101
|
Chen P, Zhou C, Li B, Yang C. Circular RNA MGAT1 regulates cell proliferation and apoptosis in hypoxia-induced cardiomyocytes through miR-34a/YAP1 axis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2474-2486. [PMID: 33165436 PMCID: PMC7642720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Congenital heart disease (CHD) has severe morbidity and mortality worldwide. Evidence suggests that circularRNAs (circRNAs) are involved in the pathogenesis of human CHD. However, the regulatory mechanism remains uncertain. This study aimed to explore that mechanism. The levels of circular RNA MGAT1 (circMGAT1) and miR-34a were measured by quantitative polymerase chain reaction (qRT-PCR). Expression of yes-associated protein isoform 1 (YAP1) was assessed by western blot. Caspase-3 activity was evaluated by Caspase 3 Activity Assay Kit. CCK-8 assay was carried out to detect cell proliferation of hypoxia-induced AC16 cells. Cell apoptosis was analyzed by flow cytometry. In addition, dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were performed to verify the relationship between miR-34a and circMGAT1 or YAP1 in vitro. The level of circMGAT1 was downregulated, while miR-34a was strikingly increased in CHD tissues and hypoxia-induced AC16 cells. CircMGAT1 was a sponge of miR-34a, and circMGAT1 targeted miR-34a to regulate cell proliferation and apoptosis in hypoxia-induced cardiomyocytes. Dual-luciferase reporter and RIP-assay verified that miR-34a directly targeted YAP1, and the expression of YAP1 was significantly suppressed by miR-34a mimics but was enhanced by miR-34a inhibitor. Interestingly, YAP1 restored the effect of miR-34a on cell proliferation and apoptosis in hypoxia-induced AC16 cells. Besides, circMGAT1 sponged miR-34a to regulate the expression of YAP1. In conclusion, circMGAT1 inhibited cell apoptosis and enhanced cell proliferation by regulating the miR-34a/YAP1 axis, providing a therapy target for the treatment of human CHD.
Collapse
Affiliation(s)
- Pengyuan Chen
- Department of Pediatrics, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Chaoran Zhou
- Department of Pediatrics, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Bo Li
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| | - Chao Yang
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science/Sichuan Provincial People’s HospitalChengdu, Sichuan, China
| |
Collapse
|
102
|
Francisco J, Zhang Y, Jeong JI, Mizushima W, Ikeda S, Ivessa A, Oka S, Zhai P, Tallquist MD, Del Re DP. Blockade of Fibroblast YAP Attenuates Cardiac Fibrosis and Dysfunction Through MRTF-A Inhibition. JACC Basic Transl Sci 2020; 5:931-945. [PMID: 33015415 PMCID: PMC7524792 DOI: 10.1016/j.jacbts.2020.07.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 10/29/2022]
Abstract
Fibrotic remodeling of the heart in response to injury contributes to heart failure, yet therapies to treat fibrosis remain elusive. Yes-associated protein (YAP) is activated in cardiac fibroblasts by myocardial infarction, and genetic inhibition of fibroblast YAP attenuates myocardial infarction-induced cardiac dysfunction and fibrosis. YAP promotes myofibroblast differentiation and associated extracellular matrix gene expression through engagement of TEA domain transcription factor 1 and subsequent de novo expression of myocardin-related transcription factor A. Thus, fibroblast YAP is a promising therapeutic target to prevent fibrotic remodeling and heart failure.
Collapse
Key Words
- AngII, angiotensin II
- Hippo signaling
- MCM, Mer-Cre-Mer
- MI, myocardial infarction
- MRTF-A, myocardin-related transcription factor A
- Mkl1, megakaryoblastic leukemia 1
- NRCF, neonatal rat cardiac fibroblast
- PDGFR, platelet-derived growth factor receptor
- PE, phenylephrine
- SMA, smooth muscle actin
- TEAD, TEA domain transcription factor
- TGF, transforming growth factor
- YAP
- YAP, yes-associated protein
- cardiac fibrosis
- heart failure
- mRNA, messenger ribonucleic acid
- myocardial infarction
Collapse
Affiliation(s)
- Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Yu Zhang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Jae Im Jeong
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Andreas Ivessa
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Michelle D. Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
103
|
Wu T, Wang S, Wang L, Zhang W, Chen W, Lv X, Li Y, Hussain Z, Sun W. Long Noncoding RNA (lncRNA) CTTN-IT1 Elevates Skeletal Muscle Satellite Cell Proliferation and Differentiation by Acting as ceRNA for YAP1 Through Absorbing miR-29a in Hu Sheep. Front Genet 2020; 11:843. [PMID: 32849826 PMCID: PMC7427492 DOI: 10.3389/fgene.2020.00843] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022] Open
Abstract
Characterizing the factors that regulate the growth and development of muscle is central to animal production. Skeletal muscle satellite cells (SMSCs) provide an important material for simulating the proliferation and differentiation of muscle cells. YAP1, which can promote muscle growth, is closely related to the proliferation of SMSCs in Hu sheep (Ovis aries). In addition, some miRNAs, such as miR-541-3p, miR-142-5p, and miR-29a, can play critical roles in muscle growth by specifically binding with their target mRNAs. Meanwhile, lncRNA can competitively bind these miRNAs and reduce the regulatory effect of miRNAs on their target genes and thus play critical roles themselves in muscle growth. However, the regulatory molecular mechanism of miRNA and lncRNA on SMSC proliferation through YAP1 remains unclear. Here, we characterized the regulatory network among YAP1 and its targeted miRNAs and lncRNAs in Hu sheep SMSCs. The potential ncRNAs that regulate YAP1 (miR-29a and CTTN-IT1) were predicted through multilevel bioinformatics analysis. Dual-luciferase assays, RT-qPCR, and western blots revealed that miR-29a can significantly reduce the mRNA and protein expression level by binding to a specific 3′-UTR of YAP1 (P < 0.05), while CTTN-IT1 can restore the expression of YAP1 through competitive binding to miR-29a. Furthermore, the mRNA and protein expression levels of MyoG, MyoD, and MyHC showed that miR-29a can inhibit the expression of genes related to the differentiation of SMSCs, and CTTN-IT1 can increase the expression of these same genes. Thus, miR-29a may inhibit the differentiation of SMSCs and CTTN-IT1 can restore this inhibition. The EdU staining assay indicated that excessive miR-29a can significantly reduce the proliferation ability of SMSCs (P < 0.05), while overexpression of CTTN-IT1 can significantly increase the proliferation of SMSCs (P < 0.01). CTTN-IT1 is a novel lncRNA that is a competing endogenous RNA (ceRNA) of miR-29a and can promote SMSC proliferation and differentiation by restoring the expression of YAP1 when it is inhibited by miR-29a in Hu sheep. Overall, our findings construct a CTTN-IT1-miR-29a-YAP1 regulatory network that will help contribute new insight into improving the muscle development of Hu sheep.
Collapse
Affiliation(s)
- Tianyi Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shanhe Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Lihong Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Weibo Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Weihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiaoyang Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yue Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zahid Hussain
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
104
|
Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, Fussenegger M, Deng X. Role of YAP/TAZ in Cell Lineage Fate Determination and Related Signaling Pathways. Front Cell Dev Biol 2020; 8:735. [PMID: 32850847 PMCID: PMC7406690 DOI: 10.3389/fcell.2020.00735] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
The penultimate effectors of the Hippo signaling pathways YAP and TAZ, are transcriptional co-activator proteins that play key roles in many diverse biological processes, ranging from cell proliferation, tumorigenesis, mechanosensing and cell lineage fate determination, to wound healing and regeneration. In this review, we discuss the regulatory mechanisms by which YAP/TAZ control stem/progenitor cell differentiation into the various major lineages that are of interest to tissue engineering and regenerative medicine applications. Of particular interest is the key role of YAP/TAZ in maintaining the delicate balance between quiescence, self-renewal, proliferation and differentiation of endogenous adult stem cells within various tissues/organs during early development, normal homeostasis and regeneration/healing. Finally, we will consider how increasing knowledge of YAP/TAZ signaling might influence the trajectory of future progress in regenerative medicine.
Collapse
Affiliation(s)
- Boon C. Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Faculty of Science and Technology, Sunway University, Subang Jaya, Malaysia
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
| | - Dominique Aubel
- IUTA Department Genie Biologique, Universite Claude Bernard Lyon 1, Villeurbanne, France
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH-Zürich, Basel, Switzerland
| | - Xuliang Deng
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
105
|
Abstract
The heart requires a high amount of energy, in the form of adenosine triphosphate, to maintain its viability and pump function. Anaerobic glycolysis and mitochondrial oxidative phosphorylation are the two main metabolic pathways by which adenosine triphosphate is generated, using fatty acids, glucose, lactate, and ketone bodies as primary substrates. Previous studies have demonstrated that, in response to stress, the heart undergoes alterations in metabolism, ranging from changes in substrate utilization to mitochondrial function, collectively called metabolic remodeling. However, the molecular mechanism mediating metabolic remodeling in the heart remains unclear. Yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which are major downstream effectors of the Hippo signaling pathway, play an important role in the regulation of heart size and cellular homeostasis of cardiomyocytes through the regulation of various transcriptional factors under both physiological and pathophysiological conditions. Recent findings in various organs and cell types have revealed that YAP and TAZ play an important role in energy metabolism. Here, we summarize what is currently known about YAP/TAZ in the regulation of metabolism of various substrates and mitochondrial function in various organs and cell types and discuss the potential role of YAP/TAZ in mediating metabolic remodeling of the heart during stress and heart failure.
