of perfused human monocytes. Locations of monocyte adhesion under pulsatile flow are identified, and the hemodynamics in the brTEBVs are characterized using particle image velocimetry (PIV) and computational fluid dynamics (CFD). Monocyte adhesion is greater at the side outlets than at the main outlets or inlets, and is greatest at larger side outlet branching angles (60° or 80° vs 45°). In PIV experiments, the branched side outlets are identified as atherosclerosis-prone areas where fluorescent particles show a transient swirling motion following flow pulses; in CFD simulations, side outlets with larger branching angles show higher vorticity magnitude and greater flow disturbance than other areas. These results suggest that the branched TEBVs with eLDL/TNF-α treatment provide a physiologically relevant model of early atherosclerosis for preclinical studies.
Collapse
Affiliation(s)
- Jounghyun H. Lee
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Joyce K. Zhou
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alexander Tsai
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
33
|
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
|
34
|
Riching AS, Danis E, Zhao Y, Cao Y, Chi C, Bagchi RA, Klein BJ, Xu H, Kutateladze TG, McKinsey TA, Buttrick PM, Song K. Suppression of canonical TGF-β signaling enables GATA4 to interact with H3K27me3 demethylase JMJD3 to promote cardiomyogenesis. J Mol Cell Cardiol 2020; 153:44-59. [PMID: 33359755 DOI: 10.1016/j.yjmcc.2020.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 11/14/2020] [Accepted: 12/12/2020] [Indexed: 01/07/2023]
Abstract
Direct reprogramming of fibroblasts into cardiomyocytes (CMs) represents a promising strategy to regenerate CMs lost after ischemic heart injury. Overexpression of GATA4, HAND2, MEF2C, TBX5, miR-1, and miR-133 (GHMT2m) along with transforming growth factor beta (TGF-β) inhibition efficiently promote reprogramming. However, the mechanisms by which TGF-β blockade promotes cardiac reprogramming remain unknown. Here, we identify interactions between the histone H3 lysine 27 trimethylation (H3K27me3) demethylase JMJD3, the SWI/SNF remodeling complex subunit BRG1, and cardiac transcription factors. Furthermore, canonical TGF-β signaling regulates the interaction between GATA4 and JMJD3. TGF-β activation impairs the ability of GATA4 to bind target genes and prevents demethylation of H3K27 at cardiac gene promoters during cardiac reprogramming. Finally, a mutation in GATA4 (V267M) that is associated with congenital heart disease exhibits reduced binding to JMJD3 and impairs cardiomyogenesis. Thus, we have identified an epigenetic mechanism wherein canonical TGF-β pathway activation impairs cardiac gene programming, in part by interfering with GATA4-JMJD3 interactions.
Collapse
Affiliation(s)
- Andrew S Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Etienne Danis
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yuanbiao Zhao
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yingqiong Cao
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rushita A Bagchi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brianna J Klein
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hongyan Xu
- Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Tatiana G Kutateladze
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Peter M Buttrick
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
35
|
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
|
36
|
Jiang L, Liang J, Huang W, Wu Z, Paul C, Wang Y. Strategies and Challenges to Improve Cellular Programming-Based Approaches for Heart Regeneration Therapy. Int J Mol Sci 2020; 21:E7662. [PMID: 33081233 PMCID: PMC7589611 DOI: 10.3390/ijms21207662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
37
|
Lee S, Park BW, Lee YJ, Ban K, Park HJ. In vivo combinatory gene therapy synergistically promotes cardiac function and vascular regeneration following myocardial infarction. J Tissue Eng 2020; 11:2041731420953413. [PMID: 35003614 PMCID: PMC8738857 DOI: 10.1177/2041731420953413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Since myocardial infarction (MI) excessively damage the myocardium and blood
vessels, the therapeutic approach for treating MI hearts should simultaneously
target these two major components in the heart to achieve comprehensive cardiac
repair. Here, we investigated a combinatory platform of ETV2 and Gata4, Mef2c
and Tbx5 (GMT) transcription factors to develop a strategy that can rejuvenate
both myocardium and vasculatures together in MI hearts. Previously ETV2
demonstrated significant effects on neovascularization and GMT was known to
directly reprogram cardiac fibroblasts into cardiomyocytes under in vivo
condition. Subsequently, intramyocardial delivery of a combination of retroviral
GMT and adenoviral ETV2 particles into the rat MI hearts significantly increased
viable myocardium area, capillary density compared to ETV2 or GMT only treated
hearts, leading to improved heart function and reduced scar formation. These
results demonstrate that this combinatorial gene therapy can be a promising
approach to enhance the cardiac repair in MI hearts.
