1
|
Burgon PG, Weldrick JJ, Talab OMSA, Nadeer M, Nomikos M, Megeney LA. Regulatory Mechanisms That Guide the Fetal to Postnatal Transition of Cardiomyocytes. Cells 2023; 12:2324. [PMID: 37759546 PMCID: PMC10528641 DOI: 10.3390/cells12182324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart disease remains a global leading cause of death and disability, necessitating a comprehensive understanding of the heart's development, repair, and dysfunction. This review surveys recent discoveries that explore the developmental transition of proliferative fetal cardiomyocytes into hypertrophic postnatal cardiomyocytes, a process yet to be well-defined. This transition is key to the heart's growth and has promising therapeutic potential, particularly for congenital or acquired heart damage, such as myocardial infarctions. Although significant progress has been made, much work is needed to unravel the complex interplay of signaling pathways that regulate cardiomyocyte proliferation and hypertrophy. This review provides a detailed perspective for future research directions aimed at the potential therapeutic harnessing of the perinatal heart transitions.
Collapse
Affiliation(s)
- Patrick G. Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Jonathan J. Weldrick
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
| | | | - Muhammad Nadeer
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Lynn A. Megeney
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
2
|
Castillo-Casas JM, Caño-Carrillo S, Sánchez-Fernández C, Franco D, Lozano-Velasco E. Comparative Analysis of Heart Regeneration: Searching for the Key to Heal the Heart-Part II: Molecular Mechanisms of Cardiac Regeneration. J Cardiovasc Dev Dis 2023; 10:357. [PMID: 37754786 PMCID: PMC10531542 DOI: 10.3390/jcdd10090357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, among which ischemic heart disease is the most representative. Myocardial infarction results from occlusion of a coronary artery, which leads to an insufficient blood supply to the myocardium. As it is well known, the massive loss of cardiomyocytes cannot be solved due the limited regenerative ability of the adult mammalian hearts. In contrast, some lower vertebrate species can regenerate the heart after an injury; their study has disclosed some of the involved cell types, molecular mechanisms and signaling pathways during the regenerative process. In this 'two parts' review, we discuss the current state-of-the-art of the main response to achieve heart regeneration, where several processes are involved and essential for cardiac regeneration.
Collapse
Affiliation(s)
- Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Cristina Sánchez-Fernández
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| |
Collapse
|
3
|
Ceja L, Escopete SS, Hughes L, Lopez LV, Camberos V, Vallejos P, Wall NR, Kearns-Jonker M. Neonatal Cardiovascular-Progenitor-Cell-Derived Extracellular Vesicles Activate YAP1 in Adult Cardiac Progenitor Cells. Int J Mol Sci 2023; 24:ijms24098088. [PMID: 37175796 PMCID: PMC10179407 DOI: 10.3390/ijms24098088] [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: 03/20/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
New stem cell and extracellular-vesicle-based therapies have the potential to improve outcomes for the increasing number of patients with heart failure. Since neonates have a significantly enhanced regenerative ability, we hypothesized that extracellular vesicles isolated from Islet-1+ expressing neonatal human cardiovascular progenitors (CPCs) will induce transcriptomic changes associated with improved regenerative capability when co-cultured with CPCs derived from adult humans. In order to test this hypothesis, we isolated extracellular vesicles from human neonatal Islet-1+ CPCs, analyzed the extracellular vesicle content using RNAseq, and treated adult CPCs with extracellular vesicles derived from neonatal CPCs to assess their functional effect. AKT, ERBB, and YAP1 transcripts were elevated in adult CPCs treated with neonatal CPC-derived extracellular vesicles. YAP1 is lost after the neonatal period but can stimulate cardiac regeneration. Our results demonstrate that YAP1 and additional transcripts associated with improved cardiovascular regeneration, as well as the activation of the cell cycle, can be achieved by the treatment of adult CPCs with neonatal CPC-derived extracellular vesicles. Progenitor cells derived from neonates secrete extracellular vesicles with the potential to stimulate and potentially improve functional effects in adult CPCs used for cardiovascular repair.
Collapse
Affiliation(s)
- Lourdes Ceja
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Sean S Escopete
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lorelei Hughes
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Larry V Lopez
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Victor Camberos
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Paul Vallejos
- Division of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Nathan R Wall
- Division of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
4
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
5
|
Defining the molecular underpinnings controlling cardiomyocyte proliferation. Clin Sci (Lond) 2022; 136:911-934. [PMID: 35723259 DOI: 10.1042/cs20211180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 12/11/2022]
Abstract
Shortly after birth, mammalian cardiomyocytes (CM) exit the cell cycle and cease to proliferate. The inability of adult CM to replicate renders the heart particularly vulnerable to injury. Restoration of CM proliferation would be an attractive clinical target for regenerative therapies that can preserve contractile function and thus prevent the development of heart failure. Our review focuses on recent progress in understanding the tight regulation of signaling pathways and their downstream molecular mechanisms that underly the inability of CM to proliferate in vivo. In this review, we describe the temporal expression of cell cycle activators e.g., cyclin/Cdk complexes and their inhibitors including p16, p21, p27 and members of the retinoblastoma gene family during gestation and postnatal life. The differential impact of members of the E2f transcription factor family and microRNAs on the regulation of positive and negative cell cycle factors is discussed. This review also highlights seminal studies that identified the coordination of signaling mechanisms that can potently activate CM cell cycle re-entry including the Wnt/Ctnnb1, Hippo, Pi3K-Akt and Nrg1-Erbb2/4 pathways. We also present an up-to-date account of landmark studies analyzing the effect of various genes such as Argin, Dystrophin, Fstl1, Meis1, Pitx2 and Pkm2 that are responsible for either inhibition or activation of CM cell division. All these reports describe bona fide therapeutically targets that could guide future clinical studies toward cardiac repair.
