1
|
Meng X, Du W, Sun Z. Fine particulate matter‑induced cardiac developmental toxicity (Review). Exp Ther Med 2025; 29:6. [PMID: 39534282 PMCID: PMC11552469 DOI: 10.3892/etm.2024.12756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Fine particulate matter (PM2.5) has become an important risk factor threatening human health. Epidemiological and toxicological investigations have revealed that PM2.5 not only leads to cardiovascular dysfunction, but it also gives rise to various adverse health effects on the human body, such as cardiovascular and cerebrovascular diseases, cancers, neurodevelopmental disorders, depression and autism. PM2.5 is able to penetrate both respiratory and placental barriers, thereby resulting in negative effects on fetal development. A large body of epidemiological evidences has suggested that gestational exposure to PM2.5 increases the incidence of congenital diseases in offspring, including congenital heart defects. In addition, animal model studies have revealed that gestational exposure to PM2.5 can disrupt normal heart development in offspring, although the potential molecular mechanisms have yet to be fully elucidated. The aim of the present review was to provide a brief overview of what is currently known regarding the molecular mechanisms underlying cardiac developmental toxicity in offspring induced by gestational exposure to PM2.5.
Collapse
Affiliation(s)
- Xiangjiang Meng
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Weiyuan Du
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Zongli Sun
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| |
Collapse
|
2
|
Zhang M, Liu C, Zhao L, Zhang X, Su Y. The Emerging Role of Protein Phosphatase in Regeneration. Life (Basel) 2023; 13:life13051216. [PMID: 37240861 DOI: 10.3390/life13051216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Maintaining normal cellular behavior is essential for the survival of organisms. One of the main mechanisms to control cellular behavior is protein phosphorylation. The process of protein phosphorylation is reversible under the regulation of protein kinases and protein phosphatases. The importance of kinases in numerous cellular processes has been well recognized. In recent years, protein phosphatases have also been demonstrated to function actively and specifically in various cellular processes and thus have gained more and more attention from researchers. In the animal kingdom, regeneration frequently occurs to replace or repair damaged or missing tissues. Emerging evidence has revealed that protein phosphatases are crucial for organ regeneration. In this review, after providing a brief overview of the classification of protein phosphatases and their functions in several representative developmental processes, we highlight the critical roles that protein phosphatases play in organ regeneration by summarizing the most recent research on the function and underlying mechanism of protein phosphatase in the regeneration of the liver, bone, neuron, and heart in vertebrates.
Collapse
Affiliation(s)
- Meiling Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Chenglin Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Xuejiao Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
3
|
Hwang S, Kim SH, Yoo KH, Chung MH, Lee JW, Son KH. Exogenous 8-hydroxydeoxyguanosine attenuates doxorubicin-induced cardiotoxicity by decreasing pyroptosis in H9c2 cardiomyocytes. BMC Mol Cell Biol 2022; 23:55. [DOI: 10.1186/s12860-022-00454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractDoxorubicin (DOX), which is widely used in cancer treatment, can induce cardiomyopathy. One of the main mechanisms whereby DOX induces cardiotoxicity involves pyroptosis through the NLR family pyrin domain containing 3 (NLRP3) inflammasome and gasdermin D (GSDMD). Increased NAPDH oxidase (NOX) and oxidative stress trigger pyroptosis. Exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases reactive oxygen species (ROS) production by inactivating NOX. Here, we examined whether 8-OHdG treatment can attenuate DOX-induced pyroptosis in H9c2 cardiomyocytes. Exposure to DOX increased the peroxidative glutathione redox status and NOX1/2/4, toll-like receptor (TLR)2/4, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) expression, while an additional 8-OHdG treatment attenuated these effects. Furthermore, DOX induced higher expression of NLRP3 inflammasome components, including NLRP3, apoptosis-associated speck-like protein containing a c-terminal caspase recruitment domain (ASC), and pro-caspase-1. Moreover, it increased caspase-1 activity, a marker of pyroptosis, and interleukin (IL)-1β expression. All these effects were attenuated by 8-OHdG treatment. In addition, the expression of the cardiotoxicity markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was increased by DOX, whereas the increase of ANP and BNP induced by DOX treatment was reversed by 8-OHdG. In conclusion, exogenous 8-OHdG attenuated DOX-induced pyroptosis by decreasing the expression of NOX1/2/3, TLR2/4, and NF-κB. Thus, 8-OHdG may attenuate DOX-induced cardiotoxicity through the inhibition of pyroptosis.