Collapse
Affiliation(s)
- Sunghun Lee
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Kowloon tong, Hong Kong
| | - Bong-Woo Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Jin Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Kowloon tong, Hong Kong
| | - Hun-Jun Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
38
|
Stem cells and new intervention measures as emerging therapy in cardiac surgery. POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2020; 17:1-7. [PMID: 32728355 PMCID: PMC7379210 DOI: 10.5114/kitp.2020.94183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/20/2020] [Indexed: 11/17/2022]
Abstract
Cardiovascular disease (CVD) presents a great burden for elderly patients, their caregivers, and health systems. Structural and functional alterations of vessels accumulate throughout life, culminating in increased risk of developing CVD. Several inflammatory pathway are involved in vascular ageing. The growing elderly population worldwide highlights the need to understand how aging promotes CVD in order to develop new strategies to confront this challenge. In this review we analyzed the role of stem cells and new intervention measures as emerging drugs for vascular aging.
Collapse
|
39
|
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
|
40
|
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
|
41
|
Mengmeng X, Yuejuan X, Sun C, Yanan L, Fen L, Kun S. Novel mutations of the SRF gene in Chinese sporadic conotruncal heart defect patients. BMC MEDICAL GENETICS 2020; 21:95. [PMID: 32380971 PMCID: PMC7203814 DOI: 10.1186/s12881-020-01032-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 04/22/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Conotruncal heart defects (CTDs) are a group of congenital heart malformations that cause anomalies of cardiac outflow tracts. In the past few decades, many genes related to CTDs have been reported. Serum response factor (SRF) is a ubiquitous nuclear protein that acts as transcription factor, and SRF was found to be a critical factor in heart development and to be strongly expressed in the myocardium of the developing mouse and chicken hearts. The targeted inactivation of SRF during heart development leads to embryonic lethality and myocardial defects in mice. METHODS To illustrate the relationship between SRF and human heart defects, we screened SRF mutations in 527 CTD patients, a cross sectional study. DNA was extracted from peripheral leukocyte cells for target sequencing. The mutations of SRF were detected and validated by Sanger sequencing. The affection of the mutations on wild-type protein was analyzed by in silico softwares. Western blot and real time PCR were used to analyze the changes of the expression of the mutant mRNA and protein. In addition, we carried out dual luciferase reporter assay to explore the transcriptional activity of the mutant SRF. RESULTS Among the target sequencing results of 527 patients, two novel mutations (Mut1: c.821A > G p.G274D, the adenine(A) was mutated to guanine(G) at position 821 of the SRF gene coding sequences (CDS), lead to the Glycine(G) mutated to Asparticacid(D) at position 274 of the SRF protein amino acid sequences; Mut2: c.880G > T p.G294C, the guanine(G) was mutated to thymine (T) at position 880 of the SRF CDS, lead to the Glycine(G) mutated to Cysteine (C) at position 294 of the SRF protein amino acid sequences.) of SRF (NM_003131.4) were identified. Western blotting and real-time PCR showed that there were no obvious differences between the protein expression and mRNA transcription of mutants and wild-type SRF. A dual luciferase reporter assay showed that both SRF mutants (G274D and G294C) impaired SRF transcriptional activity at the SRF promoter and atrial natriuretic factor (ANF) promoter (p < 0.05), additionally, the mutants displayed reduced synergism with GATA4. CONCLUSION These results suggest that SRF-p.G274D and SRF-p.G294C may have potential pathogenic effects.