Collapse
|
6
|
Mammoto T, Hunyenyiwa T, Kyi P, Hendee K, Matus K, Rao S, Lee SH, Tabima DM, Chesler NC, Mammoto A. Hydrostatic Pressure Controls Angiogenesis Through Endothelial YAP1 During Lung Regeneration. Front Bioeng Biotechnol 2022; 10:823642. [PMID: 35252132 PMCID: PMC8896883 DOI: 10.3389/fbioe.2022.823642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary artery (PA) pressure increases during lung growth after unilateral pneumonectomy (PNX). Mechanosensitive transcriptional co-activator, yes-associated protein (YAP1), in endothelial cells (ECs) is necessary for angiogenesis during post-PNX lung growth. We investigate whether increases in PA pressure following PNX control-angiogenesis through YAP1. When hydrostatic pressure is applied to human pulmonary arterial ECs (HPAECs), the expression of YAP1, transcription factor TEAD1, and angiogenic factor receptor Tie2 increases, while these effects are inhibited when HPAECs are treated with YAP1 siRNA or YAP1S94A mutant that fails to bind to TEAD1. Hydrostatic pressure also stimulates DNA synthesis, cell migration, and EC sprouting in HPAECs, while YAP1 knockdown or YAP1S94A mutant inhibits the effects. Gene enrichment analysis reveals that the levels of genes involved in extracellular matrix (ECM), cell adhesion, regeneration, or angiogenesis are altered in post-PNX mouse lung ECs, which interact with YAP1. Exosomes are known to promote tissue regeneration. Proteomics analysis reveals that exosomes isolated from conditioned media of post-PNX mouse lung ECs contain the higher levels of ECM and cell-adhesion proteins compared to those from sham-operated mouse lung ECs. Recruitment of host lung ECs and blood vessel formation are stimulated in the fibrin gel containing exosomes isolated from post-PNX mouse lung ECs or pressurized ECs, while YAP1 knockdown inhibits the effects. These results suggest that increases in PA pressure stimulate angiogenesis through YAP1 during regenerative lung growth.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tendai Hunyenyiwa
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Priscilla Kyi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kienna Matus
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sridhar Rao
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti, Milwaukee, WI, United States
| | - Sang H. Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Diana M. Tabima
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Naomi C. Chesler
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center and Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto,
| |
Collapse
|
7
|
Limyati Y, Sanjaya A, Lucretia T, Gunadi JW, Biben V, Jasaputra DK, Lesmana R. Potential Role of Exercise in Regulating YAP and TAZ During Cardiomyocytes Aging. Curr Cardiol Rev 2022; 18:24-33. [PMID: 35379136 PMCID: PMC9896415 DOI: 10.2174/1573403x18666220404152924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 01/03/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
Adaptation of cardiac muscle to regular exercise results in morphological and structural changes known as physiological cardiac hypertrophy, to which the Hippo signaling pathway might have contributed. Two major terminal effectors in the Hippo signaling pathway are Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ). The latest studies have reported the role of YAP and TAZ in different life stages, such as in fetal, neonatal, and adult hearts. Their regulation might involve several mechanisms and effectors. One of the possible coregulators is exercise. Exercise plays a role in cardiomyocyte hypertrophic changes during different stages of life, including in aged hearts. YAP/TAZ signaling pathway has a role in physiological cardiac hypertrophy induced by exercise and is associated with cardiac remodelling. Thus, it can be believed that exercise has roles in activating the signaling pathway of YAP and TAZ in aged cardiomyocytes. However, the studies regarding the roles of YAP and TAZ during cardiomyocyte aging are limited. The primary purpose of this review is to explore the response of cardiovascular aging to exercise via signaling pathway of YAP and TAZ.
Collapse
Affiliation(s)
- Yenni Limyati
- Address correspondence to this author at the Postgraduate Doctoral Program Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, 40161; Department of Physical Medicine and Rehabilitation, Unggul Karsa Medika Hospital, Bandung, West Java, 40218; Department of Clinical Skills, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia; Tel/Fax: +62222012186, +62222017621;
| | | | | | | | | | | | | |
Collapse
|
8
|
Li S, Ma W, Cai B. Targeting cardiomyocyte proliferation as a key approach of promoting heart repair after injury. MOLECULAR BIOMEDICINE 2021; 2:34. [PMID: 35006441 PMCID: PMC8607366 DOI: 10.1186/s43556-021-00047-y] [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: 01/14/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases such as myocardial infarction (MI) is a major contributor to human mortality and morbidity. The mammalian adult heart almost loses its plasticity to appreciably regenerate new cardiomyocytes after injuries, such as MI and heart failure. The neonatal heart exhibits robust proliferative capacity when exposed to varying forms of myocardial damage. The ability of the neonatal heart to repair the injury and prevent pathological left ventricular remodeling leads to preserved or improved cardiac function. Therefore, promoting cardiomyocyte proliferation after injuries to reinitiate the process of cardiomyocyte regeneration, and suppress heart failure and other serious cardiovascular problems have become the primary goal of many researchers. Here, we review recent studies in this field and summarize the factors that act upon the proliferation of cardiomyocytes and cardiac repair after injury and discuss the new possibilities for potential clinical treatment strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Shuainan Li
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Wenya Ma
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Benzhi Cai
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China. .,Institute of Clinical Pharmacy, the Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, 150086, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, 150086, China.
| |
Collapse
|
9
|
Valizadeh A, Asghari S, Mansouri P, Alemi F, Majidinia M, Mahmoodpoor A, Yousefi B. The roles of signaling pathways in cardiac regeneration. Curr Med Chem 2021; 29:2142-2166. [PMID: 34521319 DOI: 10.2174/0929867328666210914115411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
In recent years, knowledge of cardiac regeneration mechanisms has dramatically expanded. Regeneration can replace lost parts of organs, common among animal species. The heart is commonly considered an organ with terminal development, which has no reparability potential during post-natal life; however, some intrinsic regeneration capacity has been reported for cardiac muscle, which opens novel avenues in cardiovascular disease treatment. Different endogenous mechanisms were studied for cardiac repairing and regeneration in recent decades. Survival, proliferation, inflammation, angiogenesis, cell-cell communication, cardiomyogenesis, and anti-aging pathways are the most important mechanisms that have been studied in this regard. Several in vitro and animal model studies focused on proliferation induction for cardiac regeneration reported promising results. These studies have mainly focused on promoting proliferation signaling pathways and demonstrated various signaling pathways such as Wnt, PI3K/Akt, IGF-1, TGF-β, Hippo, and VEGF signaling cardiac regeneration. Therefore, in this review, we intended to discuss the connection between different critical signaling pathways in cardiac repair and regeneration.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Samira Asghari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Parinaz Mansouri
- Students Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Forough Alemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia. Iran
| | - Ata Mahmoodpoor
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
10
|
Agrin Yes-associated Protein Promotes the Proliferation of Epicardial Cells. J Cardiovasc Pharmacol 2021; 77:94-99. [PMID: 33136763 DOI: 10.1097/fjc.0000000000000926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/17/2020] [Indexed: 11/26/2022]
Abstract
ABSTRACT Embryonic epicardial cells make an important contribution to cardiac development. However, their proliferation mechanism is still unclear. Epicardial cells from E12.5 fetal hearts were used in our study. Agrin was used to treat these cells. The expression of Aurora B, Ki67, and pH3 was measured by quantitative reverse transcription-polymerase chain reaction and immunofluorescence. The proportion of cells in G1/S/G2 phase was determined by flow cytometry. The results showed that agrin significantly increased the expression of ki67, pH3, and Aurora B in epicardial cells. Flow cytometry results showed that agrin significantly increased the proportion of epicardial cells in S phase. However, blocking yes-associated protein significantly downregulated the levels of ki67, pH3, and Aurora B and the proportion of epicardial cells in S phase. Thus, our results suggest that agrin may promote the proliferation of epicardial cells by regulating the yes-associated protein activity. This may be useful in exploring heart development mechanisms and preventing congenital heart disease.