Collapse
|
106
|
Common Regulatory Pathways Mediate Activity of MicroRNAs Inducing Cardiomyocyte Proliferation. Cell Rep 2020; 27:2759-2771.e5. [PMID: 31141697 PMCID: PMC6547019 DOI: 10.1016/j.celrep.2019.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/03/2018] [Accepted: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
Loss of functional cardiomyocytes is a major determinant of heart failure after myocardial infarction. Previous high throughput screening studies have identified a few microRNAs (miRNAs) that can induce cardiomyocyte proliferation and stimulate cardiac regeneration in mice. Here, we show that all of the most effective of these miRNAs activate nuclear localization of the master transcriptional cofactor Yes-associated protein (YAP) and induce expression of YAP-responsive genes. In particular, miR-199a-3p directly targets two mRNAs coding for proteins impinging on the Hippo pathway, the upstream YAP inhibitory kinase TAOK1, and the E3 ubiquitin ligase β-TrCP, which leads to YAP degradation. Several of the pro-proliferative miRNAs (including miR-199a-3p) also inhibit filamentous actin depolymerization by targeting Cofilin2, a process that by itself activates YAP nuclear translocation. Thus, activation of YAP and modulation of the actin cytoskeleton are major components of the pro-proliferative action of miR-199a-3p and other miRNAs that induce cardiomyocyte proliferation. A few microRNAs can stimulate cardiac myocyte proliferation The most effective of these microRNAs activate YAP Several pro-proliferative microRNAs also inhibit actin depolymerization miR-199a-3p directly targets TAOK1, b-TrCP, and Cofilin2 to achieve its effects
Collapse
|
107
|
Zheng M, Jacob J, Hung SH, Wang J. The Hippo Pathway in Cardiac Regeneration and Homeostasis: New Perspectives for Cell-Free Therapy in the Injured Heart. Biomolecules 2020; 10:biom10071024. [PMID: 32664346 PMCID: PMC7407108 DOI: 10.3390/biom10071024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Intractable cardiovascular diseases are leading causes of mortality around the world. Adult mammalian hearts have poor regenerative capacity and are not capable of self-repair after injury. Recent studies of cell-free therapeutics such as those designed to stimulate endogenous cardiac regeneration have uncovered new feasible therapeutic avenues for cardiac repair. The Hippo pathway, a fundamental pathway with pivotal roles in cell proliferation, survival and differentiation, has tremendous potential for therapeutic manipulation in cardiac regeneration. In this review, we summarize the most recent studies that have revealed the function of the Hippo pathway in heart regeneration and homeostasis. In particular, we discuss the molecular mechanisms of how the Hippo pathway maintains cardiac homeostasis by directing cardiomyocyte chromatin remodeling and regulating the cell-cell communication between cardiomyocytes and non-cardiomyocytes in the heart.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
| | - Joan Jacob
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Shao-Hsi Hung
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
- Correspondence: ; Tel.: +1-7135-005-723
| |
Collapse
|
108
|
Riddell A, McBride M, Braun T, Nicklin SA, Cameron E, Loughrey CM, Martin TP. RUNX1: an emerging therapeutic target for cardiovascular disease. Cardiovasc Res 2020; 116:1410-1423. [PMID: 32154891 PMCID: PMC7314639 DOI: 10.1093/cvr/cvaa034] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/18/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Runt-related transcription factor-1 (RUNX1), also known as acute myeloid leukaemia 1 protein (AML1), is a member of the core-binding factor family of transcription factors which modulate cell proliferation, differentiation, and survival in multiple systems. It is a master-regulator transcription factor, which has been implicated in diverse signalling pathways and cellular mechanisms during normal development and disease. RUNX1 is best characterized for its indispensable role for definitive haematopoiesis and its involvement in haematological malignancies. However, more recently RUNX1 has been identified as a key regulator of adverse cardiac remodelling following myocardial infarction. This review discusses the role RUNX1 plays in the heart and highlights its therapeutic potential as a target to limit the progression of adverse cardiac remodelling and heart failure.
Collapse
Affiliation(s)
- Alexandra Riddell
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Martin McBride
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Stuart A Nicklin
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Ewan Cameron
- School of Veterinary Medicine, University of Glasgow, Garscube Campus, Glasgow G61 1BD, UK
| | - Christopher M Loughrey
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Tamara P Martin
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
109
|
Gründl M, Walz S, Hauf L, Schwab M, Werner KM, Spahr S, Schulte C, Maric HM, Ade CP, Gaubatz S. Interaction of YAP with the Myb-MuvB (MMB) complex defines a transcriptional program to promote the proliferation of cardiomyocytes. PLoS Genet 2020; 16:e1008818. [PMID: 32469866 PMCID: PMC7286521 DOI: 10.1371/journal.pgen.1008818] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/10/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023] Open
Abstract
The Hippo signalling pathway and its central effector YAP regulate proliferation of cardiomyocytes and growth of the heart. Using genetic models in mice we show that the increased proliferation of embryonal and postnatal cardiomyocytes due to loss of the Hippo-signaling component SAV1 depends on the Myb-MuvB (MMB) complex. Similarly, proliferation of postnatal cardiomyocytes induced by constitutive active YAP requires MMB. Genome studies revealed that YAP and MMB regulate an overlapping set of cell cycle genes in cardiomyocytes. Protein-protein interaction studies in cell lines and with recombinant proteins showed that YAP binds directly to B-MYB, a subunit of MMB, in a manner dependent on the YAP WW domains and a PPXY motif in B-MYB. Disruption of the interaction by overexpression of the YAP binding domain of B-MYB strongly inhibits the proliferation of cardiomyocytes. Our results point to MMB as a critical downstream effector of YAP in the control of cardiomyocyte proliferation. YAP, the major downstream transducer of the Hippo pathway, is a potent inducer of proliferation. Here we show that the Myb-MuvB complex (MMB) mediates cardiomyocyte proliferation by YAP. We find that YAP and MMB regulate an overlapping set of pro-proliferative genes which involves binding of MMB to the promoters of these genes. We also identified a direct interaction between the B-MYB subunit of MMB and YAP. Based on the binding studies, we created a tool called MY-COMP that interferes with the association YAP to B-MYB and strongly inhibits proliferation of cardiomyocytes. Together, our data suggests that the YAP-MMB interaction is essential for division of cardiomyocytes, underscoring the functional relevance of the crosstalk between these two pathways for proper heart development.
Collapse
Affiliation(s)
- Marco Gründl
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Susanne Walz
- Comprehensive Cancer Center Mainfranken, Core Unit Bioinformatics, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Laura Hauf
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Melissa Schwab
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Kerstin Marcela Werner
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Susanne Spahr
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Clemens Schulte
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Wuerzburg, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Wuerzburg, Germany
| | - Carsten P. Ade
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
| | - Stefan Gaubatz
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
110
|
|
111
|
Chen X, Li Y, Luo J, Hou N. Molecular Mechanism of Hippo-YAP1/TAZ Pathway in Heart Development, Disease, and Regeneration. Front Physiol 2020; 11:389. [PMID: 32390875 PMCID: PMC7191303 DOI: 10.3389/fphys.2020.00389] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/01/2020] [Indexed: 01/20/2023] Open
Abstract
The Hippo-YAP1/TAZ pathway is a highly conserved central mechanism that controls organ size through the regulation of cell proliferation and other physical attributes of cells. The transcriptional factors Yes-associated protein 1 (YAP1) and PDZ-binding motif (TAZ) act as downstream effectors of the Hippo pathway, and their subcellular location and transcriptional activities are affected by multiple post-translational modifications (PTMs). Studies have conclusively demonstrated a pivotal role of the Hippo-YAP1/TAZ pathway in cardiac development, disease, and regeneration. Targeted therapeutics for the YAP1/TAZ could be an effective treatment option for cardiac regeneration and disease. This review article provides an overview of the Hippo-YAP1/TAZ pathway and the increasing impact of PTMs in fine-tuning YAP1/TAZ activation; in addition, we discuss the potential contributions of the Hippo-YAP1/TAZ pathway in cardiac development, disease, and regeneration.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yilang Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
112
|
Ye L, Qiu L, Feng B, Jiang C, Huang Y, Zhang H, Zhang H, Hong H, Liu J. Role of Blood Oxygen Saturation During Post-Natal Human Cardiomyocyte Cell Cycle Activities. JACC Basic Transl Sci 2020; 5:447-460. [PMID: 32478207 PMCID: PMC7251192 DOI: 10.1016/j.jacbts.2020.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
Blood oxygen saturation (SaO2) is one of the most important environmental factors in clinical heart protection. This study used human heart samples and human induced pluripotent stem cell-cardiomyocytes (iPSC-CMs) to assess how SaO2 affects human CM cell cycle activities. The results showed that there were significantly more cell cycle markers in the moderate hypoxia group (SaO2: 75% to 85%) than in the other 2 groups (SaO2 <75% or >85%). In iPSC-CMs 15% and 10% oxygen (O2) treatment increased cell cycle markers, whereas 5% and rapid change of O2 decreased the markers. Moderate hypoxia is beneficial to the cell cycle activities of post-natal human CMs.
Collapse
Key Words
- CHD, congenital heart disease
- CM, cardiomyocytes
- IF, immunofluorescence
- LV, lentivirus
- O2, oxygen
- SaO2, blood oxygen saturation
- TOF, tetralogy of Fallot
- YAP1, yes-associated protein 1
- blood oxygen saturation
- cardiomyocyte
- congenital heart disease
- iPSC, induced pluripotent stem cell
- pATM, phosphorylated ataxia telangiectasia mutated
- pHH3, phospho-histone H3
- pediatric patients
- proliferation
- qPCR, quantitative polymerase chain reaction
- sh, short hairpin
Collapse
Affiliation(s)
- Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lisheng Qiu
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Feng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chuan Jiang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanhui Huang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haibo Zhang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hao Zhang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haifa Hong
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinfen Liu
- Shanghai Institute for Pediatric Congenital Heart Diseases, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
113
|
Ma C, Peng P, Zhou Y, Liu T, Wang L, Lu C. MicroRNA‑93 promotes angiogenesis and attenuates remodeling via inactivation of the Hippo/Yap pathway by targeting Lats2 after myocardial infarctionω. Mol Med Rep 2020; 22:483-493. [PMID: 32319642 PMCID: PMC7248469 DOI: 10.3892/mmr.2020.11085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 04/01/2020] [Indexed: 01/10/2023] Open
Abstract
Inactivation of the Hippo pathway protects the myocardium from cardiac ischemic injury. MicroRNAs (miRs) have been reported to play pivotal roles in the progression of myocardial infarction (MI). The present study examined whether miR-93 could promote angiogenesis and attenuate remodeling after MI via inactivation of the Hippo/Yes-associated protein (Yap) pathway, by targeting large tumor suppressor kinase 2 (Lats2). It was identified that transfection of human umbilical vein endothelial cells with miR-93 mimic significantly decreased Lats2 expression and Yap phosphorylation, increased cell viability and migration, and attenuated cell apoptosis following hypoxia/reoxygenation injury. Moreover, increased expression of miR-93 resulted in an improvement of cardiac function, promotion of angiogenesis and attenuation of remodeling after MI. Additionally, miR-93 overexpression significantly decreased intracellular adhesion molecule 1 and vascular cell adhesion protein 1 expression levels, as well as attenuated the infiltration of neutrophils and macrophages into the myocardium after MI. Furthermore, it was found that miR-93 overexpression significantly suppressed Lats2 expression and decreased the levels of phosphorylated Yap in the myocardium after MI. Collectively, the present results suggested that miR-93 may exert a protective effect against MI via inactivation of the Hippo/Yap pathway by targeting Lats2.