Collapse
Affiliation(s)
- Xu Mengmeng
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Xu Yuejuan
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China.
| | - Chen Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Lu Yanan
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China
| | - Li Fen
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, No. 1678, Dongfang Road, Shanghai, 200127, China
| | - Sun Kun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang road, Shanghai, 200092, China.
| |
Collapse
|
42
|
Isoform Specific Effects of Mef2C during Direct Cardiac Reprogramming. Cells 2020; 9:cells9020268. [PMID: 31979018 PMCID: PMC7072587 DOI: 10.3390/cells9020268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 01/14/2023] Open
Abstract
Direct conversion of cardiac fibroblasts into induced cardiomyocytes (iCMs) by forced expression of defined factors holds great potential for regenerative medicine by offering an alternative strategy for treatment of heart disease. Successful iCM conversion can be achieved by minimally using three transcription factors, Mef2c (M), Gata4(G), and Tbx5 (T). Despite increasing interest in iCM mechanistic studies using MGT(polycistronic construct with optimal expression of M,G and T), the reprogramming efficiency varies among different laboratories. Two main Mef2c isoforms (isoform2, Mi2 and isoform4, Mi4) are present in heart and are used separately by different labs, for iCM reprogramming. It is currently unknown if differently spliced isoform of Mef2c contributes to varied reprogramming efficiency. Here, we used Mi2 and Mi4 together with Gata4 and Tbx5 in separate vectors or polycistronic vector, to convert fibroblasts to iCMs. We found that Mi2 can induce higher reprogramming efficiency than Mi4 in MEFs. Addition of Hand2 to MGT retroviral cocktail or polycistronic Mi2-GT retroviruses further enhanced the iCM conversion. Overall, this study demonstrated the isoform specific effects of Mef2c, during iCM reprogramming, clarified some discrepancy about varied efficiency among labs and might lead to future research into the role of alternative splicing and the consequent variants in cell fate determination.
Collapse
|
43
|
Spotlight on epigenetic reprogramming in cardiac regeneration. Semin Cell Dev Biol 2020; 97:26-37. [PMID: 31002867 DOI: 10.1016/j.semcdb.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
44
|
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
|
45
|
Stone NR, Gifford CA, Thomas R, Pratt KJB, Samse-Knapp K, Mohamed TMA, Radzinsky EM, Schricker A, Ye L, Yu P, van Bemmel JG, Ivey KN, Pollard KS, Srivastava D. Context-Specific Transcription Factor Functions Regulate Epigenomic and Transcriptional Dynamics during Cardiac Reprogramming. Cell Stem Cell 2019; 25:87-102.e9. [PMID: 31271750 PMCID: PMC6632093 DOI: 10.1016/j.stem.2019.06.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
Ectopic expression of combinations of transcription factors (TFs) can drive direct lineage conversion, thereby reprogramming a somatic cell's identity. To determine the molecular mechanisms by which Gata4, Mef2c, and Tbx5 (GMT) induce conversion from a cardiac fibroblast toward an induced cardiomyocyte, we performed comprehensive transcriptomic, DNA-occupancy, and epigenomic interrogation throughout the reprogramming process. Integration of these datasets identified new TFs involved in cardiac reprogramming and revealed context-specific roles for GMT, including the ability of Mef2c and Tbx5 to independently promote chromatin remodeling at previously inaccessible sites. We also find evidence for cooperative facilitation and refinement of each TF's binding profile in a combinatorial setting. A reporter assay employing newly defined regulatory elements confirmed that binding of a single TF can be sufficient for gene activation, suggesting that co-binding events do not necessarily reflect synergy. These results shed light on fundamental mechanisms by which combinations of TFs direct lineage conversion.