Collapse
|
4
|
Huang Y, Wang WF, Huang CX, Li XH, Liu H, Wang HL. miR-731 modulates the zebrafish heart morphogenesis via targeting Calcineurin/Nfatc3a pathway. Biochim Biophys Acta Gen Subj 2022; 1866:130133. [PMID: 35346765 DOI: 10.1016/j.bbagen.2022.130133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Zebrafish miR-731 is orthologous of human miR-425, which has been demonstrated to have cardio-protective roles by a variety of mechanisms. The miR-731 morphants show pericardium enlargement, and many DEGs (differentially expressed genes) are enriched in 'Cardiac muscle contraction' and 'Calcium signaling pathway', implying that miR-731 plays a potential role in heart function and development. However,the in vivo physiological role of miR-731 in the heart needs to be fully defined. METHODS Zebrafish miR-731 morphants were generated by morpholino knockdown, and miR-731 knockout zebrafish was generated by CRISRP/Cas9. We observed cardiac morphogenesis based on whole-mount in situ hybridization. Furthermore, RNA-seq and qRT-PCR were used to elucidate the molecular mechanism and analyze the gene expression. Double luciferase verification and Western blot were used to verify the target gene. RESULTS The depletion of miR-731 in zebrafish embryos caused the deficiency of cardiac development and function, which was associated with reduced heart rate, ventricular enlargement and heart looping disorder. In addition, mechanistic study demonstrated that Calcineurin/Nfatc3a signaling involved in miR-731 depletion induced abnormal cardiac function and developmental defects. CONCLUSION MiR-731 regulates cardiac function and morphogenesis through Calcineurin/Nfatc3a signaling. GENERAL SIGNIFICANCE Our studies highlight the potential importance of miR-731 in cardiac development.
Collapse
Affiliation(s)
- Yan Huang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Wei-Feng Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Chun-Xiao Huang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Xuan-Hui Li
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Hong Liu
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Huan-Ling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, Hubei, PR China.
| |
Collapse
|
5
|
Shimizu K, Sunagawa Y, Funamoto M, Honda H, Katanasaka Y, Murai N, Kawase Y, Hirako Y, Katagiri T, Yabe H, Shimizu S, Sari N, Wada H, Hasegawa K, Morimoto T. The Selective Serotonin 2A Receptor Antagonist Sarpogrelate Prevents Cardiac Hypertrophy and Systolic Dysfunction via Inhibition of the ERK1/2-GATA4 Signaling Pathway. Pharmaceuticals (Basel) 2021; 14:ph14121268. [PMID: 34959669 PMCID: PMC8708651 DOI: 10.3390/ph14121268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/15/2021] [Accepted: 12/01/2021] [Indexed: 01/02/2023] Open
Abstract
Drug repositioning has recently emerged as a strategy for developing new treatments at low cost. In this study, we used a library of approved drugs to screen for compounds that suppress cardiomyocyte hypertrophy. We identified the antiplatelet drug sarpogrelate, a selective serotonin-2A (5-HT2A) receptor antagonist, and investigated the drug's anti-hypertrophic effect in cultured cardiomyocytes and its effect on heart failure in vivo. Primary cultured cardiomyocytes pretreated with sarpogrelate were stimulated with angiotensin II, endothelin-1, or phenylephrine. Immunofluorescence staining showed that sarpogrelate suppressed the cardiomyocyte hypertrophy induced by each of the stimuli. Western blotting analysis revealed that 5-HT2A receptor level was not changed by phenylephrine, and that sarpogrelate suppressed phenylephrine-induced phosphorylation of ERK1/2 and GATA4. C57BL/6J male mice were subjected to transverse aortic constriction (TAC) surgery followed by daily oral administration of sarpogrelate for 8 weeks. Echocardiography showed that 5 mg/kg of sarpogrelate suppressed TAC-induced cardiac hypertrophy and systolic dysfunction. Western blotting revealed that sarpogrelate suppressed TAC-induced phosphorylation of ERK1/2 and GATA4. These results indicate that sarpogrelate suppresses the development of heart failure and that it does so at least in part by inhibiting the ERK1/2-GATA4 signaling pathway.