Collapse
|
11
|
Tao RH, Kobayashi M, Yang Y, Kleinerman ES. Exercise Inhibits Doxorubicin-Induced Damage to Cardiac Vessels and Activation of Hippo/YAP-Mediated Apoptosis. Cancers (Basel) 2021; 13:cancers13112740. [PMID: 34205942 PMCID: PMC8198139 DOI: 10.3390/cancers13112740] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 01/16/2023] Open
Abstract
Dose-related cardiomyopathy is a major side effect following doxorubicin (Dox). To investigate whether exercise (Ex)-induced vasculogenesis plays a role in reducing Dox-induced cardiotoxicity, GFP+ bone marrow (BM) cells from GFP transgenic mice were transplanted into wild-type mice. Transplanted mice were treated with Dox, Ex, Dox+Ex, or control. We found Dox therapy resulted in decreased systolic and diastolic blood flow, decreased ejection fraction and fractional shortening, and decreased vascular endothelial cells and pericytes. These abnormalities were not seen in Dox+Ex hearts. Heart tissues from control-, Ex-, or Dox-treated mice showed a small number of GFP+ cells. By contrast, the Dox+Ex-treated hearts had a significant increase in GFP+ cells. Further analyses demonstrated these GFP+ BM cells had differentiated into vascular endothelial cells (GFP+CD31+) and pericytes (GFP+NG2+). Decreased cardiomyocytes were also seen in Dox-treated but not Dox+Ex-treated hearts. Ex induced an increase in GFP+c-Kit+ cells. However, these c-Kit+ BM stem cells had not differentiated into cardiomyocytes. Dox therapy induced phosphorylation of MST1/2, LATS1, and YAP; a decrease in total YAP; and cleavage of caspase-3 and PARP in the heart tissues. Dox+Ex prevented these effects. Our data demonstrated Dox-induced cardiotoxicity is mediated by vascular damage resulting in decreased cardiac blood flow and through activation of Hippo-YAP signaling resulting in cardiomyocyte apoptosis. Furthermore, Ex inhibited these effects by promoting migration of BM stem cells into the heart to repair the cardiac vessels damaged by Dox and through inhibiting Dox-induced Hippo-YAP signaling-mediated apoptosis. These data support the concept of using exercise as an intervention to decrease Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rong-Hua Tao
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| | - Masato Kobayashi
- School of Health Sciences, Institutes of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-0942, Japan;
| | - Yuanzheng Yang
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Eugenie S. Kleinerman
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| |
Collapse
|
12
|
Elde S, Wang H, Woo YJ. Navigating the Crossroads of Cell Therapy and Natural Heart Regeneration. Front Cell Dev Biol 2021; 9:674180. [PMID: 34046410 PMCID: PMC8148343 DOI: 10.3389/fcell.2021.674180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite significant advances in our understanding of the disease and its treatment. Consequently, the therapeutic potential of cell therapy and induction of natural myocardial regeneration have stimulated a recent surge of research and clinical trials aimed at addressing this challenge. Recent developments in the field have shed new light on the intricate relationship between inflammation and natural regeneration, an intersection that warrants further investigation.
Collapse
Affiliation(s)
- Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
13
|
Jiang M, Hu H, Zhao K, Di R, Huang X, Shi Y, Yue Y, Nie J, Yu S, Wang W, Yang Z. The G4 resolvase RHAU modulates mRNA translation and stability to sustain postnatal heart function and regeneration. J Biol Chem 2021; 296:100080. [PMID: 33199370 PMCID: PMC7948451 DOI: 10.1074/jbc.ra120.014948] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Post-transcriptional regulation of mRNA translation and stability is primarily achieved by RNA-binding proteins, which are of increasing importance for heart function. Furthermore, G-quadruplex (G4) and G4 resolvase activity are involved in a variety of biological processes. However, the role of G4 resolvase activity in heart function remains unknown. The present study aims to investigate the role of RNA helicase associated with adenylate- and uridylate-rich element (RHAU), an RNA-binding protein with G4 resolvase activity in postnatal heart function through deletion of Rhau in the cardiomyocytes of postnatal mice. RHAU-deficient mice displayed progressive pathological remodeling leading to heart failure and mortality and impaired neonatal heart regeneration. RHAU ablation reduced the protein levels but enhanced mRNA levels of Yap1 and Hexim1 that are important regulators for heart development and postnatal heart function. Furthermore, RHAU was found to associate with both the 5' and 3' UTRs of these genes to destabilize mRNA and enhance translation. Thus, we have demonstrated the important functions of RHAU in the dual regulation of mRNA translation and stability, which is vital for heart physiology.
Collapse
Affiliation(s)
- Mingyang Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Han Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ke Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Ruomin Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xinyi Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yingchao Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yunyun Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Junwei Nie
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Shan Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China.
| |
Collapse
|
14
|
VGLL4 Protects against Oxidized-LDL-Induced Endothelial Cell Dysfunction and Inflammation by Activating Hippo-YAP/TEAD1 Signaling Pathway. Mediators Inflamm 2020; 2020:8292173. [PMID: 33456372 PMCID: PMC7787722 DOI: 10.1155/2020/8292173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Vestigial-like 4 (VGLL4) has been found to have multiple functions in tumor development; however, its role in cardiovascular disease is unknown. The aim of this study was to investigate the effect of VGLL4 on the dysfunction and inflammatory response of Ox-LDL-induced human umbilical vein endothelial cells (HUVECs) and its mechanism, so as to provide a new theoretical basis for the diagnosis and treatment of atherosclerosis. In the present study, the protective activity of VGLL4 inhibiting Ox-LDL-induced apoptosis, oxidative stress, inflammation, and injury as well as its molecular mechanisms was examined using human umbilical vein endothelial cells (HUVECs). The results showed that the expression of VGLL4 was decreased with the increase of Ox-LDL concentration in HUVECs. In addition, the functional study found that VGLL4 overexpression alleviated Ox-LDL-induced oxidative stress, inflammation, and dysfunction and inhibited apoptosis. Further research found that VGLL4 regulated Hippo-YAP/TEAD1 signaling pathway, and the Hippo-YAP/TEAD1 signaling pathway was involved in the protective mechanism of VGLL4 on HUVECs. In conclusion, it suggests that VGLL4 protects against oxidized-LDL-induced endothelial cell dysfunction by activating the Hippo-YAP/TEAD1 signaling pathway.
Collapse
|
15
|
Tabrizi A, Soori R, Choobineh S, Gholipour M. Aerobic Training-induced Upregulation of YAP1 and Prevention of Cardiac Pathological Hypertrophy in Male Rats. Int J Prev Med 2020; 11:119. [PMID: 33088447 PMCID: PMC7554557 DOI: 10.4103/ijpvm.ijpvm_356_19] [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: 10/02/2019] [Accepted: 02/21/2020] [Indexed: 11/04/2022] Open
Abstract
Background Pathological hypertrophy is one of the negative consequences of cardiac sympathetic hyperactivity. Recent studies have shown that YAP1 plays a critical role in cardiomyocytes hypertrophy. Considering the preventive role of exercise training in cardiovascular diseases, the present study was conducted to examine the effect of aerobic exercise training on YAP1 gene expression and its upstream components. Methods Eighteen male Wistar rats were randomly divided into aerobic training and control groups. Aerobic training was performed one hour/day, five days per week, for eight weeks, on a treadmill at 65-75% VO2 max. Pathological hypertrophy was induced by injecting 3 mg/kg-1 of isoproterenol for seven days. The left ventricle was separated, and YAP1, 3-mercaptopyruvate sulfurtransferase (MST), large tumor suppressor (LATS), and mitogen-activated protein 4 kinase (MAP4K) gene expressions were assessed and YAP1 protein levels were also assessed by western blotting. Cell apoptosis was detected by TUNEL assays. The between-group differences were evaluated using the T-test and P value <0.05 was considered statistically significant. Results There were no significant between-group differences in MST gene expression (P = 0.061); meanwhile, in the training group, LATS and Map4K expressions were suppressed, followed by a significant increase in YAP1 expression (P < 0.001). Compared to the control group, the left ventricular weight increased significantly in the training group while the cardiomyocyte apoptosis decreased. Conclusions The results showed that, by reducing LATS, aerobic training-induced YAP1 upregulation can help prevent the propagation of cardiomyocyte apoptosis due to pathological conditions.