Collapse
Affiliation(s)
- Nicole R Stone
- Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Pediatrics and Biochemistry & Biophysics, University of California, San Francisco, CA 94143, USA
| | - Casey A Gifford
- Gladstone Institutes, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | | | | | | | - Tamer M A Mohamed
- Gladstone Institutes, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | | | | | - Lin Ye
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Pengzhi Yu
- Gladstone Institutes, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | | | - Kathryn N Ivey
- Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Pediatrics and Biochemistry & Biophysics, University of California, San Francisco, CA 94143, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Epidemiology & Biostatistics, University of California, San Francisco, CA 94143, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Deepak Srivastava
- Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Pediatrics and Biochemistry & Biophysics, University of California, San Francisco, CA 94143, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA 94158, USA.
| |
Collapse
|
46
|
Sauls K, Greco TM, Wang L, Zou M, Villasmil M, Qian L, Cristea IM, Conlon FL. Initiating Events in Direct Cardiomyocyte Reprogramming. Cell Rep 2019; 22:1913-1922. [PMID: 29444441 DOI: 10.1016/j.celrep.2018.01.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 11/30/2017] [Accepted: 01/17/2018] [Indexed: 01/14/2023] Open
Abstract
Direct reprogramming of fibroblasts into cardiomyocyte-like cells (iCM) holds great potential for heart regeneration and disease modeling and may lead to future therapeutic applications. Currently, application of this technology is limited by our lack of understanding of the molecular mechanisms that drive direct iCM reprogramming. Using a quantitative mass spectrometry-based proteomic approach, we identified the temporal global changes in protein abundance that occur during initial phases of iCM reprogramming. Collectively, our results show systematic and temporally distinct alterations in levels of specific functional classes of proteins during the initiating steps of reprogramming including extracellular matrix proteins, translation factors, and chromatin-binding proteins. We have constructed protein relational networks associated with the initial transition of a fibroblast into an iCM. These findings demonstrate the presence of an orchestrated series of temporal steps associated with dynamic changes in protein abundance in a defined group of protein pathways during the initiating events of direct reprogramming.
Collapse
Affiliation(s)
- Kimberly Sauls
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Li Wang
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Pathology and Laboratory Medicine, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Meng Zou
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA
| | - Michelle Villasmil
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA; Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Qian
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Pathology and Laboratory Medicine, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Frank L Conlon
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, UNC-Chapel Hill, Chapel Hill, NC 27599 USA; Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, Chapel Hill, NC 27599, USA; Integrative Program for Biological and Genome Sciences, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
47
|
Cardiac Reprogramming Factors Synergistically Activate Genome-wide Cardiogenic Stage-Specific Enhancers. Cell Stem Cell 2019; 25:69-86.e5. [PMID: 31080136 DOI: 10.1016/j.stem.2019.03.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/07/2018] [Accepted: 03/25/2019] [Indexed: 11/21/2022]
Abstract
The cardiogenic transcription factors (TFs) Mef2c, Gata4, and Tbx5 can directly reprogram fibroblasts to induced cardiac-like myocytes (iCLMs), presenting a potential source of cells for cardiac repair. While activity of these TFs is enhanced by Hand2 and Akt1, their genomic targets and interactions during reprogramming are not well studied. We performed genome-wide analyses of cardiogenic TF binding and enhancer profiling during cardiac reprogramming. We found that these TFs synergistically activate enhancers highlighted by Mef2c binding sites and that Hand2 and Akt1 coordinately recruit other TFs to enhancer elements. Intriguingly, these enhancer landscapes collectively resemble patterns of enhancer activation during embryonic cardiogenesis. We further constructed a cardiac reprogramming gene regulatory network and found repression of EGFR signaling pathway genes. Consistently, chemical inhibition of EGFR signaling augmented reprogramming. Thus, by defining epigenetic landscapes these findings reveal synergistic transcriptional activation across a broad landscape of cardiac enhancers and key signaling pathways that govern iCLM reprogramming.