Collapse
Affiliation(s)
- Chengjie Ma
- Clinical and Research Center of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Peipei Peng
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Tianya Liu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lijuan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
114
|
Xie Y, Wang Q, Gao N, Wu F, Lan F, Zhang F, Jin L, Huang Z, Ge J, Wang H, Wang Y. MircroRNA-10b Promotes Human Embryonic Stem Cell-Derived Cardiomyocyte Proliferation via Novel Target Gene LATS1. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:437-445. [PMID: 31902743 PMCID: PMC6948266 DOI: 10.1016/j.omtn.2019.11.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 01/20/2023]
Abstract
Adult mammalian cardiomyocytes (CMs) retain a limited proliferative ability, which is insufficient for the repair of CM loss in ischemic cardiac injury. Regulation of the Hippo signaling pathway to promote endogenous CM proliferation has emerged as a promising strategy for heart regeneration. Previous studies have shown that the microRNA cluster miR302-367 negatively regulates the Hippo pathway, promoting CM proliferation. In this study, we identified another microRNA, miR-10b, that regulates the Hippo pathway and promotes cell proliferation in human embryonic stem cell-derived CMs (hESC-CMs). We observed that miR-10b expression was enriched in the early stage of CMs, but its expression was reduced over time. Overexpression of miR-10b promoted CM proliferation, while knockdown of miR-10b suppressed CM proliferation. Moreover, miR-10b protected CMs against apoptosis. miR-10b functions, in part, by directly targeting LATS1, which is a major component of the Hippo pathway. Our study suggests that miR-10b has promising potential for heart regeneration.
Collapse
Affiliation(s)
- Yifang Xie
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Qiaozi Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Ning Gao
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Fujian Wu
- Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Capital Medical University, Beijing 100029, China
| | - Feng Lan
- Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Capital Medical University, Beijing 100029, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Zheyong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Hongyan Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering at School of Life Sciences, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai 200032, China; Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Yongming Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
115
|
Li HR, Wang C, Sun P, Liu DD, Du GQ, Tian JW. Melatonin attenuates doxorubicin-induced cardiotoxicity through preservation of YAP expression. J Cell Mol Med 2020; 24:3634-3646. [PMID: 32068341 PMCID: PMC7131936 DOI: 10.1111/jcmm.15057] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
There are increasing concerns related to the cardiotoxicity of doxorubicin in the clinical setting. Recently, melatonin has been shown to exert a cardioprotective effect in various cardiovascular diseases, including cardiotoxic conditions. In this study, we examined the possible protective effects of melatonin on doxorubicin‐induced cardiotoxicity and explored the underlying mechanisms related to this process. We found that in vitro doxorubicin treatment significantly decreased H9c2 cell viability and induced apoptosis as manifested by increased TUNEL‐positive cells, down‐regulation of anti‐apoptotic protein Bcl‐2, as well as up‐regulation of pro‐apoptotic protein Bax. This was associated with increased reactive oxygen species (ROS) levels and decreased mitochondrial membrane potentials (MMP). In vivo, five weeks of doxorubicin treatment significantly decreased cardiac function, as evaluated by echocardiography. TUNEL staining results confirmed the increased apoptosis caused by doxorubicin. On the other hand, combinational treatment of doxorubicin with melatonin decreased cardiomyocyte ROS and apoptosis levels, along with increasing MMP. Such doxorubicin‐melatonin co‐treatment alleviated in vivo doxorubicin‐induced cardiac injury. Western Blots, along with in vitro immunofluorescence and in vivo immunohistochemical staining confirmed that doxorubicin treatment significantly down‐regulated Yes‐associated protein (YAP) expression, while YAP levels were maintained under co‐treatment of doxorubicin and melatonin. YAP inhibition by siRNA abolished the protective effects of melatonin on doxorubicin‐treated cardiomyocytes, with reversed ROS level and apoptosis. Our findings suggested that melatonin treatment attenuated doxorubicin‐induced cardiotoxicity through preserving YAP levels, which in turn decreases oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Hai-Ru Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Dan-Dan Liu
- Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guo-Qing Du
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jia-Wei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
116
|
The HIPPO Transducer YAP and Its Targets CTGF and Cyr61 Drive a Paracrine Signalling in Cold Atmospheric Plasma-Mediated Wound Healing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4910280. [PMID: 32104533 PMCID: PMC7040405 DOI: 10.1155/2020/4910280] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Reactive species play a pivotal role in orchestrating wound healing responses. They act as secondary messengers and drive redox-signalling pathways that are involved in the homeostatic, inflammatory, proliferative, and remodelling phases of wound healing. The application of Cold Atmospheric Plasma (CAP) to the wound site produces a profusion of short- and long-lived reactive species that have been demonstrated to be effective in promoting wound healing; however, knowledge of the mechanisms underlying CAP-mediated wound healing remains scarce. To address this, an in vitro coculture model was used to study the effects of CAP on wound healing and on paracrine crosstalk between dermal keratinocytes and fibroblasts. Using this coculture model, we observed a stimulatory effect on the migration ability of HaCaT cells that were cocultured with dermal fibroblasts. Additionally, CAP treatment resulted in an upregulation of the HIPPO transcription factor YAP in HaCaTs and fibroblasts. Downstream effectors of the HIPPO signalling pathway (CTGF and Cyr61) were also upregulated in dermal fibroblasts, and the administration of antioxidants could inhibit CAP-mediated wound healing and abrogate the gene expression of the HIPPO downstream effectors. Interestingly, we observed that HaCaT cells exhibited an improved cell migration rate when incubated with CAP-treated fibroblast-conditioned media compared to that observed after incubation with untreated media. An induction of CTGF and Cyr61 secretion was also observed upon CAP treatment in the fibroblast-conditioned media. Finally, exposure to recombinant CTGF and Cyr61 could also significantly improve HaCaT cell migration. In summary, our results validated that CAP activates a regenerative signalling pathway at the onset of wound healing. Additionally, CAP also stimulated a reciprocal communication between dermal fibroblasts and keratinocytes, resulting in improved keratinocyte wound healing in coculture.
Collapse
|
117
|
Yatsenko AS, Kucherenko MM, Xie Y, Aweida D, Urlaub H, Scheibe RJ, Cohen S, Shcherbata HR. Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 2020; 18:8. [PMID: 31959160 PMCID: PMC6971923 DOI: 10.1186/s12916-019-1478-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Physiology, Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Yuanbin Xie
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: University Medical Center, Centre for Anatomy, Institute of Neuroanatomy, Georg-August-University Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Dina Aweida
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Research Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Bioanalytics Institute for Clinical Chemistry, University Medical Center Goettingen, Robert Koch Strasse 40, 37075, Göttingen, Germany
| | - Renate J Scheibe
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | | | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany. .,Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
118
|
Chen J, Ma Q, King JS, Sun Y, Xu B, Zhang X, Zohrabian S, Guo H, Cai W, Li G, Bruno I, Cooke JP, Wang C, Kontaridis M, Wang DZ, Luo H, Pu WT, Lin Z. aYAP modRNA reduces cardiac inflammation and hypertrophy in a murine ischemia-reperfusion model. Life Sci Alliance 2020; 3:e201900424. [PMID: 31843959 PMCID: PMC6918510 DOI: 10.26508/lsa.201900424] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
Myocardial recovery from ischemia-reperfusion (IR) is shaped by the interaction of many signaling pathways and tissue repair processes, including the innate immune response. We and others previously showed that sustained expression of the transcriptional co-activator yes-associated protein (YAP) improves survival and myocardial outcome after myocardial infarction. Here, we asked whether transient YAP expression would improve myocardial outcome after IR injury. After IR, we transiently activated YAP in the myocardium with modified mRNA encoding a constitutively active form of YAP (aYAP modRNA). Histological studies 2 d after IR showed that aYAP modRNA reduced cardiomyocyte (CM) necrosis and neutrophil infiltration. 4 wk after IR, aYAP modRNA-treated mice had better heart function as well as reduced scar size and hypertrophic remodeling. In cultured neonatal and adult CMs, YAP attenuated H2O2- or LPS-induced CM necrosis. TLR signaling pathway components important for innate immune responses were suppressed by YAP/TEAD1. In summary, our findings demonstrate that aYAP modRNA treatment reduces CM necrosis, cardiac inflammation, and hypertrophic remodeling after IR stress.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/administration & dosage
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Cardiomegaly/drug therapy
- Cardiomegaly/etiology
- Cell Survival/drug effects
- Cells, Cultured
- Disease Models, Animal
- Humans
- Injections, Intramuscular
- Mice
- Mice, Inbred C57BL
- Myocardial Reperfusion Injury/complications
- Myocarditis/drug therapy
- Myocarditis/etiology
- Myocardium/immunology
- Myocytes, Cardiac/metabolism
- Neutrophil Infiltration/drug effects
- RNA Editing
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- Transcription Factors/administration & dosage
- Transcription Factors/genetics
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Jinmiao Chen
- Boston Children's Hospital, Boston, MA, USA
- Department of Cardiovascular Surgery and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Ma
- Boston Children's Hospital, Boston, MA, USA
| | | | - Yan Sun
- Masonic Medical Research Institute, Utica, NY, USA
| | - Bing Xu
- Masonic Medical Research Institute, Utica, NY, USA
| | | | | | - Haipeng Guo
- Boston Children's Hospital, Boston, MA, USA
- Department of Critical Care Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China
| | - Wenqing Cai
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Gavin Li
- Boston Children's Hospital, Boston, MA, USA
| | - Ivone Bruno
- Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Houston Methodist Research Institute, Houston, TX, USA
| | - Chunsheng Wang
- Department of Cardiovascular Surgery and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | - Hongbo Luo
- Boston Children's Hospital, Boston, MA, USA
| | - William T Pu
- Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Zhiqiang Lin
- Boston Children's Hospital, Boston, MA, USA
- Masonic Medical Research Institute, Utica, NY, USA
| |
Collapse
|
119
|
FibroAtlas: A Database for the Exploration of Fibrotic Diseases and Their Genes. Cardiol Res Pract 2019; 2019:4237285. [PMID: 32082621 PMCID: PMC7012261 DOI: 10.1155/2019/4237285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022] Open
Abstract
Background Fibrosis is a highly dynamic process caused by prolonged injury, deregulation of the normal processes of wound healing, and extensive deposition of extracellular matrix (ECM) proteins. During fibrosis process, multiple genes interact with environmental factors. Over recent decades, tons of fibrosis-related genes have been identified to shed light on the particular clinical manifestations of this complex process. However, the genetics information about fibrosis is dispersed in lots of extensive literature. Methods We extracted data from literature abstracts in PubMed by text mining, and manually curated the literature and identified the evidence sentences. Results We presented FibroAtlas, which included 1,439 well-annotated fibrosis-associated genes. FibroAtlas 1.0 is the first attempt to build a nonredundant and comprehensive catalog of fibrosis-related genes with supporting evidence derived from curated published literature and allows us to have an overview of human fibrosis-related genes.