Collapse
Affiliation(s)
- Kana Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
| | - Hiroki Honda
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Noriyuki Murai
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Yuto Kawase
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Yuta Hirako
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Takahiro Katagiri
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Harumi Yabe
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Satoshi Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
| | - Nurmila Sari
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
| | - Hiromichi Wada
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
| | - Koji Hasegawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.S.); (Y.S.); (M.F.); (H.H.); (Y.K.); (N.M.); (Y.K.); (Y.H.); (T.K.); (H.Y.); (S.S.); (N.S.); (K.H.)
- National Hospital Organization Kyoto Medical Center, Division of Translational Research, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, Shizuoka 420-8527, Japan
- Correspondence: ; Tel.: +81-54-264-5763
| |
Collapse
|
6
|
Papaver somniferum L. mediated novel bioinspired lead oxide (PbO) and iron oxide (Fe2O3) nanoparticles: In-vitro biological applications, biocompatibility and their potential towards HepG2 cell line. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109740. [DOI: 10.1016/j.msec.2019.109740] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/27/2019] [Accepted: 05/10/2019] [Indexed: 11/22/2022]
|
7
|
Chen J, Wang S, Pang S, Cui Y, Yan B, Hawley RG. Functional genetic variants of the GATA4 gene promoter in acute myocardial infarction. Mol Med Rep 2019; 19:2861-2868. [PMID: 30720078 DOI: 10.3892/mmr.2019.9914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/25/2019] [Indexed: 11/05/2022] Open
Abstract
Coronary artery disease (CAD), including acute myocardial infarction (AMI), is a common complex disease; however, the genetic causes remain largely unknown. Recent epidemiological investigations indicated that the incidence of CAD in patients with congenital heart diseases is markedly higher than that observed in healthy controls. It was therefore hypothesized that the dysregulated expression of cardiac developmental genes may be involved in CAD development. GATA binding protein 4 (GATA4) serves essential roles in heart development and coronary vessel formation. In the present study, the GATA4 gene promoter was analyzed in patients with AMI (n=395) and in ethnically‑matched healthy controls (n=397). A total of 14 DNA variants were identified, including two single‑nucleotide polymorphisms. Three novel heterozygous DNA variants (g.31806C>T, g.31900G>C and g.32241C>T) were reported in three patients with AMI. These DNA variants significantly increased the activity of the GATA4 gene promoter. The electrophoretic mobility shift assay revealed that the DNA variant g.32241C>T influenced the binding ability of transcription factors. Taken together, the DNA variants may alter GATA4 gene promoter activity and affect GATA4 levels, thus contributing to AMI development.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Shuai Wang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Robert G Hawley
- Department of Anatomy and Regenerative Biology, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
8
|
Wu X, Pan B, Liu L, Zhao W, Zhu J, Huang X, Tian J. In utero exposure to PM2.5 during gestation caused adult cardiac hypertrophy through histone acetylation modification. J Cell Biochem 2018; 120:4375-4384. [PMID: 30269375 DOI: 10.1002/jcb.27723] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/29/2018] [Indexed: 01/27/2023]
Affiliation(s)
- Xiaoqi Wu
- Heart Centre, Children’s Hospital of Chongqing Medical University Chongqing China
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education Chongqing China
- Key Laboratory of Pediatrics Chongqing China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders Chongqing China