Collapse
Affiliation(s)
- Arezoo Tabrizi
- Department of Physical Education, Sharif University of Technology Tehran, Iran
| | - Rahman Soori
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences. University of Tehran, Tehran, Iran
| | - Siroos Choobineh
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences. University of Tehran, Tehran, Iran
| | - Majid Gholipour
- Department of Physical Education, Sharif University of Technology Tehran, Iran
| |
Collapse
|
16
|
Guo H, Lu YW, Lin Z, Huang ZP, Liu J, Wang Y, Seok HY, Hu X, Ma Q, Li K, Kyselovic J, Wang Q, Lin JLC, Lin JJC, Cowan DB, Naya F, Chen Y, Pu WT, Wang DZ. Intercalated disc protein Xinβ is required for Hippo-YAP signaling in the heart. Nat Commun 2020; 11:4666. [PMID: 32938943 PMCID: PMC7494909 DOI: 10.1038/s41467-020-18379-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Intercalated discs (ICD), specific cell-to-cell contacts that connect adjacent cardiomyocytes, ensure mechanical and electrochemical coupling during contraction of the heart. Mutations in genes encoding ICD components are linked to cardiovascular diseases. Here, we show that loss of Xinβ, a newly-identified component of ICDs, results in cardiomyocyte proliferation defects and cardiomyopathy. We uncovered a role for Xinβ in signaling via the Hippo-YAP pathway by recruiting NF2 to the ICD to modulate cardiac function. In Xinβ mutant hearts levels of phosphorylated NF2 are substantially reduced, suggesting an impairment of Hippo-YAP signaling. Cardiac-specific overexpression of YAP rescues cardiac defects in Xinβ knock-out mice—indicating a functional and genetic interaction between Xinβ and YAP. Our study reveals a molecular mechanism by which cardiac-expressed intercalated disc protein Xinβ modulates Hippo-YAP signaling to control heart development and cardiac function in a tissue specific manner. Consequently, this pathway may represent a therapeutic target for the treatment of cardiovascular diseases. Intercalated discs ensure mechanical and electrochemical coupling during contraction of the heart. Here, the authors show that loss of Xinβ results in cardiomyocyte proliferation defects and cardiomyopathy by influencing the Hippo-YAP signalling pathway, thus affecting cardiac development and function.
Collapse
Affiliation(s)
- Haipeng Guo
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA.,Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA.,Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA.,Department of Cardiology, Center for Translational Medicine, The First Affiliated Hospital, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Yi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Hee Young Seok
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA.,Institute of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Qing Ma
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Kathryn Li
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Jan Kyselovic
- Department of Internal Medicine, Faculty of Medicine, Comenius University, Ruzinovska 6, 826 06, Bratislava, Slovak Republic
| | - Qingchuan Wang
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 20215, USA
| | - Jenny L-C Lin
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Jim J-C Lin
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA
| | - Francisco Naya
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Yuguo Chen
- Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, 02115, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
17
|
Desgres M, Menasché P. Clinical Translation of Pluripotent Stem Cell Therapies: Challenges and Considerations. Cell Stem Cell 2020; 25:594-606. [PMID: 31703770 DOI: 10.1016/j.stem.2019.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the clinical outcomes of cell therapy trials have not met initial expectations, emerging evidence suggests that injury-mediated tissue damage might benefit from the delivery of cells or their secreted products. Pluripotent stem cells (PSCs) are promising cell sources primarily because of their capacity to generate stage- and lineage-specific differentiated derivatives. However, they carry inherent challenges for safe and efficacious clinical translation. This Review describes completed or ongoing trials of PSCs, discusses their potential mechanisms of action, and considers how to address the challenges required for them to become a major therapy, using heart repair as a case study.
Collapse
Affiliation(s)
- Manon Desgres
- Université de Paris, PARCC, INSERM, 75015 Paris, France
| | - Philippe Menasché
- Université de Paris, PARCC, INSERM, 75015 Paris, France; Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
18
|
Cardiac regeneration as an environmental adaptation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118623. [DOI: 10.1016/j.bbamcr.2019.118623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
19
|
Cardiac cell therapy: Current status, challenges and perspectives. Arch Cardiovasc Dis 2020; 113:285-292. [PMID: 32171698 DOI: 10.1016/j.acvd.2020.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/08/2020] [Indexed: 12/28/2022]
Abstract
Although the initial clinical trials of cardiac cell therapy have failed to demonstrate unequivocal clinical benefits, the accumulation of preclinical data gathered in parallel can now help us to understand the main causes of failures, while providing mechanistic insights that may be leveraged to improve the outcomes of subsequent clinical studies using cells or their secreted products. This review briefly describes the current status of clinical trials, discusses the potential mechanisms of action of the grafted cells, and the impact of this knowledge on the design of future studies, and finally draws some perspectives.
Collapse
|
20
|
Antiapoptosis and Antifibrosis Effects of Qishen Granules on Heart Failure Rats via Hippo Pathway. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1642575. [PMID: 31915683 PMCID: PMC6930732 DOI: 10.1155/2019/1642575] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/07/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
Qishen granules (QSG) are a famous formula with cardioprotective properties to heart failure (HF). The aim of this study was to investigate the underlying mechanism of QSG on apoptosis and fibrosis in the treatment of HF. HF model was induced by left anterior descending artery ligation on Sprague-Dawley rats. Transcriptome analysis was used to investigate the regulatory pathways of QSG on HF. Interestingly, downregulated genes of QSG were significantly enriched in Hippo pathway which plays a crucial role in regulating cell apoptosis and proliferation. We found that QSG inhibited the expressions of proapoptotic key proteins P-53 and fibrosis-related proteins TGF-β1, SMAD3, and CTGF. Further, we conducted research on the key proteins in the Hippo pathway upstream of CTGF and P-53. The results showed that MST1, P-MST1, P-LATS1, and RASSF1A that exert proapoptotic function were downregulated after QSG intervention. Similarly, P-YAP and P-TAZ, mediating self-degradation and apoptosis, were both observably decreased after QSG administration. Taken together, QSG are shown to be likely to exert cardioprotective effects by inhibiting the progression of apoptosis and fibrosis through Hippo pathway.
Collapse
|
21
|
Camberos V, Baio J, Bailey L, Hasaniya N, Lopez LV, Kearns-Jonker M. Effects of Spaceflight and Simulated Microgravity on YAP1 Expression in Cardiovascular Progenitors: Implications for Cell-Based Repair. Int J Mol Sci 2019; 20:E2742. [PMID: 31167392 PMCID: PMC6600678 DOI: 10.3390/ijms20112742] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 01/06/2023] Open
Abstract
Spaceflight alters many processes of the human body including cardiac function and cardiac progenitor cell behavior. The mechanism behind these changes remains largely unknown; however, simulated microgravity devices are making it easier for researchers to study the effects of microgravity. To study the changes that take place in cardiac progenitor cells in microgravity environments, adult cardiac progenitor cells were cultured aboard the International Space Station (ISS) as well as on a clinostat and examined for changes in Hippo signaling, a pathway known to regulate cardiac development. Cells cultured under microgravity conditions, spaceflight-induced or simulated, displayed upregulation of downstream genes involved in the Hippo pathway such as YAP1 and SOD2. YAP1 is known to play a role in cardiac regeneration which led us to investigate YAP1 expression in a sheep model of cardiovascular repair. Additionally, to mimic the effects of microgravity, drug treatment was used to induce Hippo related genes as well as a regulator of the Hippo pathway, miRNA-302a. These studies provide insight into the changes that occur in space and how the effects of these changes relate to cardiac regeneration studies.