Collapse
|
120
|
Avalos-de León CG, Jiménez-Castro MB, Cornide-Petronio ME, Gulfo J, Rotondo F, Gracia-Sancho J, Casillas-Ramírez A, Peralta C. The Effect of Fibroblast Growth Factor 15 Signaling in Non-Steatotic and Steatotic Liver Transplantation from Cardiocirculatory Death. Cells 2019; 8:1640. [PMID: 31847428 PMCID: PMC6952771 DOI: 10.3390/cells8121640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022] Open
Abstract
We elucidate the relevance of fibroblast growth factor 15 (FGF15) in liver transplantation (LT) using rats with both steatotic and non-steatotic organs from donors after cardiocirculatory death (DCD). Compared to LT from non-DCDs, the induction of cardiocirculatory death (CD) increases hepatic damage, proliferation, and intestinal and circulatory FGF15. This is associated with high levels of FGF15, bilirubin and bile acids (BAs), and overexpression of the enzyme involved in the alternative BA synthesis pathway, CYP27A1, in non-steatotic livers. Furthermore, CD activates the proliferative pathway, Hippo/YAP, in these types of liver. Blocking FGF15 action in LT from DCDs does not affect CYP27A1 but causes an overexpression of CYP7A, an enzyme from the classic BA synthesis pathway, and this is related to further accumulation of BAs and exacerbated damage. FGF15 inhibition also impairs proliferation without changing Hippo/YAP. In spite of worse damage, steatosis prevents a proliferative response in livers from DCDs. In steatotic grafts, CD does not modify CYP7A1, CYP27A1, BA, or the Hippo/YAP pathway, and FGF15 is not involved in damage or proliferation. Thus, endogenous FGF15 protects against BA accumulation and damage and promotes regeneration independently of the Hippo/YAP pathway, in non-steatotic LT from DCDs. Herein we show a minor role of FGF15 in steatotic LT from DCDs.
Collapse
Affiliation(s)
- Cindy G. Avalos-de León
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
| | - Mónica B. Jiménez-Castro
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
| | - María Eugenia Cornide-Petronio
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
| | - José Gulfo
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
| | - Floriana Rotondo
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
| | - Jordi Gracia-Sancho
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain;
- Liver Vascular Biology Research Group, IDIBAPS, 08036 Barcelona, Spain
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros 87300, Mexico
| | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain; (C.G.A.-d.L.); (M.B.J.-C.); (M.E.C.-P.); (J.G.); (F.R.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain;
| |
Collapse
|
121
|
Takaguri A, Akihiro O, Sasano J, Satoh K. Involvement of Yes-associated protein 1 (YAP1) in doxorubicin-induced cytotoxicity in H9c2 cardiac cells. Cell Biol Int 2019; 44:873-881. [PMID: 31833156 DOI: 10.1002/cbin.11285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/10/2019] [Indexed: 11/06/2022]
Abstract
Cardiac cell death is one of the major events implicated in doxorubicin-induced cardiotoxicity, which leads to heart failure. We recently reported that Yes-associated protein 1 (YAP1) regulates cell survival and apoptosis. However, it is unclear whether YAP1 regulates doxorubicin-induced cell death in cardiomyocytes. We investigated whether YAP1 is involved in doxorubicin-induced cell death using H9c2 cardiac cells and mouse heart. In an in vivo study, YAP1 protein expression was significantly decreased in hearts of doxorubicin-treated mice with increased caspase-3 activation. Doxorubicin also caused cell death by increasing caspase-3 activation in H9c2 cells. Doxorubicin reduced YAP1 protein expression and messenger RNA expression accompanied by increased phosphorylation of YAP1 at Ser127. Doxorubicin further increased cell death with increased caspase-3/7 activation in the absence of YAP1 when compared with doxorubicin or siYAP1 treatment alone. Overexpression of constitutively active YAP1 (YAP1-5SA) using an adenovirus gene transfer technique significantly reversed doxorubicin-induced cell death by decreasing caspase-3/7 activation in H9c2 cells. Akt, a potential prosurvival factor, decreased in doxorubicin- and YAP1 short interfering RNA (siRNA)-treated cells. Doxorubicin further significantly decreased Akt protein expression when YAP1 was silenced. Overexpression of YAP1 canceled decreased Akt protein expression induced by doxorubicin treatment in H9c2 cells. In conclusion, these results suggest that doxorubicin-induced cardiac cell death is mediated in part by down-regulation of YAP1 and YAP1-targeted gene, Akt. Modulating YAP1 and its related Hippo pathway on local cardiomyocytes may be a promising therapeutic approach for doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Akira Takaguri
- Department of Pharmacology, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Ohmiya Akihiro
- Department of Pharmacology, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Jun Sasano
- Department of Pharmacology, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| | - Kumi Satoh
- Department of Pharmacology, Hokkaido University of Science, 7-15-4-1 Maeda, Teine-ku, Sapporo, 006-8590, Japan
| |
Collapse
|
122
|
Judd J, Lovas J, Huang GN. Defined factors to reactivate cell cycle activity in adult mouse cardiomyocytes. Sci Rep 2019; 9:18830. [PMID: 31827131 PMCID: PMC6906479 DOI: 10.1038/s41598-019-55027-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
Adult mammalian cardiomyocytes exit the cell cycle during the neonatal period, commensurate with the loss of regenerative capacity in adult mammalian hearts. We established conditions for long-term culture of adult mouse cardiomyocytes that are genetically labeled with fluorescence. This technique permits reliable analyses of proliferation of pre-existing cardiomyocytes without complications from cardiomyocyte marker expression loss due to dedifferentiation or significant contribution from cardiac progenitor cell expansion and differentiation in culture. Using this system, we took a candidate gene approach to screen for fetal-specific proliferative gene programs that can induce proliferation of adult mouse cardiomyocytes. Using pooled gene delivery and subtractive gene elimination, we identified a novel functional interaction between E2f Transcription Factor 2 (E2f2) and Brain Expressed X-Linked (Bex)/Transcription elongation factor A-like (Tceal) superfamily members Bex1 and Tceal8. Specifically, Bex1 and Tceal8 both preserved cell viability during E2f2-induced cell cycle re-entry. Although Tceal8 inhibited E2f2-induced S-phase re-entry, Bex1 facilitated DNA synthesis while inhibiting cell death. In sum, our study provides a valuable method for adult cardiomyocyte proliferation research and suggests that Bex family proteins may function in modulating cell proliferation and death decisions during cardiomyocyte development and maturation.
Collapse
Affiliation(s)
- Justin Judd
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jonathan Lovas
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Guo N Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, 94158, USA. .,Department of Physiology, University of California, San Francisco, San Francisco, CA, 94158, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
123
|
Li W, Wang L, Wu Y, Yuan Z, Zhou J. Weighted gene co‑expression network analysis to identify key modules and hub genes associated with atrial fibrillation. Int J Mol Med 2019; 45:401-416. [PMID: 31894294 PMCID: PMC6984797 DOI: 10.3892/ijmm.2019.4416] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Atrial fibrillation (AF) is the most common form of cardiac arrhythmia and significantly increases the risks of morbidity, mortality and health care expenditure; however, treatment for AF remains unsatisfactory due to the complicated and incompletely understood underlying mechanisms. In the present study, weighted gene co‑expression network analysis (WGCNA) was conducted to identify key modules and hub genes to determine their potential associations with AF. WGCNA was performed in an AF dataset GSE79768 obtained from the Gene Expression Omnibus, which contained data from paired left and right atria in cardiac patients with persistent AF or sinus rhythm. Differentially expressed gene (DEG) analysis was used to supplement and validate the results of WGCNA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were also performed. Green and magenta modules were identified as the most critical modules associated with AF, from which 6 hub genes, acetyl‑CoA Acetyltransferase 1, death domain‑containing protein CRADD, gypsy retrotransposon integrase 1, FTX transcript, XIST regulator, transcription elongation factor A like 2 and minichromosome maintenance complex component 3 associated protein, were hypothesized to serve key roles in the pathophysiology of AF due to their increased intramodular connectivity. Functional enrichment analysis results demonstrated that the green module was associated with energy metabolism, and the magenta module may be associated with the Hippo pathway and contain multiple interactive pathways associated with apoptosis and inflammation. In addition, the blue module was identified to be an important regulatory module in AF with a higher specificity for the left atria, the genes of which were primarily correlated with complement, coagulation and extracellular matrix formation. These results suggest that may improve understanding of the underlying mechanisms of AF, and assist in identifying biomarkers and potential therapeutic targets for treating patients with AF.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lijun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Juan Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
124
|
Ikeda S, Mizushima W, Sciarretta S, Abdellatif M, Zhai P, Mukai R, Fefelova N, Oka SI, Nakamura M, Del Re DP, Farrance I, Park JY, Tian B, Xie LH, Kumar M, Hsu CP, Sadayappan S, Shimokawa H, Lim DS, Sadoshima J. Hippo Deficiency Leads to Cardiac Dysfunction Accompanied by Cardiomyocyte Dedifferentiation During Pressure Overload. Circ Res 2019; 124:292-305. [PMID: 30582455 DOI: 10.1161/circresaha.118.314048] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE The Hippo pathway plays an important role in determining organ size through regulation of cell proliferation and apoptosis. Hippo inactivation and consequent activation of YAP (Yes-associated protein), a transcription cofactor, have been proposed as a strategy to promote myocardial regeneration after myocardial infarction. However, the long-term effects of Hippo deficiency on cardiac function under stress remain unknown. OBJECTIVE We investigated the long-term effect of Hippo deficiency on cardiac function in the presence of pressure overload (PO). METHODS AND RESULTS We used mice with cardiac-specific homozygous knockout of WW45 (WW45cKO), in which activation of Mst1 (Mammalian sterile 20-like 1) and Lats2 (large tumor suppressor kinase 2), the upstream kinases of the Hippo pathway, is effectively suppressed because of the absence of the scaffolding protein. We used male mice at 3 to 4 month of age in all animal experiments. We subjected WW45cKO mice to transverse aortic constriction for up to 12 weeks. WW45cKO mice exhibited higher levels of nuclear YAP in cardiomyocytes during PO. Unexpectedly, the progression of cardiac dysfunction induced by PO was exacerbated in WW45cKO mice, despite decreased apoptosis and activated cardiomyocyte cell cycle reentry. WW45cKO mice exhibited cardiomyocyte sarcomere disarray and upregulation of TEAD1 (transcriptional enhancer factor) target genes involved in cardiomyocyte dedifferentiation during PO. Genetic and pharmacological inactivation of the YAP-TEAD1 pathway reduced the PO-induced cardiac dysfunction in WW45cKO mice and attenuated cardiomyocyte dedifferentiation. Furthermore, the YAP-TEAD1 pathway upregulated OSM (oncostatin M) and OSM receptors, which played an essential role in mediating cardiomyocyte dedifferentiation. OSM also upregulated YAP and TEAD1 and promoted cardiomyocyte dedifferentiation, indicating the existence of a positive feedback mechanism consisting of YAP, TEAD1, and OSM. CONCLUSIONS Although activation of YAP promotes cardiomyocyte regeneration after cardiac injury, it induces cardiomyocyte dedifferentiation and heart failure in the long-term in the presence of PO through activation of the YAP-TEAD1-OSM positive feedback mechanism.