| | - Bo Pan
- Heart Centre, Children’s Hospital of Chongqing Medical University Chongqing China
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education Chongqing China
- Key Laboratory of Pediatrics Chongqing China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders Chongqing China
| | - Lingjuan Liu
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education Chongqing China
- Key Laboratory of Pediatrics Chongqing China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders Chongqing China
| | - Weian Zhao
- Heart Centre, Children’s Hospital of Chongqing Medical University Chongqing China
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education Chongqing China
- Key Laboratory of Pediatrics Chongqing China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders Chongqing China
| | - Jing Zhu
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education Chongqing China
- Key Laboratory of Pediatrics Chongqing China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders Chongqing China
| | - Xupei Huang
- Department of Biomedical Science Charlie E. Schmidt College of Medicine, Florida Atlantic University Boca Raton Florida
| | - Jie Tian
- Heart Centre, Children’s Hospital of Chongqing Medical University Chongqing China
| |
Collapse
|
9
|
Abstract
GATA transcription factors are emerging as critical players in mammalian reproductive development and function. GATA-4 contributes to fetal male gonadal development by regulating genes mediating Müllerian duct regression and the onset of testosterone production. GATA-2 expression appears to be sexually dimorphic being transiently expressed in the germ cell lineage of the fetal ovary but not the fetal testis. In the reproductive system, GATA-1 is exclusively expressed in Sertoli cells at specific seminiferous tubule stages. In addition, GATA-4 and GATA-6 are localized primary to ovarian and testicular somatic cells. The majority of cell transfection studies demonstrate that GATA-1 and GATA-4 can stimulate inhibin subunit gene promoter constructs. Other studies provide strong evidence that GATA-4 and GATA-6 can activate genes mediating gonadal cell steroidogenesis. GATA-2 and GATA-3 are found in pituitary and placental cells and can regulate alpha-glycoprotein subunit gene expression. Gonadal expression and activation of GATAs appear to be regulated in part by gonadotropin signaling via the cyclic AMP-protein kinase A pathway. This review will cover the current knowledge regarding GATA expression and function at all levels of the reproductive axis.
Collapse
Affiliation(s)
- Holly A LaVoie
- Department of Cell and Developmental Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USA.
| |
Collapse
|
10
|
Katanasaka Y, Suzuki H, Sunagawa Y, Hasegawa K, Morimoto T. Regulation of Cardiac Transcription Factor GATA4 by Post-Translational Modification in Cardiomyocyte Hypertrophy and Heart Failure. Int Heart J 2016; 57:672-675. [PMID: 27818483 DOI: 10.1536/ihj.16-404] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Heart failure is a leading cause of cardiovascular mortality in industrialized countries. During development and deterioration of heart failure, cardiomyocytes undergo maladaptive hypertrophy, and changes in the cellular phenotype are accompanied by reinduction of the fetal gene program. Gene expression in cardiomyocytes is regulated by various nuclear transcription factors, co-activators, and co-repressors. The zinc finger protein GATA4 is one such transcription factor involved in the regulation of cardiomyocyte hypertrophy. In response to hypertrophic stimuli such as those involving the sympathetic nervous and renin-angiotensin systems, changes in protein interaction and/or post-translational modifications of GATA4 cause hypertrophic gene transcription in cardiomyocytes. In this article, we focus on cardiac nuclear signaling molecules, especially GATA4, that are promising as potential targets for heart failure therapy.