Collapse
Affiliation(s)
- Victor Camberos
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Jonathan Baio
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Leonard Bailey
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Nahidh Hasaniya
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Larry V Lopez
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
22
|
Khan K, Makhoul G, Yu B, Schwertani A, Cecere R. The cytoprotective impact of yes-associated protein 1 after ischemia-reperfusion injury in AC16 human cardiomyocytes. Exp Biol Med (Maywood) 2019; 244:802-812. [PMID: 31142144 DOI: 10.1177/1535370219851243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Hippo-signaling pathway is a mechanism implicated in cardiomyocyte cytoprotection and regeneration after a myocardial infarction. Yes-associated protein 1, the main effector protein of this pathway, acts as a co-transcriptional activator to promote cardiomyocyte proliferation and survival. However, the biological mechanisms by which yes-associated protein 1 protects the heart post-MI are currently unknown. Here, we propose that yes-associated protein 1 plays a critical role in cardiomyocyte cytoprotection after simulated ischemia-reperfusion injury. AC16 human cardiomyocytes were infected with lentiviral plasmids containing normal human yes-associated protein 1 and a constitutively active form of YAP, YAP1S127A. Cells were exposed to ischemia-reperfusion injury using a hypoxic chamber. Hippo-signaling characterization after ischemia-reperfusion injury was performed via Western blotting and reverse transcriptase polymerase chain reaction. Cell viability, apoptosis, and cellular hypertrophy were assessed as a measure of cytoprotection. The GSK3β inhibitor CHIR99021 was used to investigate cross-talk between Hippo and Wnt-signaling and their role in cytoprotection after ischemia-reperfusion-injury. Ischemia-reperfusion injury resulted in significant decreased expression of the non-phosphorylated Hippo signaling kinases MST1 and LATS1, along with decreased expression of YAP/TAZ. Overexpression of yes-associated protein 1 improved cellular viability, while reducing hypertrophy and apoptosis via the ATM/ATR DNA damage response pathway. Activation of β-catenin in YAP-infected cardiomyocytes synergistically reduced cellular hypertrophy after ischemia-reperfusion-injury. Our findings indicate that yes-associated protein 1 is cytoprotective in AC16 human cardiomyocytes after ischemia-reperfusion injury, which may be mediated by co-activation of the canonical Wnt/β-catenin pathway. Thus, activation of yes-associated protein 1 may be a novel therapeutic to repair the infarcted myocardium. Impact statement Genetically engineering the cells of the heart after myocardial infarction to display a more regenerative phenotype is a promising therapy for heart failure patients. Here, we support a regenerative role for yes-associated protein 1, the main effector protein of the Hippo signaling pathway, in AC16 human cardiomyocytes as a potential therapeutic gene target for cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Kashif Khan
- Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec H4AJ1, Canada
| | - Georges Makhoul
- Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec H4AJ1, Canada
| | - Bin Yu
- Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec H4AJ1, Canada
| | - Adel Schwertani
- Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec H4AJ1, Canada
| | - Renzo Cecere
- Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec H4AJ1, Canada
| |
Collapse
|
23
|
Abstract
After decades of directed research, no effective regenerative therapy is currently available to repair the injured human heart. The epicardium, a layer of mesothelial tissue that envelops the heart in all vertebrates, has emerged as a new player in cardiac repair and regeneration. The epicardium is essential for muscle regeneration in the zebrafish model of innate heart regeneration, and the epicardium also participates in fibrotic responses in mammalian hearts. This structure serves as a source of crucial cells, such as vascular smooth muscle cells, pericytes, and fibroblasts, during heart development and repair. The epicardium also secretes factors that are essential for proliferation and survival of cardiomyocytes. In this Review, we describe recent advances in our understanding of the biology of the epicardium and the effect of these findings on the candidacy of this structure as a therapeutic target for heart repair and regeneration.
Collapse
Affiliation(s)
- Jingli Cao
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
- Regeneration Next, Duke University, Durham, NC, USA.
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA.
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
- Regeneration Next, Duke University, Durham, NC, USA.
| |
Collapse
|
24
|
Abstract
Death of adult cardiac myocytes and supportive tissues resulting from cardiovascular diseases such as myocardial infarction is the proximal driver of pathological ventricular remodeling that often culminates in heart failure. Unfortunately, no currently available therapeutic barring heart transplantation can directly replenish myocytes lost from the injured heart. For decades, the field has struggled to define the intrinsic capacity and cellular sources for endogenous myocyte turnover in pursuing more innovative therapeutic strategies aimed at regenerating the injured heart. Although controversy persists to this day as to the best therapeutic regenerative strategy to use, a growing consensus has been reached that the very limited capacity for new myocyte formation in the adult mammalian heart is because of proliferation of existing cardiac myocytes but not because of the activity of an endogenous progenitor cell source of some sort. Hence, future therapeutic approaches should take into consideration the fundamental biology of myocyte renewal in designing strategies to potentially replenish these cells in the injured heart.
Collapse
Affiliation(s)
| | - Jeffery D Molkentin
- From the Department of Pediatrics (R.J.V., J.D.M.)
- Howard Hughes Medical Institute (J.D.M.)
| | - Steven R Houser
- Cincinnati Children's Hospital Medical Center, OH; and the Lewis Katz School of Medicine, Cardiovascular Research Center, Temple University, Philadelphia, PA (S.R.H.)
| |
Collapse
|
25
|
El-Nachef D, Oyama K, Wu YY, Freeman M, Zhang Y, MacLellan WR. Repressive histone methylation regulates cardiac myocyte cell cycle exit. J Mol Cell Cardiol 2018; 121:1-12. [PMID: 29800554 DOI: 10.1016/j.yjmcc.2018.05.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 02/07/2023]
Abstract
Mammalian cardiac myocytes (CMs) stop proliferating soon after birth and subsequent heart growth comes from hypertrophy, limiting the adult heart's regenerative potential after injury. The molecular events that mediate CM cell cycle exit are poorly understood. To determine the epigenetic mechanisms limiting CM cycling in adult CMs (ACMs) and whether trimethylation of lysine 9 of histone H3 (H3K9me3), a histone modification associated with repressed chromatin, is required for the silencing of cell cycle genes, we developed a transgenic mouse model where H3K9me3 is specifically removed in CMs by overexpression of histone demethylase, KDM4D. Although H3K9me3 is found across the genome, its loss in CMs preferentially disrupts cell cycle gene silencing. KDM4D binds directly to cell cycle genes and reduces H3K9me3 levels at these promotors. Loss of H3K9me3 preferentially leads to increased cell cycle gene expression resulting in enhanced CM cycling. Heart mass was increased in KDM4D overexpressing mice by postnatal day 14 (P14) and continued to increase until 9-weeks of age. ACM number, but not size, was significantly increased in KDM4D expressing hearts, suggesting CM hyperplasia accounts for the increased heart mass. Inducing KDM4D after normal development specifically in ACMs resulted in increased cell cycle gene expression and cycling. We demonstrated that H3K9me3 is required for CM cell cycle exit and terminal differentiation in ACMs. Depletion of H3K9me3 in adult hearts prevents and reverses permanent cell cycle exit and allows hyperplastic growth in adult hearts in vivo.