Collapse
Affiliation(s)
- Shohei Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Wataru Mizushima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S. Sciarretta).,Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S. Sciarretta)
| | - Maha Abdellatif
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Risa Mukai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Nadezhda Fefelova
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Shin-Ichi Oka
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Michinari Nakamura
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Dominic P Del Re
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | | | - Ji Yeon Park
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Lai-Hua Xie
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Dae-Sik Lim
- Department of Biological Science, National Creative Research Initiatives Center for Cell Division and Differentiation, Korea Advanced Institute of Science and Technology, Daejeon (D.-S.L.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| |
Collapse
|
125
|
Triastuti E, Nugroho AB, Zi M, Prehar S, Kohar YS, Bui TA, Stafford N, Cartwright EJ, Abraham S, Oceandy D. Pharmacological inhibition of Hippo pathway, with the novel kinase inhibitor XMU-MP-1, protects the heart against adverse effects during pressure overload. Br J Pharmacol 2019; 176:3956-3971. [PMID: 31328787 PMCID: PMC6811740 DOI: 10.1111/bph.14795] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/13/2019] [Accepted: 07/05/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The Hippo pathway has emerged as a potential therapeutic target to control pathological cardiac remodelling. The core components of the Hippo pathway, mammalian Ste-20 like kinase 1 (Mst1) and mammalian Ste-20 like kinase 2 (Mst2), modulate cardiac hypertrophy, apoptosis, and fibrosis. Here, we study the effects of pharmacological inhibition of Mst1/2 using a novel inhibitor XMU-MP-1 in controlling the adverse effects of pressure overload-induced hypertrophy. EXPERIMENTAL APPROACH We used cultured neonatal rat cardiomyocytes (NRCM) and C57Bl/6 mice with transverse aortic constriction (TAC) as in vitro and in vivo models, respectively, to test the effects of XMU-MP-1 treatment. We used luciferase reporter assays, western blots and immunofluorescence assays in vitro, with echocardiography, qRT-PCR and immunohistochemical methods in vivo. KEY RESULTS XMU-MP-1 treatment significantly increased activity of the Hippo pathway effector yes-associated protein and inhibited phenylephrine-induced hypertrophy in NRCM. XMU-MP-1 improved cardiomyocyte survival and reduced apoptosis following oxidative stress. In vivo, mice 3 weeks after TAC, were treated with XMU-MP-1 (1 mg·kg-1 ) every alternate day for 10 further days. XMU-MP-1-treated mice showed better cardiac contractility than vehicle-treated mice. Cardiomyocyte cross-sectional size and expression of the hypertrophic marker, brain natriuretic peptide, were reduced in XMU-MP-1-treated mice. Improved heart function in XMU-MP-1-treated mice with TAC, was accompanied by fewer TUNEL positive cardiomyocytes and lower levels of fibrosis, suggesting inhibition of cardiomyocyte apoptosis and decreased fibrosis. CONCLUSIONS AND IMPLICATIONS The Hippo pathway inhibitor, XMU-MP-1, reduced cellular hypertrophy and improved survival in cultured cardiomyocytes and, in vivo, preserved cardiac function following pressure overload.
Collapse
Affiliation(s)
- Efta Triastuti
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
- Department of Pharmacy, Faculty of MedicineUniversitas BrawijayaMalangIndonesia
| | - Ardiansah Bayu Nugroho
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Min Zi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Sukhpal Prehar
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Yulia Suciati Kohar
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
- Department of Biochemistry, Faculty of MedicineYARSI UniversityJakartaIndonesia
| | - Thuy Anh Bui
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Nicholas Stafford
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Elizabeth J. Cartwright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Sabu Abraham
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and HealthThe University of Manchester, Manchester Academic Health Science CentreManchesterUK
| |
Collapse
|
126
|
Mia MM, Singh MK. The Hippo Signaling Pathway in Cardiac Development and Diseases. Front Cell Dev Biol 2019; 7:211. [PMID: 31632964 PMCID: PMC6779857 DOI: 10.3389/fcell.2019.00211] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/12/2019] [Indexed: 01/07/2023] Open
Abstract
Heart disease continues to be the leading cause of morbidity and mortality worldwide. Cardiac malformation during development could lead to embryonic or postnatal death. However, matured heart tissue has a very limited regenerative capacity. Thus, loss of cardiomyocytes from injury or diseases in adults could lead to heart failure. The Hippo signaling pathway is a newly identified signaling cascade that modulates regenerative response by regulating cardiomyocyte proliferation in the embryonic heart, as well as in postnatal hearts after injury. In this review, we summarize recent findings highlighting the function and regulation of the Hippo signaling pathway in cardiac development and diseases.
Collapse
Affiliation(s)
- Masum M Mia
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Manvendra K Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore
| |
Collapse
|
127
|
Ikeda S, Mukai R, Mizushima W, Zhai P, Oka SI, Nakamura M, Del Re DP, Sciarretta S, Hsu CP, Shimokawa H, Sadoshima J. Yes-Associated Protein (YAP) Facilitates Pressure Overload-Induced Dysfunction in the Diabetic Heart. JACC Basic Transl Sci 2019; 4:611-622. [PMID: 31768477 PMCID: PMC6872826 DOI: 10.1016/j.jacbts.2019.05.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/19/2019] [Accepted: 05/09/2019] [Indexed: 01/09/2023]
Abstract
Patients with diabetes are more prone to developing heart failure in the presence of high blood pressure than those without diabetes. Yes-associated protein (YAP), a key effector of the Hippo signaling pathway, is persistently activated in diabetic hearts, and YAP plays an essential role in mediating the exacerbation of heart failure in response to pressure overload in the hearts of mice fed a high-fat diet. YAP induced dedifferentiation of cardiomyocytes through activation of transcriptional enhancer factor 1 (TEAD1), a transcription factor. Thus, YAP and TEAD1 are promising therapeutic targets for diabetic patients with high blood pressure to prevent the development of heart failure.
Collapse
Key Words
- HF, heart failure
- HFD, high-fat diet
- Hippo pathway
- LV, left ventricular
- Lats2, large tumor suppressor kinase 2
- Mst1, mammalian sterile 20-like 1
- ND, normal diet
- OSM, oncostatin M
- PO, pressure overload
- Runx1, runt-related transcription factor 1
- TAC, transverse aortic constriction
- TAZ, transcriptional coactivator with PDZ-binding motif
- TEAD, transcriptional enhancer factor
- YAP, Yes-associated protein
- diabetes
- diabetic cardiomyopathy
- pressure overload
Collapse
Affiliation(s)
- Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey.,Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Istituto Di Ricovero e Cura a Carattere Scientifico Neuromed, Pozzilli, Italy
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
128
|
Abstract
The Hippo-YAP (Yes-associated protein) pathway is an evolutionarily and functionally conserved regulator of organ size and growth with crucial roles in cell proliferation, apoptosis, and differentiation. This pathway has great potential for therapeutic manipulation in different disease states and to promote organ regeneration. In this Review, we summarize findings from the past decade revealing the function and regulation of the Hippo-YAP pathway in cardiac development, growth, homeostasis, disease, and regeneration. In particular, we highlight the roles of the Hippo-YAP pathway in endogenous heart muscle renewal, including the pivotal role of the Hippo-YAP pathway in regulating cardiomyocyte proliferation and differentiation, stress response, and mechanical signalling. The human heart lacks the capacity to self-repair; therefore, the loss of cardiomyocytes after injury such as myocardial infarction can result in heart failure and death. Despite substantial advances in the treatment of heart failure, an enormous unmet clinical need exists for alternative treatment options. Targeting the Hippo-YAP pathway has tremendous potential for developing therapeutic strategies for cardiac repair and regeneration for currently intractable cardiovascular diseases such as heart failure. The lessons learned from cardiac repair and regeneration studies will also bring new insights into the regeneration of other tissues with limited regenerative capacity.
Collapse
|
129
|
Lee EH, Park KI, Kim KY, Lee JH, Jang EJ, Ku SK, Kim SC, Suk HY, Park JY, Baek SY, Kim YW. Liquiritigenin inhibits hepatic fibrogenesis and TGF-β1/Smad with Hippo/YAP signal. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152780. [PMID: 31121384 DOI: 10.1016/j.phymed.2018.12.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 11/23/2018] [Accepted: 12/09/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Recent reports highlighted the possibility that Yes-associated protein (YAP) and transforming growth factor-β1 (TGF-β1) can act as critical regulators of hepatic stellate cells (HSCs) activation; therefore, it is natural for compounds targeting Hippo/YAP and TGF-β1/Smad signaling pathways to be identified as potential anti-fibrotic candidates. PURPOSE Liquiritigenin (LQ) is an aglycone of liquiritin and has been reported to protect the liver from injury. However, its effects on the Hippo/YAP and TGF-β1/Smad pathways have not been identified to date. METHODS We conducted a series of experiments using CCl4-induced fibrotic mice and cultured LX-2 cells. RESULT LQ significantly inhibited liver fibrosis, as indicated by decreases in regions of hepatic degeneration, inflammatory cell infiltration, and the intensity of α-smooth muscle actin (α-SMA) staining in mice. Moreover, LQ blocked the TGF-β1-induced phosphorylation of Smad 3, and the transcript levels of plasminogen activator inhibitor-1 (PAI-1) and matrix metalloproteinase-2 (MMP-2) in LX-2 cells, which is similar with resveratrol and oxyresveratrol (positive controls). Furthermore, LQ increased activation of large tumor suppressor kinase 1 (LATS1) with the induction of YAP phosphorylation, thereby preventing YAP transcriptional activity and suppressing the expression of exacerbated TGF-β1/Smad signaling molecules. CONCLUSION These results clearly show that LQ ameliorated experimental liver fibrosis by acting on the TGF-β1/Smad and Hippo/YAP pathways, indicating that LQ has the potential for effective treatment of liver fibrosis.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Pathology, School of Medical Science, Kyungpook National University, Daegu 41566, South Korea
| | - Kwang-Il Park
- Korea Institute of Oriental Medicine, Daegu 41062, South Korea
| | - Kwang-Youn Kim
- Korea Institute of Oriental Medicine, Daegu 41062, South Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Gyeongju 38066, South Korea
| | - Eun Jeong Jang
- College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Sae Kwang Ku
- College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Sang Chan Kim
- College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Ho Young Suk
- Yeungnam University, Gyeongsan 38541, South Korea
| | - Ji Young Park
- Department of Pathology, School of Medical Science, Kyungpook National University, Daegu 41566, South Korea
| | - Su Youn Baek
- College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, South Korea.