Collapse
Affiliation(s)
- Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | | | | | | | | |
Collapse
|
11
|
ANGPTL2 activity in cardiac pathologies accelerates heart failure by perturbing cardiac function and energy metabolism. Nat Commun 2016; 7:13016. [PMID: 27677409 PMCID: PMC5052800 DOI: 10.1038/ncomms13016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/25/2016] [Indexed: 12/21/2022] Open
Abstract
A cardioprotective response that alters ventricular contractility or promotes cardiomyocyte enlargement occurs with increased workload in conditions such as hypertension. When that response is excessive, pathological cardiac remodelling occurs, which can progress to heart failure, a leading cause of death worldwide. Mechanisms underlying this response are not fully understood. Here, we report that expression of angiopoietin-like protein 2 (ANGPTL2) increases in pathologically-remodeled hearts of mice and humans, while decreased cardiac ANGPTL2 expression occurs in physiological cardiac remodelling induced by endurance training in mice. Mice overexpressing ANGPTL2 in heart show cardiac dysfunction caused by both inactivation of AKT and sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a signalling and decreased myocardial energy metabolism. Conversely, Angptl2 knockout mice exhibit increased left ventricular contractility and upregulated AKT-SERCA2a signalling and energy metabolism. Finally, ANGPTL2-knockdown in mice subjected to pressure overload ameliorates cardiac dysfunction. Overall, these studies suggest that therapeutic ANGPTL2 suppression could antagonize development of heart failure. Heart responds to increased workload by enlarging cardiomyocytes to preserve function, but in pathologies hypertrophy leads to heart failure. Here the authors show that ANGPTL2 activity in the heart is critical for determining beneficial vs. pathological hypertrophy via its effect on AKT-SERCA2a signaling and myocardial energy.
Collapse
|
12
|
Watts RP, Bilska I, Diab S, Dunster KR, Bulmer AC, Barnett AG, Fraser JF. Novel 24-h ovine model of brain death to study the profile of the endothelin axis during cardiopulmonary injury. Intensive Care Med Exp 2015; 3:31. [PMID: 26596583 PMCID: PMC4656265 DOI: 10.1186/s40635-015-0067-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 11/13/2015] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Upregulation of the endothelin axis has been observed in pulmonary tissue after brain death, contributing to primary graft dysfunction and ischaemia reperfusion injury. The current study aimed to develop a novel, 24-h, clinically relevant, ovine model of brain death to investigate the profile of the endothelin axis during brain death-associated cardiopulmonary injury. We hypothesised that brain death in sheep would also result in demonstrable injury to other transplantable organs. METHODS Twelve merino cross ewes were randomised into two groups. Following induction of general anaesthesia and placement of invasive monitoring, brain death was induced in six animals by inflation of an extradural catheter. All animals were supported in an intensive care unit environment for 24 h. Animal management reflected current human donor management, including administration of vasopressors, inotropes and hormone resuscitation therapy. Activation of the endothelin axis and transplantable organ injury were assessed using ELISA, immunohistochemistry and standard biochemical markers. RESULTS All animals were successfully supported for 24 h. ELISA suggested early endothelin-1 and big endothelin-1 release, peaking 1 and 6 h after BD, respectively, but there was no difference at 24 h. Immunohistochemistry confirmed the presence of the endothelin axis in pulmonary tissue. Brain dead animals demonstrated tachycardia and hypertension, followed by haemodynamic collapse, typified by a reduction in systemic vascular resistance to 46 ± 1 % of baseline. Mean pulmonary artery pressure rose to 186 ± 20 % of baseline at induction and remained elevated throughout the protocol, reaching 25 ± 2.2 mmHg at 24 h. Right ventricular stroke work increased 25.9 % above baseline by 24 h. Systemic markers of cardiac and hepatocellular injury were significantly elevated, with no evidence of renal dysfunction. CONCLUSIONS This novel, clinically relevant, ovine model of brain death demonstrated that increased pulmonary artery pressures are observed after brain death. This may contribute to right ventricular dysfunction and pulmonary injury. The development of this model will allow for further investigation of therapeutic strategies to minimise the deleterious effects of brain death on potentially transplantable organs.
Collapse
Affiliation(s)
- Ryan P Watts
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Queensland, Australia.
- University of Queensland, Brisbane, Queensland, Australia.
- Royal Brisbane and Women's Hospital, Herston, Queensland, Australia.
| | - Izabela Bilska
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Queensland, Australia.
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia.
| | - Sara Diab
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Queensland, Australia.
| | - Kimble R Dunster
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Queensland, Australia.