Collapse
Affiliation(s)
- Danny El-Nachef
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Kyohei Oyama
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Yun-Yu Wu
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Miles Freeman
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Yiqiang Zhang
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - W Robb MacLellan
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States.
| |
Collapse
|
26
|
Zhou W, Zhao M. How Hippo Signaling Pathway Modulates Cardiovascular Development and Diseases. J Immunol Res 2018; 2018:3696914. [PMID: 29577047 PMCID: PMC5822808 DOI: 10.1155/2018/3696914] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/12/2017] [Indexed: 01/26/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death around the globe. Cardiac deterioration is associated with irreversible cardiomyocyte loss. Understanding how the cardiovascular system develops and the pathological processes of cardiac disease will contribute to finding novel and preventive therapeutic methods. The canonical Hippo tumor suppressor pathway in mammalian cells is primarily composed of the MST1/2-SAV1-LATS1/2-MOB1-YAP/TAZ cascade. Continuing research on this pathway has identified other factors like RASSF1A, Nf2, MAP4Ks, and NDR1/2, further enriching our knowledge of the Hippo-YAP pathway. YAP, the core effecter of the Hippo pathway, may accumulate in the nucleus and initiate transcriptional activity if the pathway is inhibited. The role of Hippo signaling has been widely investigated in organ development and cancers. A heart of normal size and function which is critical for survival could not be generated without the proper regulation of the Hippo tumor suppressor pathway. Recent research has demonstrated a novel role of Hippo signaling in cardiovascular disease in the context of development, hypertrophy, angiogenesis, regeneration, apoptosis, and autophagy. In this review, we summarize the current knowledge of how Hippo signaling modulates pathological processes in cardiovascular disease and discuss potential molecular therapeutic targets.
Collapse
Affiliation(s)
- Wenyi Zhou
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
- Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510000, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
27
|
Understanding the role of mammalian sterile 20-like kinase 1 (MST1) in cardiovascular disorders. J Mol Cell Cardiol 2018; 114:141-149. [DOI: 10.1016/j.yjmcc.2017.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
|
28
|
Landry NM, Cohen S, Dixon IMC. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res Cardiol 2017; 113:1. [PMID: 29101484 DOI: 10.1007/s00395-017-0659-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022]
Abstract
Tissue development and homeostasis are dependent upon the concerted synthesis, maintenance, and degradation of extracellular matrix (ECM) molecules. Cardiac fibrosis is now recognized as a primary contributor to incidence of heart failure, particularly heart failure with preserved ejection fraction, wherein cardiac filling in diastole is compromised. Periostin is a cell-associated protein involved in cell fate determination, proliferation, tumorigenesis, and inflammatory responses. As a non-structural component of the ECM, secreted 90 kDa periostin is emerging as an important matricellular factor in cardiac mesenchymal tissue development. In addition, periostin's role as a mediator in cell-matrix crosstalk has also garnered attention for its association with fibroproliferative diseases in the myocardium, and for its association with TGF-β/BMP signaling. This review summarizes the phylogenetic history of periostin, its role in cardiac development, and the major signaling pathways influencing its expression in cardiovascular pathology. Further, we provide a synthesis of the current literature to distinguish the multiple roles of periostin in cardiac health, development and disease. As periostin may be targeted for therapeutic treatment of cardiac fibrosis, these insights may shed light on the putative timing for application of periostin-specific therapies.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada
| | - Smadar Cohen
- Regenerative Medicine and Stem Cell Research Center, Ilse Katz Institute for Nanoscale Science and Technology, Beersheba, Israel.,Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada. .,Laboratory of Molecular Cardiology, St. Boniface Hospital Albrechtsen Research Centre, R3010-351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada.
| |
Collapse
|
29
|
Abstract
The heart has a markedly limited capacity for regeneration. Reporting in Nature, Bassat et al. (2017) and Morikawa et al. (2017) have uncovered a new mechanism of Yap inhibition by the dystrophin glycoprotein complex (DGC) that is released by the extracellular matrix protein Agrin in order to promote cardiac regeneration.
Collapse
|
30
|
Jeong MG, Song H, Shin JH, Jeong H, Kim HK, Hwang ES. Transcriptional coactivator with PDZ-binding motif is required to sustain testicular function on aging. Aging Cell 2017; 16:1035-1042. [PMID: 28613007 PMCID: PMC5595677 DOI: 10.1111/acel.12631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2017] [Indexed: 12/15/2022] Open
Abstract
Transcriptional coactivator with PDZ‐binding motif (TAZ) directly interacts with transcription factors and regulates their transcriptional activity. Extensive functional studies have shown that TAZ plays critical regulatory roles in stem cell proliferation, differentiation, and survival and also modulates the development of organs such as the lung, kidney, heart, and bone. Despite the importance of TAZ in stem cell maintenance, TAZ function has not yet been evaluated in spermatogenic stem cells of the male reproductive system. Here, we investigated the expression and functions of TAZ in mouse testis. TAZ was expressed in spermatogenic stem cells; however, its deficiency caused significant structural abnormalities, including atrophied tubules, widened interstitial space, and abnormal Leydig cell expansion, thereby resulting in lowered sperm counts and impaired fertility. Furthermore, TAZ deficiency increased the level of apoptosis and senescence in spermatogenic cells and Leydig cells upon aging. The expression of senescence‐associated β‐galactosidase (SA‐βgal), secretory phenotypes, and cyclin‐dependent kinase inhibitors (p16, p19, and p21) significantly increased in the absence of TAZ. TAZ downregulation in testicular cells further increased SA‐βgal and p21 expression induced by oxidative stress, whereas TAZ overexpression decreased p21 induction and prevented senescence. Mechanistic studies showed that TAZ suppressed DNA‐binding activity of p53 through a direct interaction and thus attenuated p53‐induced p21 gene transcription. Our results suggested that TAZ may suppress apoptosis and premature senescence in spermatogenic cells by inhibiting the p53‐p21 signaling pathway, thus playing important roles in the maintenance and control of reproductive function.
Collapse
Affiliation(s)
- Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hyuna Song
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Ji Hyun Shin
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hana Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hyo Kyeong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| |
Collapse
|
31
|
Wang J, Martin JF. Hippo Pathway: An Emerging Regulator of Craniofacial and Dental Development. J Dent Res 2017; 96:1229-1237. [PMID: 28700256 DOI: 10.1177/0022034517719886] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The evolutionarily conserved Hippo signaling pathway is a vital regulator of organ size that fine-tunes cell proliferation, apoptosis, and differentiation. A number of important studies have revealed critical roles of Hippo signaling and its effectors Yap (Yes-associated protein) and Taz (transcriptional coactivator with PDZ binding motif) in tissue development, homeostasis, and regeneration, as well as in tumorigenesis. In addition, recent studies have shown evidence of crosstalk between the Hippo pathway and other key signaling pathways, such as Wnt signaling, that not only regulates developmental processes but also contributes to disease pathogenesis. In this review, we summarize the major discoveries in the field of Hippo signaling and what has been learned about its regulation and crosstalk with other signaling pathways, with a particular focus on recent findings involving the Hippo-Yap pathway in craniofacial and tooth development. New and exciting studies of the Hippo pathway are anticipated that will significantly improve our understanding of the molecular mechanisms of human craniofacial and tooth development and disease and will ultimately lead to the development of new therapies.
Collapse
Affiliation(s)
- J Wang
- 1 Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - J F Martin
- 1 Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.,2 Texas Heart Institute, Houston, TX, USA
| |
Collapse
|
32
|
Cardiac stem cells for myocardial regeneration: promising but not ready for prime time. Curr Opin Biotechnol 2017; 47:30-35. [PMID: 28591641 DOI: 10.1016/j.copbio.2017.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 02/06/2023]
Abstract
Remarkable strides have been made in the treatment of ischemic heart disease in decades. As the initial loss of cardiomyocytes associated with myocardial infarction serves as an impetus for myocardial remodeling, the ability to replace these cells with healthy counterparts would represent an effective treatment for many forms of cardiovascular disease. The discovery of cardiac stem cells (that can differentiate into multiple lineages) highlighted the possibility for development of cell-based therapeutics to achieve this ultimate goal. Recent research features cardiac stem cell maintenance, proliferation, and differentiation, as well as direct reprogramming of various somatic cells into cardiomyocytes, all within the context of the holy grail of regeneration of the injured heart. Much work remains to be done, but the future looks bright!