| | - Young Woo Kim
- College of Korean Medicine, Dongguk University, Gyeongju 38066, South Korea; College of Oriental Medicine, Daegu Haany University, Gyeongsan 38610, South Korea.
| |
Collapse
|
130
|
Kakiuchi-Kiyota S, Schutten MM, Zhong Y, Crawford JJ, Dey A. Safety Considerations in the Development of Hippo Pathway Inhibitors in Cancers. Front Cell Dev Biol 2019; 7:156. [PMID: 31475147 PMCID: PMC6707765 DOI: 10.3389/fcell.2019.00156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/25/2019] [Indexed: 01/04/2023] Open
Abstract
The Hippo pathway is a critical regulator of cell and organ growth and has emerged as a target for therapeutic intervention in cancers. Its signaling is thought to play an important role in various physiological processes including homeostasis and tissue regeneration. To date there has been limited information about potential pharmacology-related (on-target) safety liabilities of Hippo pathway inhibitors in the context of cancer indications. Herein, we review data from human genetic disorders and genetically engineered rodent models to gain insight into safety liabilities that may emerge from the inhibition of Hippo pathway. Germline systemic deletion of murine Hippo pathway effectors (Yap, Taz, and Teads) resulted in embryonic lethality or developmental phenotypes. Mouse models with tissue-specific deletion (or mutant overexpression) of the key effectors in Hippo pathways have indicated that, at least in some tissues, Hippo signaling may be dispensable for physiological homeostasis; and appears to be critical for regeneration upon tissue damage, indicating that patients with underlying comorbidities and/or insults caused by therapeutic agents and/or comedications may have a higher risk. Caution should be taken in interpreting phenotypes from tissue-specific transgenic animal models since some tissue-specific promoters are turned on during development. In addition, therapeutic agents may result in systemic effects not well-predicted by animal models with tissue-specific gene deletion. Therefore, the development of models that allows for systemic deletion of Yap and/or Taz in adult animals will be key in evaluating the potential safety liabilities of Hippo pathway modulation. In this review, we focus on potential challenges and strategies for targeting the Hippo pathway in cancers.
Collapse
Affiliation(s)
- Satoko Kakiuchi-Kiyota
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - Melissa M Schutten
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - Yu Zhong
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - James J Crawford
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, CA, United States
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
131
|
Francisco J, Byun J, Zhang Y, Kalloo OB, Mizushima W, Oka S, Zhai P, Sadoshima J, Del Re DP. The tumor suppressor RASSF1A modulates inflammation and injury in the reperfused murine myocardium. J Biol Chem 2019; 294:13131-13144. [PMID: 31311858 DOI: 10.1074/jbc.ra119.008970] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/11/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammation is a central feature of cardiovascular disease, including myocardial infarction and heart failure. Reperfusion of the ischemic myocardium triggers a complex inflammatory response that can exacerbate injury and worsen heart function, as well as prevent myocardial rupture and mediate wound healing. Therefore, a more complete understanding of this process could contribute to interventions that properly balance inflammatory responses for improved outcomes. In this study, we leveraged several approaches, including global and regional ischemia/reperfusion (I/R), genetically modified mice, and primary cell culture, to investigate the cell type-specific function of the tumor suppressor Ras association domain family member 1 isoform A (RASSF1A) in cardiac inflammation. Our results revealed that genetic inhibition of RASSF1A in cardiomyocytes affords cardioprotection, whereas myeloid-specific deletion of RASSF1A exacerbates inflammation and injury caused by I/R in mice. Cell-based studies revealed that RASSF1A negatively regulates NF-κB and thereby attenuates inflammatory cytokine expression. These findings indicate that myeloid RASSF1A antagonizes I/R-induced myocardial inflammation and suggest that RASSF1A may be a promising target in immunomodulatory therapy for the management of acute heart injury.
Collapse
Affiliation(s)
- Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Yu Zhang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Olivia Berman Kalloo
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103.
| |
Collapse
|
132
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
133
|
Camberos V, Baio J, Bailey L, Hasaniya N, Lopez LV, Kearns-Jonker M. Effects of Spaceflight and Simulated Microgravity on YAP1 Expression in Cardiovascular Progenitors: Implications for Cell-Based Repair. Int J Mol Sci 2019; 20:E2742. [PMID: 31167392 PMCID: PMC6600678 DOI: 10.3390/ijms20112742] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 01/06/2023] Open
Abstract
Spaceflight alters many processes of the human body including cardiac function and cardiac progenitor cell behavior. The mechanism behind these changes remains largely unknown; however, simulated microgravity devices are making it easier for researchers to study the effects of microgravity. To study the changes that take place in cardiac progenitor cells in microgravity environments, adult cardiac progenitor cells were cultured aboard the International Space Station (ISS) as well as on a clinostat and examined for changes in Hippo signaling, a pathway known to regulate cardiac development. Cells cultured under microgravity conditions, spaceflight-induced or simulated, displayed upregulation of downstream genes involved in the Hippo pathway such as YAP1 and SOD2. YAP1 is known to play a role in cardiac regeneration which led us to investigate YAP1 expression in a sheep model of cardiovascular repair. Additionally, to mimic the effects of microgravity, drug treatment was used to induce Hippo related genes as well as a regulator of the Hippo pathway, miRNA-302a. These studies provide insight into the changes that occur in space and how the effects of these changes relate to cardiac regeneration studies.
Collapse
Affiliation(s)
- Victor Camberos
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Jonathan Baio
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Leonard Bailey
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Nahidh Hasaniya
- Department of Cardiovascular and Thoracic Surgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Larry V Lopez
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
134
|
Zhao WB, Lu Q, Nguyen MN, Su Y, Ziemann M, Wang LN, Kiriazis H, Puthalakath H, Sadoshima J, Hu HY, Du XJ. Stimulation of β-adrenoceptors up-regulates cardiac expression of galectin-3 and BIM through the Hippo signalling pathway. Br J Pharmacol 2019; 176:2465-2481. [PMID: 30932177 PMCID: PMC6592853 DOI: 10.1111/bph.14674] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/29/2019] [Accepted: 03/04/2019] [Indexed: 01/01/2023] Open
Abstract
Background and Purpose Expression of the pro‐fibrotic galectin‐3 and the pro‐apoptotic BIM is elevated in diseased heart or after β‐adrenoceptor stimulation, but the underlying mechanisms are unclear. This question was addressed in the present study. Experimental Approach Wild‐type mice and mice with cardiac transgenic expression of β2‐adrenoceptors, mammalian sterile‐20 like kinase 1 (Mst1) or dominant‐negative Mst1, and non‐specific galectin‐3 knockout mice were used. Effects of the β‐adrenoceptor agonist isoprenaline or β‐adrenoceptor antagonists were studied. Rat cardiomyoblasts (H9c2) were used for mechanistic exploration. Biochemical assays were performed. Key Results Isoprenaline treatment up‐regulated expression of galectin‐3 and BIM, and this was inhibited by non‐selective or selective β‐adrenoceptor antagonists (by 60–70%). Cardiac expression of galectin‐3 and BIM was increased in β2‐adrenoceptor transgenic mice. Isoprenaline‐induced up‐regulation of galectin‐3 and BIM was attenuated by Mst1 inactivation, but isoprenaline‐induced galectin‐3 expression was exaggerated by transgenic Mst1 activation. Pharmacological or genetic activation of β‐adrenoceptors induced Mst1 expression and yes‐associated protein (YAP) phosphorylation. YAP hyper‐phosphorylation was also evident in Mst1 transgenic hearts with up‐regulated expression of galectin‐3 (40‐fold) and BIM as well as up‐regulation of many YAP‐target genes by RNA sequencing. In H9c2 cells, isoprenaline induced YAP phosphorylation and expression of galectin‐3 and BIM, effects simulated by forskolin but abolished by PKA inhibitors, and YAP knockdown induced expression of galectin‐3 and BIM. Conclusions and Implications Stimulation of cardiac β‐adrenoceptors activated the Mst1/Hippo pathway leading to YAP hyper‐phosphorylation with enhanced expression of galectin‐3 and BIM. This signalling pathway would have therapeutic potential. Linked Articles This article is part of a themed section on Adrenoceptors—New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc
Collapse
Affiliation(s)
- Wei-Bo Zhao
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Qun Lu
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Cardiovascular Medicine, First Hospital and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - My-Nhan Nguyen
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yidan Su
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University, Geelong, Australia
| | - Li-Na Wang
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Hou-Yuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Cardiovascular Medicine, First Hospital and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
135
|
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
|
136
|
Kim CL, Choi SH, Mo JS. Role of the Hippo Pathway in Fibrosis and Cancer. Cells 2019; 8:cells8050468. [PMID: 31100975 PMCID: PMC6562634 DOI: 10.3390/cells8050468] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway is the key player in various signaling processes, including organ development and maintenance of tissue homeostasis. This pathway comprises a core kinases module and transcriptional activation module, representing a highly conserved mechanism from Drosophila to vertebrates. The central MST1/2-LATS1/2 kinase cascade in this pathway negatively regulates YAP/TAZ transcription co-activators in a phosphorylation-dependent manner. Nuclear YAP/TAZ bind to transcription factors to stimulate gene expression, contributing to the regenerative potential and regulation of cell growth and death. Recent studies have also highlighted the potential role of Hippo pathway dysfunctions in the pathology of several diseases. Here, we review the functional characteristics of the Hippo pathway in organ fibrosis and tumorigenesis, and discuss its potential as new therapeutic targets.