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - Andrew C Bulmer
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Southport, Queensland, Australia.
| | - Adrian G Barnett
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Queensland, Australia.
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.
- University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
13
|
Clapp LH, Gurung R. The mechanistic basis of prostacyclin and its stable analogues in pulmonary arterial hypertension: Role of membrane versus nuclear receptors. Prostaglandins Other Lipid Mediat 2015; 120:56-71. [PMID: 25917921 DOI: 10.1016/j.prostaglandins.2015.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of distal pulmonary arteries in which patients suffer from elevated pulmonary arterial pressure, extensive vascular remodelling and right ventricular failure. To date prostacyclin (PGI2) therapy remains the most efficacious treatment for PAH and is the only approved monotherapy to have a positive impact on long-term survival. A key thing to note is that improvement exceeds that predicted from vasodilator testing strongly suggesting that additional mechanisms contribute to the therapeutic benefit of prostacyclins in PAH. Given these agents have potent antiproliferative, anti-inflammatory and endothelial regenerating properties suggests therapeutic benefit might result from a slowing, stabilization or even some reversal of vascular remodelling in vivo. This review discusses evidence that the pharmacology of each prostacyclin (IP) receptor agonist so far developed is distinct, with non-IP receptor targets clearly contributing to the therapeutic and side effect profile of PGI2 (EP3), iloprost (EP1), treprostinil (EP2, DP1) along with a family of nuclear receptors known as peroxisome proliferator-activated receptors (PPARs), to which PGI2 and some analogues directly bind. These targets are functionally expressed to varying degrees in arteries, veins, platelets, fibroblasts and inflammatory cells and are likely to be involved in the biological actions of prostacylins. Recently, a highly selective IP agonist, selexipag has been developed for PAH. This agent should prove useful in distinguishing IP from other prostanoid receptors or PPAR binding effects in human tissue. It remains to be determined whether selectivity for the IP receptor gives rise to a superior or inferior clinical benefit in PAH.
Collapse
Affiliation(s)
- Lucie H Clapp
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK.
| | - Rijan Gurung
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK
| |
Collapse
|
14
|
Cardiopathogenic mediators generated by GATA4 signaling upon co-activation with endothelin-1 and Trypanosoma cruzi infection. Microb Pathog 2014; 73:47-52. [DOI: 10.1016/j.micpath.2014.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/26/2014] [Accepted: 06/03/2014] [Indexed: 01/17/2023]
|
15
|
Intracrine endothelin signaling evokes IP3-dependent increases in nucleoplasmic Ca²⁺ in adult cardiac myocytes. J Mol Cell Cardiol 2013; 62:189-202. [PMID: 23756157 DOI: 10.1016/j.yjmcc.2013.05.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 12/17/2022]
Abstract
Endothelin receptors are present on the nuclear membranes in adult cardiac ventricular myocytes. The objectives of the present study were to determine 1) which endothelin receptor subtype is in cardiac nuclear membranes, 2) if the receptor and ligand traffic from the cell surface to the nucleus, and 3) the effect of increased intracellular ET-1 on nuclear Ca(2+) signaling. Confocal microscopy using fluorescently-labeled endothelin analogs confirmed the presence of ETB at the nuclear membrane of rat cardiomyocytes in skinned-cells and isolated nuclei. Furthermore, in both cardiac myocytes and aortic endothelial cells, endocytosed ET:ETB complexes translocated to lysosomes and not the nuclear envelope. Although ETA and ETB can form heterodimers, the presence or absence of ETA did not alter ETB trafficking. Treatment of isolated nuclei with peptide: N-glycosidase F did not alter the electrophoretic mobility of ETB. The absence of N-glycosylation further indicates that these receptors did not originate at the cell surface. Intracellular photolysis of a caged ET-1 analog ([Trp-ODMNB(21)]ET-1) evoked an increase in nucleoplasmic Ca(2+) ([Ca(2+)]n) that was attenuated by inositol 1,4,5-trisphosphate receptor inhibitor 2-aminoethoxydiphenyl borate and prevented by pre-treatment with ryanodine. A caged cell-permeable analog of the ETB-selective antagonist IRL-2500 blocked the ability of intracellular cET-1 to increase [Ca(2+)]n whereas extracellular application of ETA and ETB receptor antagonists did not. These data suggest that 1) the endothelin receptor in the cardiac nuclear membranes is ETB, 2) ETB traffics directly to the nuclear membrane after biosynthesis, 3) exogenous endothelins are not ligands for ETB on nuclear membranes, and 4) ETB associated with the nuclear membranes regulates nuclear Ca(2+) signaling.