Collapse
|
33
|
Galdos FX, Guo Y, Paige SL, VanDusen NJ, Wu SM, Pu WT. Cardiac Regeneration: Lessons From Development. Circ Res 2017; 120:941-959. [PMID: 28302741 DOI: 10.1161/circresaha.116.309040] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023]
Abstract
Palliative surgery for congenital heart disease has allowed patients with previously lethal heart malformations to survive and, in most cases, to thrive. However, these procedures often place pressure and volume loads on the heart, and over time, these chronic loads can cause heart failure. Current therapeutic options for initial surgery and chronic heart failure that results from failed palliation are limited, in part, by the mammalian heart's low inherent capacity to form new cardiomyocytes. Surmounting the heart regeneration barrier would transform the treatment of congenital, as well as acquired, heart disease and likewise would enable development of personalized, in vitro cardiac disease models. Although these remain distant goals, studies of heart development are illuminating the path forward and suggest unique opportunities for heart regeneration, particularly in fetal and neonatal periods. Here, we review major lessons from heart development that inform current and future studies directed at enhancing cardiac regeneration.
Collapse
Affiliation(s)
- Francisco X Galdos
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Yuxuan Guo
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sharon L Paige
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Nathan J VanDusen
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sean M Wu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - William T Pu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
34
|
Chimenti I, Massai D, Morbiducci U, Beltrami AP, Pesce M, Messina E. Stem Cell Spheroids and Ex Vivo Niche Modeling: Rationalization and Scaling-Up. J Cardiovasc Transl Res 2017; 10:150-166. [PMID: 28289983 DOI: 10.1007/s12265-017-9741-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Improved protocols/devices for in vitro culture of 3D cell spheroids may provide essential cues for proper growth and differentiation of stem/progenitor cells (S/PCs) in their niche, allowing preservation of specific features, such as multi-lineage potential and paracrine activity. Several platforms have been employed to replicate these conditions and to generate S/PC spheroids for therapeutic applications. However, they incompletely reproduce the niche environment, with partial loss of its highly regulated network, with additional hurdles in the field of cardiac biology, due to debated resident S/PCs therapeutic potential and clinical translation. In this contribution, the essential niche conditions (metabolic, geometric, mechanical) that allow S/PCs maintenance/commitment will be discussed. In particular, we will focus on both existing bioreactor-based platforms for the culture of S/PC as spheroids, and on possible criteria for the scaling-up of niche-like spheroids, which could be envisaged as promising tools for personalized cardiac regenerative medicine, as well as for high-throughput drug screening.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Rome, Italy
| | - Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino", IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry, "Umberto I" Hospital, "La Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
35
|
Pfleger CM. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr Top Dev Biol 2017; 123:181-228. [PMID: 28236967 DOI: 10.1016/bs.ctdb.2016.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Hippo Pathway is a master regulatory network that regulates proliferation, cell growth, stemness, differentiation, and cell death. Coordination of these processes by the Hippo Pathway throughout development and in mature organisms in response to diverse external and internal cues plays a role in morphogenesis, in controlling organ size, and in maintaining organ homeostasis. Given the importance of these processes, the Hippo Pathway also plays an important role in organismal health and has been implicated in a variety of diseases including eye disease, cardiovascular disease, neurodegeneration, and cancer. This review will focus on Drosophila reports that identified the core components of the Hippo Pathway revealing specific downstream biological outputs of this complicated network. A brief description of mammalian reports will complement review of the Drosophila studies. This review will also survey upstream regulation of the core components with a focus on feedback mechanisms.
Collapse
Affiliation(s)
- Cathie M Pfleger
- The Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
36
|
Abstract
In this review, we focus on new approaches that could lead to the regeneration of heart muscle and the restoration of cardiac muscle function derived from newly-formed cardiomyocytes. Various strategies for the production of cardiomyocytes from embryonic stem cells, induced pluripotent stem cells, adult bone marrow stem cells and cardiac spheres from human heart biopsies are described. Pathological conditions which lead to atherosclerosis and coronary artery disease often are followed by myocardial infarction causing myocardial cell death. After cell death, there is very little self-regeneration of the cardiac muscle tissue, which is replaced by non-contractile connective tissue, thus weakening the ability of the heart muscle to contract fully and leading to heart failure. A number of experimental research approaches to stimulate heart muscle regeneration with the hope of regaining normal or near normal heart function in the damaged heart muscle have been attempted. Some of these very interesting studies have used a variety of stem cell types in combination with potential cardiogenic differentiation factors in an attempt to promote differentiation of new cardiac muscle for possible future use in the clinical treatment of patients who have suffered heart muscle damage from acute myocardial infarctions or related cardiovascular diseases. Although progress has been made in recent years relative to promoting the differentiation of cardiac muscle tissue from non-muscle cells, much work remains to be done for this technology to be used routinely in translational clinical medicine to treat patients with damaged heart muscle tissue and return such individuals to pre-heart-attack activity levels.
Collapse
Affiliation(s)
- Andrei Kochegarov
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| | - Larry F Lemanski
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| |
Collapse
|
37
|
Pesce M, Messina E, Chimenti I, Beltrami AP. Cardiac Mechanoperception: A Life-Long Story from Early Beats to Aging and Failure. Stem Cells Dev 2016; 26:77-90. [PMID: 27736363 DOI: 10.1089/scd.2016.0206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The life-long story of the heart starts concomitantly with primary differentiation events occurring in multipotent progenitors located in the so-called heart tube. This initially tubular structure starts a looping process, which leads to formation of the final four-chambered heart with a primary contribution of geometric and position-associated cell sensing. While this establishes the correct patterning of the final cardiac structure, it also provides feedbacks to fundamental cellular machineries controlling proliferation and differentiation, thus ensuring a coordinated restriction of cell growth and a myocyte terminal differentiation. Novel evidences provided by embryological and cell engineering studies have clarified the relevance of mechanics-supported position sensing for the correct recognition of cell fate inside developing embryos and multicellular aggregates. One of the main components of this pathway, the Hippo-dependent signal transduction machinery, is responsible for cell mechanics intracellular transduction with important consequences for gene transcription and cell growth control. Being the Hippo pathway also directly connected to stress responses and altered metabolism, it is tempting to speculate that permanent alterations of mechanosensing may account for modifying self-renewal control in tissue homeostasis. In the present contribution, we translate these concepts to the aging process and the failing of the human heart, two pathophysiologic conditions that are strongly affected by stress responses and altered metabolism.
Collapse
Affiliation(s)
- Maurizio Pesce
- 1 Tissue Engineering Research Unit, Centro Cardiologico Monzino, IRCCS , Milan, Italy
| | - Elisa Messina
- 2 Department of Pediatric Cardiology, "Sapienza" University , Rome, Italy
| | - Isotta Chimenti
- 3 Department of Medical Surgical Science and Biotechnology, "Sapienza" University , Rome, Italy
| | | |
Collapse
|
38
|
Cardiomyocyte proliferation in zebrafish and mammals: lessons for human disease. Cell Mol Life Sci 2016; 74:1367-1378. [PMID: 27812722 PMCID: PMC5357290 DOI: 10.1007/s00018-016-2404-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes proliferate profusely during early development and for a brief period after birth in mammals. Within a month after birth, this proliferative capability is dramatically reduced in mammals unlike lower vertebrates where it persists into adult life. The zebrafish, for example, retains the ability to regenerate the apex of the heart following resection by a mechanism predominantly driven by cardiomyocyte proliferation. Differences in proliferative capacity of cardiomyocytes in adulthood between mammals and lower vertebrates are closely liked to ontogenetic or phylogenetic factors. Elucidation of these factors has the potential to provide enormous benefits if they lead to the development of therapeutic strategies that facilitate cardiomyocyte proliferation. In this review, we highlight the differences between Mammalian and Zebrafish cardiomyocytes, which could explain at least in part the different proliferative capacities in these two species. We discuss the advantages of the zebrafish as a model of cardiomyocyte proliferation, particularly at the embryonic stage. We also identify a number of key molecular pathways with potential to reveal key steps in switching cardiomyocytes from a quiescent to a proliferative phenotype.