Collapse
Affiliation(s)
- Cho-Long Kim
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Sue-Hee Choi
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jung-Soon Mo
- Genomic Instability Research Center (GIRC), Ajou University School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
137
|
Brand CS, Lighthouse JK, Trembley MA. Protective transcriptional mechanisms in cardiomyocytes and cardiac fibroblasts. J Mol Cell Cardiol 2019; 132:1-12. [PMID: 31042488 DOI: 10.1016/j.yjmcc.2019.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Heart failure is the leading cause of morbidity and mortality worldwide. Several lines of evidence suggest that physical activity and exercise can pre-condition the heart to improve the response to acute cardiac injury such as myocardial infarction or ischemia/reperfusion injury, preventing the progression to heart failure. It is becoming more apparent that cardioprotection is a concerted effort between multiple cell types and converging signaling pathways. However, the molecular mechanisms of cardioprotection are not completely understood. What is clear is that the mechanisms underlying this protection involve acute activation of transcriptional activators and their corresponding gene expression programs. Here, we review the known stress-dependent transcriptional programs that are activated in cardiomyocytes and cardiac fibroblasts to preserve function in the adult heart after injury. Focus is given to prominent transcriptional pathways such as mechanical stress or reactive oxygen species (ROS)-dependent activation of myocardin-related transcription factors (MRTFs) and transforming growth factor beta (TGFβ), and gene expression that positively regulates protective PI3K/Akt signaling. Together, these pathways modulate both beneficial and pathological responses to cardiac injury in a cell-specific manner.
Collapse
Affiliation(s)
- Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California - San Diego, 9500 Gilman Drive, Biomedical Sciences Building, La Jolla, CA 92093, USA.
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14624, USA.
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
138
|
Ma Y, Hu Y, Wu J, Wen J, Li S, Zhang L, Zhang J, Li Y, Li J. Epigallocatechin-3-gallate inhibits angiotensin II-induced cardiomyocyte hypertrophy via regulating Hippo signaling pathway in H9c2 rat cardiomyocytes. Acta Biochim Biophys Sin (Shanghai) 2019; 51:422-430. [PMID: 30877756 DOI: 10.1093/abbs/gmz018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 12/20/2022] Open
Abstract
Angiotensin II (AII) has been well known to induce cardiomyocyte hypertrophy. Epigallocatechin-3-gallate (EGCG) is the main active component of green tea and it has been shown to exhibit strong cardioprotective potential, although the underlying molecular mechanisms remain unclear. In this study, we investigated the role and mechanism of EGCG in preventing AII-induced cardiomyocyte hypertrophy using rat H9c2 cardiomyocytes cells. Reactive oxygen species assay, cell size, and mRNA expression of cardiac hypertrophy markers ANP and BNP were assessed in response to AII treatment. In addition, expression of proteins involved in Hippo signaling pathway were determined by western blot analysis. We found that AII treatment resulted in significant upregulation of ANP and BNP expression levels and increase in H9c2 cell size, which were markedly attenuated by EGCG treatment. Furthermore, our results suggested that EGCG inhibited AII-induced cardiac hypertrophy via regulating the Hippo signaling pathway. Therefore, EGCG may be an effective agent for preventing cardiac hypertrophy.
Collapse
Affiliation(s)
- Yuan Ma
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Yongjia Hu
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Jiawen Wu
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Junru Wen
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sen Li
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Lijuan Zhang
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Jie Zhang
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| | - Yanfei Li
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jue Li
- Institute of Clinical Epidemiology and Evidence-Based Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
139
|
Chen YA, Lu CY, Cheng TY, Pan SH, Chen HF, Chang NS. WW Domain-Containing Proteins YAP and TAZ in the Hippo Pathway as Key Regulators in Stemness Maintenance, Tissue Homeostasis, and Tumorigenesis. Front Oncol 2019; 9:60. [PMID: 30805310 PMCID: PMC6378284 DOI: 10.3389/fonc.2019.00060] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/21/2019] [Indexed: 12/29/2022] Open
Abstract
The Hippo pathway is a conserved signaling pathway originally defined in Drosophila melanogaster two decades ago. Deregulation of the Hippo pathway leads to significant overgrowth in phenotypes and ultimately initiation of tumorigenesis in various tissues. The major WW domain proteins in the Hippo pathway are YAP and TAZ, which regulate embryonic development, organ growth, tissue regeneration, stem cell pluripotency, and tumorigenesis. Recent reports reveal the novel roles of YAP/TAZ in establishing the precise balance of stem cell niches, promoting the production of induced pluripotent stem cells (iPSCs), and provoking signals for regeneration and cancer initiation. Activation of YAP/TAZ, for example, results in the expansion of progenitor cells, which promotes regeneration after tissue damage. YAP is highly expressed in self-renewing pluripotent stem cells. Overexpression of YAP halts stem cell differentiation and yet maintains the inherent stem cell properties. A success in reprograming iPSCs by the transfection of cells with Oct3/4, Sox2, and Yap expression constructs has recently been shown. In this review, we update the current knowledge and the latest progress in the WW domain proteins of the Hippo pathway in relevance to stem cell biology, and provide a thorough understanding in the tissue homeostasis and identification of potential targets to block tumor development. We also provide the regulatory role of tumor suppressor WWOX in the upstream of TGF-β, Hyal-2, and Wnt signaling that cross talks with the Hippo pathway.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yu Lu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tian-You Cheng
- Department of Optics and Photonics, National Central University, Chungli, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University, Taipei, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, United States.,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
140
|
Lauzi J, Anders F, Liu H, Pfeiffer N, Grus F, Thanos S, Arnhold S, Prokosch V. Neuroprotective and neuroregenerative effects of CRMP-5 on retinal ganglion cells in an experimental in vivo and in vitro model of glaucoma. PLoS One 2019; 14:e0207190. [PMID: 30673694 PMCID: PMC6343933 DOI: 10.1371/journal.pone.0207190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/28/2018] [Indexed: 01/15/2023] Open
Abstract
Purpose To analyze the potential neuro-protective and neuro-regenerative effects of Collapsin-response-mediator-protein-5 (CRMP-5) on retinal ganglion cells (RGCs) using in vitro and in vivo animal models of glaucoma. Methods Elevated intraocular pressure (IOP) was induced in adult female Sprague-Dawley (SD) rats by cauterization of three episcleral veins. Changes in CRMP-5 expression within the retinal proteome were analyzed via label-free mass spectrometry. In vitro, retinal explants were cultured under elevated pressure (60 mmHg) within a high-pressure incubation chamber with and without addition of different concentrations of CRMP-5 (4 μg/l, 200 μg/l and 400 μg/l). In addition, retinal explants were cultured under regenerative conditions with and without application of 200 μg/l CRMP-5 after performing an optic nerve crush (ONC). Thirdly, an antibody against Protein Kinase B (PKB) was added to examine the possible effects of CRMP-5. RGC count was performed. Number and length of the axons were determined and compared. To undermine a signal-transduction pathway via CRMP-5 and PKB microarray and immunohistochemistry were performed. Results CRMP-5 was downregulated threefold in animals showing chronically elevated IOP. The addition of CRMP-5 to retinal culture significantly increased RGC numbers under pressure in a dose-dependent manner and increased and elongated outgrowing axons in retinal explants significantly which could be blocked by PKB. Especially the number of neurites longer than 400 μm significantly increased after application of CRMP-5. CRMP-5 as well as PKB were detected higher in the experimental than in the control group. Conclusion CRMP-5 seems to play an important role in an animal model of glaucoma. Addition of CRMP-5 exerts neuro-protective and neuro-regenerative effects in vitro. This effect could be mediated via activation of PKB affecting intra-cellular apoptosis pathways.
Collapse
Affiliation(s)
- Jasmin Lauzi
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Fabian Anders
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Hanhan Liu
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Franz Grus
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Solon Thanos
- Department of Experimental Ophthalmology, School of Medicine, University of Münster, Münster, Germany
| | - Stefan Arnhold
- Institute of Veterinary-Anatomy, -Histology and–Embryology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Verena Prokosch
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
- * E-mail:
| |
Collapse
|
141
|
Byun J, Del Re DP, Zhai P, Ikeda S, Shirakabe A, Mizushima W, Miyamoto S, Brown JH, Sadoshima J. Yes-associated protein (YAP) mediates adaptive cardiac hypertrophy in response to pressure overload. J Biol Chem 2019; 294:3603-3617. [PMID: 30635403 DOI: 10.1074/jbc.ra118.006123] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/27/2018] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death globally, and heart failure is a major component of CVD-related morbidity and mortality. The development of cardiac hypertrophy in response to hemodynamic overload is initially considered to be beneficial; however, this adaptive response is limited and, in the presence of prolonged stress, will transition to heart failure. Yes-associated protein (YAP), the central downstream effector of the Hippo signaling pathway, regulates proliferation and survival in mammalian cells. Our previous work demonstrated that cardiac-specific loss of YAP leads to increased cardiomyocyte (CM) apoptosis and impaired CM hypertrophy during chronic myocardial infarction (MI) in the mouse heart. Because of its documented cardioprotective effects, we sought to determine the importance of YAP in response to acute pressure overload (PO). Our results indicate that endogenous YAP is activated in the heart during acute PO. YAP activation that depended upon RhoA was also observed in CMs subjected to cyclic stretch. To examine the function of endogenous YAP during acute PO, Yap +/ flox;Cre α-MHC (YAP-CHKO) and Yap +/ flox mice were subjected to transverse aortic constriction (TAC). We found that YAP-CHKO mice had attenuated cardiac hypertrophy and significant increases in CM apoptosis and fibrosis that correlated with worsened cardiac function after 1 week of TAC. Loss of CM YAP also impaired activation of the cardioprotective kinase Akt, which may underlie the YAP-CHKO phenotype. Together, these data indicate a prohypertrophic, prosurvival function of endogenous YAP and suggest a critical role for CM YAP in the adaptive response to acute PO.
Collapse
Affiliation(s)
- Jaemin Byun
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Dominic P Del Re
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Shohei Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Akihiro Shirakabe
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Wataru Mizushima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| | - Shigeki Miyamoto
- the Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Joan H Brown
- the Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey 07103 and
| |
Collapse
|
142
|
Zheng J, Peng B, Zhang Y, Ai F, Hu X. miR-9 knockdown inhibits hypoxia-induced cardiomyocyte apoptosis by targeting Yap1. Life Sci 2019; 219:129-135. [PMID: 30639391 DOI: 10.1016/j.lfs.2019.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022]
Abstract
AIMS Aberrantly expressed miRNAs are demonstrated to be involved in the development of congenital heart disease (CHD). miR-9 was proposed to be upregulated in cardiac tissues from CHD cases. However, the role of miR-9 in hypoxia-induced cardiomyocytes and the potential mechanism are far from being addressed. MAIN METHODS qRT-PCR and western blot analysis were performed to detect miR-9 and Yes-associated protein 1 (Yap1) expressions in hypoxic H9c2 cells. CCK-8, flow cytometry analysis, caspase-3/7 activity assay were applied to evaluate cell proliferation, apoptosis, and caspase-3/7 activity, respectively. The interaction between miR-9 and Yap1 was explored by luciferase reporter assay, qRT-PCR and western blot. KEY FINDINGS miR-9 was upregulated and Yap1 was downregulated in H9c2 cells in response to hypoxia in a time-dependent manner. Knockdown of miR-9 promoted cell proliferation, and inhibited apoptosis and caspase-3/7 activity in hypoxic H9c2 cells, while miR-9 overexpression exerted the opposite effects on hypoxic H9c2 cells. In addition, Yap1 was a direct target of miR-9 in H9c2 cells. Yap1 knockdown suppressed cell proliferation and promoted apoptosis in hypoxia-exposed H9c2 cells. Yap1 knockdown attenuated the effect of anti-miR-9 on cell proliferation and apoptosis in hypoxia-exposed H9c2 cells. SIGNIFICANCE miR-9 knockdown inhibited hypoxia-induced cardiomyocyte apoptosis by targeting Yap1. Our study provided a novel insight into the mechanism of the adaptation of cardiomyocytes to chronic hypoxia.