Collapse
|
48
|
Guo Y, Lei I, Tian S, Gao W, Hacer K, Li Y, Wang S, Liu L, Wang Z. Chemical suppression of specific C-C chemokine signaling pathways enhances cardiac reprogramming. J Biol Chem 2019; 294:9134-9146. [PMID: 31023824 DOI: 10.1074/jbc.ra118.006000] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 04/25/2019] [Indexed: 01/02/2023] Open
Abstract
Reprogramming of fibroblasts into induced cardiomyocytes (iCMs) is a potentially promising strategy for regenerating a damaged heart. However, low fibroblast-cardiomyocyte conversion rates remain a major challenge in this reprogramming. To this end, here we conducted a chemical screen and identified four agents, insulin-like growth factor-1, Mll1 inhibitor MM589, transforming growth factor-β inhibitor A83-01, and Bmi1 inhibitor PTC-209, termed IMAP, which coordinately enhanced reprogramming efficiency. Using α-muscle heavy chain-GFP-tagged mouse embryo fibroblasts as a starting cell type, we observed that the IMAP treatment increases iCM formation 6-fold. IMAP stimulated higher cardiac troponin T and α-actinin expression and increased sarcomere formation, coinciding with up-regulated expression of many cardiac genes and down-regulated fibroblast gene expression. Furthermore, IMAP promoted higher spontaneous beating and calcium transient activities of iCMs derived from neonatal cardiac fibroblasts. Intriguingly, we also observed that the IMAP treatment repressed many genes involved in immune responses, particularly those in specific C-C chemokine signaling pathways. We therefore investigated the roles of C-C motif chemokine ligand 3 (CCL3), CCL6, and CCL17 in cardiac reprogramming and observed that they inhibited iCM formation, whereas inhibitors of C-C motif chemokine receptor 1 (CCR1), CCR4, and CCR5 had the opposite effect. These results indicated that the IMAP treatment directly suppresses specific C-C chemokine signaling pathways and thereby enhances cardiac reprogramming. In conclusion, a combination of four chemicals, named here IMAP, suppresses specific C-C chemokine signaling pathways and facilitates Mef2c/Gata4/Tbx5 (MGT)-induced cardiac reprogramming, providing a potential means for iCM formation in clinical applications.
Collapse
Affiliation(s)
- Yijing Guo
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109.,Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ienglam Lei
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109.,Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Shuo Tian
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109
| | - Wenbin Gao
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109.,First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Karatas Hacer
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109.,Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, and.,Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109
| | - Yangbing Li
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109.,Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, and.,Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109.,Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, Michigan 48109, and.,Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109
| | - Liu Liu
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109,
| | - Zhong Wang
- From the Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan 48109,
| |
Collapse
|
49
|
Wang M, Ling W, Xiong C, Xie D, Chu X, Li Y, Qiu X, Li Y, Xiao X. Potential Strategies for Cardiac Diseases: Lineage Reprogramming of Somatic Cells into Induced Cardiomyocytes. Cell Reprogram 2019; 21:63-77. [DOI: 10.1089/cell.2018.0052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mingyu Wang
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Wenhui Ling
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chunxia Xiong
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Dengfeng Xie
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xinyue Chu
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yunxin Li
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yuemin Li
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Animal Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
50
|
Dorr KM, Conlon FL. Proteomic-based approaches to cardiac development and disease. Curr Opin Chem Biol 2019; 48:150-157. [PMID: 30711722 DOI: 10.1016/j.cbpa.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 01/14/2023]
Abstract
Congenital malformations, or structural birth defects, are now the leading cause of infant mortality in the United States and Europe (Dolk et al., 2010; Heron et al., 2009). Of the congenital malformations, congenital heart disease (CHD) is the most common (Dolk et al., 2010; Heron et al., 2009). Thus, a molecular understanding of heart development is an essential goal for improving clinical approaches to CHD. However, CHDs are commonly a result of genetic defects that manifest themselves in a spatial and temporal manner during the early stages of embryogenesis, leaving them mostly intractable to mass spectrometry-based analysis. Here, we describe the technologies and advancements in the field of mass spectrometry over the past few years that have begun to provide insights into the molecular and cellular basis of CHD and prospects for these types of approaches in the future.
Collapse
Affiliation(s)
- Kerry M Dorr
- Department of Biology and Genetics, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank L Conlon
- Department of Biology and Genetics, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|