Collapse
|
39
|
Targeting the Hippo Signaling Pathway for Tissue Regeneration and Cancer Therapy. Genes (Basel) 2016; 7:genes7090055. [PMID: 27589805 PMCID: PMC5042386 DOI: 10.3390/genes7090055] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
The Hippo signaling pathway is a highly-conserved developmental pathway that plays an essential role in organ size control, tumor suppression, tissue regeneration and stem cell self-renewal. The YES-associated protein (YAP) and the transcriptional co-activator with PDZ-binding motif (TAZ) are two important transcriptional co-activators that are negatively regulated by the Hippo signaling pathway. By binding to transcription factors, especially the TEA domain transcription factors (TEADs), YAP and TAZ induce the expression of growth-promoting genes, which can promote organ regeneration after injury. Therefore, controlled activation of YAP and TAZ can be useful for regenerative medicine. However, aberrant activation of YAP and TAZ due to deregulation of the Hippo pathway or overexpression of YAP/TAZ and TEADs can promote cancer development. Hence, pharmacological inhibition of YAP and TAZ may be a useful approach to treat tumors with high YAP and/or TAZ activity. In this review, we present the mechanisms regulating the Hippo pathway, the role of the Hippo pathway in tissue repair and cancer, as well as a detailed analysis of the different strategies to target the Hippo signaling pathway and the genes regulated by YAP and TAZ for regenerative medicine and cancer therapy.
Collapse
|
40
|
Peng X, He Q, Li G, Ma J, Zhong TP. Rac1-PAK2 pathway is essential for zebrafish heart regeneration. Biochem Biophys Res Commun 2016; 472:637-42. [PMID: 26966072 DOI: 10.1016/j.bbrc.2016.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/05/2016] [Indexed: 01/08/2023]
Abstract
P-21 activated kinases, or PAKs, are serine-threonine kinases that play important roles in diverse heart functions include heart development, cardiovascular development and function in a range of models; however, the mechanisms by which PAKs mediate heart regeneration are unknown. Here, we demonstrate that PAK2 and PAK4 expression is induced in cardiomyocytes and vessels, respectively, following zebrafish heart injury. Inhibition of PAK2 and PAK4 using a specific small molecule inhibitor impedes cardiomyocyte proliferation/dedifferentiation and cardiovascular regeneration, respectively. Cdc42 is specifically expressed in the ventricle and may function upstream of PAK2 but not PAK4 under normal conditions and that cardiomyocyte proliferentation during heart regeneration relies on Rac1-mediated activation of Pak2. Our results indicate that PAKs play a key role in heart regeneration.
Collapse
Affiliation(s)
- Xiangwen Peng
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Quanze He
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Jiangsu 215002, China
| | - Guobao Li
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Jinmin Ma
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China
| | - Tao P Zhong
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 201203, China; Department of Medicine, Vanderbilt University School of Medicine, TN 37232, USA.
| |
Collapse
|
41
|
Gabriel BM, Hamilton DL, Tremblay AM, Wackerhage H. The Hippo signal transduction network for exercise physiologists. J Appl Physiol (1985) 2016; 120:1105-17. [PMID: 26940657 DOI: 10.1152/japplphysiol.01076.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/20/2022] Open
Abstract
The ubiquitous transcriptional coactivators Yap (gene symbol Yap1) and Taz (gene symbol Wwtr1) regulate gene expression mainly by coactivating the Tead transcription factors. Being at the center of the Hippo signaling network, Yap and Taz are regulated by the Hippo kinase cassette and additionally by a plethora of exercise-associated signals and signaling modules. These include mechanotransduction, the AKT-mTORC1 network, the SMAD transcription factors, hypoxia, glucose homeostasis, AMPK, adrenaline/epinephrine and angiotensin II through G protein-coupled receptors, and IL-6. Consequently, exercise should alter Hippo signaling in several organs to mediate at least some aspects of the organ-specific adaptations to exercise. Indeed, Tead1 overexpression in muscle fibers has been shown to promote a fast-to-slow fiber type switch, whereas Yap in muscle fibers and cardiomyocytes promotes skeletal muscle hypertrophy and cardiomyocyte adaptations, respectively. Finally, genome-wide association studies in humans have linked the Hippo pathway members LATS2, TEAD1, YAP1, VGLL2, VGLL3, and VGLL4 to body height, which is a key factor in sports.
Collapse
Affiliation(s)
- Brendan M Gabriel
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, Scotland, UK; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, University of Copenhagen, Denmark; and Integrative physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Annie M Tremblay
- Stem Cell Program, Children's Hospital, Boston, Massachusetts; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Henning Wackerhage
- School of Medicine, Dentistry and Nutrition, University of Aberdeen, Scotland, UK; Faculty of Sport and Health Science, Technical University Munich, Germany;
| |
Collapse
|
42
|
JUDD J, XUAN W, HUANG GN. Cellular and molecular basis of cardiac regeneration. Turk J Biol 2016. [DOI: 10.3906/biy-1504-43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
43
|
Giacca M, Zacchigna S. Harnessing the microRNA pathway for cardiac regeneration. J Mol Cell Cardiol 2015; 89:68-74. [PMID: 26431632 DOI: 10.1016/j.yjmcc.2015.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/28/2015] [Accepted: 09/28/2015] [Indexed: 10/23/2022]
Abstract
Mounting evidence over the last few years has indicated that the rate of cardiomyocyte proliferation, and thus the extent of cardiac renewal, is under the control of the microRNA network. Several microRNAs (e.g. miR-1) regulate expansion of the cardiomyocyte pool and its terminal differentiation during the embryonic life; some not only promote cardiomyocyte proliferation but also their de-differentiation towards an embryonic cell phenotype (e.g. the miR-302/367 cluster); a few others are involved in the repression of cardiomyocyte proliferation occurring suddenly after birth (e.g. the miR-15 family); others again are not physiologically involved in the regulation of cardiomyocyte turnover, but nevertheless are able to promote cardiomyocyte proliferation and cardiac regeneration when delivered exogenously (e.g. miR-199a-3p). With a few exceptions, the molecular mechanisms underlying the pro-proliferative effect of these microRNAs, most of which appear to act at the level of already differentiated cardiomyocytes, remain to be thoroughly elucidated. The possibility of harnessing the miRNA network to achieve cardiac regeneration paves the way to exciting therapeutic applications. This could be achieved by either administering miRNA mimics or inhibitors, or transducing the heart with viral vectors expressing miRNA-encoding genes.
Collapse
Affiliation(s)
- Mauro Giacca
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy.
| | - Serena Zacchigna
- Cardiovascular Biology Laboratories, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, Padriciano 99, 34149 Trieste, Italy.
| |
Collapse
|
44
|
RNA mimics as therapeutics for cardiac regeneration: a paradigm shift. Mol Ther 2015; 23:984-986. [PMID: 26022627 DOI: 10.1038/mt.2015.86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|