Collapse
Affiliation(s)
- Jiayong Zheng
- Department of Children's Heart Center, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Avenue, Zhengzhou 450000, China
| | - Bangtian Peng
- Department of Children's Heart Center, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Avenue, Zhengzhou 450000, China.
| | - Yanwei Zhang
- Department of Children's Heart Center, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Avenue, Zhengzhou 450000, China
| | - Feng Ai
- Department of Children's Heart Center, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Avenue, Zhengzhou 450000, China
| | - Xiaosong Hu
- Department of Children's Heart Center, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, No. 1 Fuwai Avenue, Zhengzhou 450000, China
| |
Collapse
|
143
|
Iliev A, Kotov G, Dimitrova IN, Landzhov B. Hypertension-induced changes in the rat myocardium during the development of cardiac hypertrophy - a comparison between the left and the right ventricle. Acta Histochem 2019; 121:16-28. [PMID: 30336951 DOI: 10.1016/j.acthis.2018.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023]
Abstract
The hypertrophy of the cardiac muscle is one of the most significant maladaptive mechanisms activated in response to increased workload. It is associated with histological and ultrastructural alterations, changes in the quantitative parameters and the expression of different enzymes. While the structural and functional consequences of systemic hypertension on the left ventricle have been well evaluated, the right ventricle has received less attention. The aim of the present study was to analyse and compare the changes in the left and right ventricle during the development of cardiac hypertrophy initiated by systemic hypertension in different age groups of spontaneously hypertensive rats. Therefore, we studied the histology and ultrastructure of the cells of the myocardium, evaluated the immunohistochemical expression of the enzyme neuronal nitric oxide synthase and conducted a quantitative analysis of several morphometric parameters. We used three groups of spontaneously hypertensive rats. For the quantitative analysis we also used three age groups of age- and weight-matched control animals (normotensive Wistar rats). In both ventricles, we described cardiomyocytic hypertrophy, focal myocytolysis and increased collagen deposition in the interstitial space. Our observations on the ultrastructural level were associated with changes in the cardiomyocytic nuclei, the arrangement, maturity and organisation of the myofibrils, the localisation and ultrastructure of the mitochondria, the development and maturity of the intercalated discs, as well as changes in the components of the interstitium. The immunohistochemical expression of neuronal nitric oxide synthase in the left ventricle was stronger than that in the right ventricle across all age groups. The comparative quantitative analysis revealed that changes in the studied morphometric parameters in the two ventricles occurred disproportionately. In conclusion, the present study characterised the development of cardiac hypertrophy in response to systemic hypertension in both ventricles and demonstrated the involvement of the right ventricle.
Collapse
|
144
|
Abstract
Hippo signaling is an evolutionarily conserved network that has a central role in regulating cell proliferation and cell fate to control organ growth and regeneration. It promotes activation of the LATS kinases, which control gene expression by inhibiting the activity of the transcriptional coactivator proteins YAP and TAZ in mammals and Yorkie in Drosophila. Diverse upstream inputs, including both biochemical cues and biomechanical cues, regulate Hippo signaling and enable it to have a key role as a sensor of cells' physical environment and an integrator of growth control signals. Several components of this pathway localize to cell-cell junctions and contribute to regulation of Hippo signaling by cell polarity, cell contacts, and the cytoskeleton. Downregulation of Hippo signaling promotes uncontrolled cell proliferation, impairs differentiation, and is associated with cancer. We review the current understanding of Hippo signaling and highlight progress in the elucidation of its regulatory mechanisms and biological functions.
Collapse
Affiliation(s)
- Jyoti R Misra
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| |
Collapse
|
145
|
Affiliation(s)
- Kartik Mani
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
- John Cochran VA Medical Center, St. Louis, Missouri
| | - Abhinav Diwan
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
- John Cochran VA Medical Center, St. Louis, Missouri
| |
Collapse
|
146
|
Liu X, Liu K, Li C, Cai J, Huang L, Chen H, Wang H, Zou J, Liu M, Wang K, Tan S, Zhang H. Heat-shock protein B1 upholds the cytoplasm reduced state to inhibit activation of the Hippo pathway in H9c2 cells. J Cell Physiol 2018; 234:5117-5133. [PMID: 30256412 DOI: 10.1002/jcp.27322] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/02/2018] [Indexed: 12/29/2022]
Abstract
Heat-shock protein B1 (HSPB1) is a multifunctional protein that protects against oxidative stress; however, its function in antioxidant pathways remains largely unknown. Here, we sought to determine the roles of HSPB1 in H9c2 cells subjected to oxidative stress. Using nonreducing sodium dodecyl sulfate polyacrylamide gel electrophoresis, we found that increased HSPB1 expression promoted the reduced states of glutathione reductase (GR), peroxiredoxin 1 (Prx1), and thioredoxin 1, whereas knockdown of HSPB1 attenuated these responses following oxidative stress. Increased HSPB1 expression promoted the activation of GR and thioredoxin reductase. Conversely, knockdown of HSPB1 attenuated these responses following oxidative stress. Importantly, overexpression of HSPB1 promoted the complex formation between HSPB1 and oxidized Prx1, leading to dephosphorylation of STE-mammalian STE20-like kinase 1 (MST1) in H9c2 cells exposed to H2 O 2 , whereas downregulation of HSPB1 induced the opposite results. Mechanistically, HSPB1 regulated the Hippo pathway by enhancing the dephosphorylation of MST1, resulting in reduced phosphorylation of LATS1 and Yes-associated protein (YAP). Moreover, HSPB1 regulated YAP-dependent gene expression. Thus, HSPB1 promoted the reduced state of endogenous antioxidant pathways following oxidative stress in H9c2 cells and improved the redox state of the cytoplasm via modulation of the Hippo signaling pathway.
Collapse
Affiliation(s)
- Xiehong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Ke Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Caiyan Li
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Jiaodi Cai
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Li Huang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Huan Chen
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Hao Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Jiang Zou
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Meidong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Sipin Tan
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Huali Zhang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| |
Collapse
|
147
|
Liu S, Martin JF. The regulation and function of the Hippo pathway in heart regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e335. [PMID: 30169913 DOI: 10.1002/wdev.335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/30/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Heart failure caused by cardiomyocyte loss and fibrosis is a leading cause of death worldwide. Although current treatments for heart failure such as heart transplantation and left ventricular assist device implantation have obvious value, new approaches are needed. Endogenous adult cardiomyocyte renewal is measurable but inefficient and inadequate in response to extensive acute heart damage. Stimulating self-renewal of endogenous cardiomyocytes holds great promise for heart repair. Uncovering the genetic mechanisms underlying cardiomyocyte renewal is a critical step in developing new approaches to repairing the heart. Recent studies have revealed that the inhibition of the Hippo pathway is sufficient to promote the proliferation of endogenous cardiomyocytes, indicating that the manipulation of the Hippo pathway in the heart may be a promising treatment for heart failure in the future. We summarize recent findings that have shed light on the function of the Hippo pathway in heart regeneration. We also discuss the mechanisms by which Hippo pathway inhibition promotes heart regeneration and how the Hippo pathway responds to different types of injury or stress during the regenerative process. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
148
|
Xu W, Yang Z, Xie C, Zhu Y, Shu X, Zhang Z, Li N, Chai N, Zhang S, Wu K, Nie Y, Lu N. PTEN lipid phosphatase inactivation links the hippo and PI3K/Akt pathways to induce gastric tumorigenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:198. [PMID: 30134988 PMCID: PMC6104022 DOI: 10.1186/s13046-018-0795-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/12/2018] [Indexed: 02/06/2023]
Abstract
Background Phosphatase and tensin homolog (PTEN) is an important tumor suppressor gene, and its encoded protein has activities of both a protein phosphatase and a lipid phosphatase. However, the substitution effect of protein phosphatase activity remains unclear. PI3K/Akt is the most common pathway negatively regulated by PTEN. The Hippo and PI3K/Akt pathways have a joint effect in regulating cell proliferation and apoptosis. Therefore, how PTEN lipid phosphatase inactivation contributes to the occurrence and development of gastric cancer and the potential role of the Hippo and PI3K/Akt pathways in PTEN lipid phosphatase inactivation mediated gastric tumorigenesis remain to be explored. Methods Immunohistochemical staining was performed to detect the expression of p-PTEN and YAP in a gastric cancer tissue microarray. Stable cell lines expressing a wild-type or dominant-negative mutant PTEN were established. The proliferation and migration of stable cells were detected by MTT, BrdU, and colony-formation, transwell assay and high content analysis in vitro, and tumor growth differences were observed in xenograft nude mice. Changes in the expression of key molecules in the Hippo and Akt signaling pathways were detected by western blot. Nuclear-cytoplasm separation, immunofluorescence and coimmunoprecipitation analyses were conducted to explore the dysregulation of Hippo in the stable cell lines. Results PTEN lipid phosphatase inactivation strongly promoted the proliferation and migration of gastric cancer cells in vitro and tumor growth in vivo. A immunohistochemical analysis of gastric cancer tissues revealed a significant correlation between phosphorylated PTEN and nuclear YAP expression, and both were determined to be independent prognostic factors for gastric cancer. Mechanistically, PTEN lipid phosphatase inactivation abolished the MOB1-LATS1/2 interaction, decreased YAP phosphorylation and finally promoted YAP nuclear translocation, which enhanced the synergistic effect of YAP-TEAD, thus inducing cell proliferation and migration. Moreover, PTEN lipid phosphatase inactivation promoted the PI3K/Akt pathway, and disruption of YAP-TEAD-driven transcription decreased the activation of Akt in a dose-dependent manner. Conclusions Taken together, our findings indicate that PTEN lipid phosphatase inactivation links the Hippo and PI3K/Akt pathways to promote gastric tumorigenesis and cancer development. Electronic supplementary material The online version of this article (10.1186/s13046-018-0795-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhen Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xu Shu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhe Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Nianshuang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Na Chai
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Song Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Kaichun Wu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yongzhan Nie
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
149
|
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
|
150
|
|