Collapse
|
16
|
Liu JF, Zhao SH, Wu SS. Depleting NFAT1 expression inhibits the ability of invasion and migration of human lung cancer cells. Cancer Cell Int 2013; 13:41. [PMID: 23663403 PMCID: PMC3658948 DOI: 10.1186/1475-2867-13-41] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/15/2013] [Indexed: 11/10/2022] Open
Abstract
Background Nuclear factor of activated T-cells (NFAT) is a general name applied to a family of transcription factors shown to be important in immune response. One or more members of the NFAT family are expressed in most cells of the immune system. NFAT1 is considered to involve in the development of cardiac, skeletal muscle, nervous systems, and tumorigenesis. Methods In the current study, we analyzed MEKK1 expression in 159 surgically resection non-small cell lung cancer patient’s samples by immunohistochemistry and determined its role in SK-EMS-1 cells via RNAi experiment. Results The abilities of invasion, motility, and adhesion of SK-EMS-1 cells were detected by transwell assay, wound healing assay and adhesion assay, respectively. The result showed NFAT1 was highly expressed in lung tumor tissues instead of adjacent lung tissues (54.1% vs 8.8%, p < 0.05); its overexpression was positively correlated with lymph node metastasis (p < 0.05). Depleting its expression in SK-EMS-1 cells can inhibit its invasion and migration abilities significantly (p < 0.05); and also can reduce proliferation of lung cancer cells (p < 0.05). Conclusion Our study showed NFAT1 plays an important role in origination, invasion and metastasis of non-small lung cancer cells; its underlying action mechanism needs further study.
Collapse
Affiliation(s)
- Ji-Fu Liu
- Department of thoracic surgery, General Hospital of Beijing Military Region, Nan Men Cang 5, Dongcheng District, Beijing, China.
| | | | | |
Collapse
|
17
|
Nagendran J, Sutendra G, Paterson I, Champion HC, Webster L, Chiu B, Haromy A, Rebeyka IM, Ross DB, Michelakis ED. Endothelin axis is upregulated in human and rat right ventricular hypertrophy. Circ Res 2012; 112:347-54. [PMID: 23233754 DOI: 10.1161/circresaha.111.300448] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Right ventricular (RV) function is the most important determinant of morbidity and mortality in pulmonary arterial hypertension (PAH). Endothelin (ET)-1 receptor antagonists (ERAs) are approved therapies for PAH. It is not known whether ERAs have effects on the RV, in addition to their vasodilating/antiproliferative effects in pulmonary arteries. OBJECTIVE We hypothesized that the ET axis is upregulated in RV hypertrophy (RVH) and that ERAs have direct effects on the RV myocardium. METHODS AND RESULTS RV myocardial samples from 34 patients with RVH were compared with 16 nonhypertrophied RV samples, and from rats with normal RV versus RVH attributable to PAH. Confocal immunohistochemistry showed that RVH myocardial ET type A (but not type B) receptor and ET-1 protein levels were increased compared with the nonhypertrophied RVs and positively correlated with the degree of RVH (RV thickness/body surface area; r(2)=0.838 and r(2)=0.818, respectively; P<0.01). These results were recapitulated in the rat model. In modified Langendorff perfusions, ERAs (BQ-123 and bosentan 10(-7,-6,-5) mol/L) decreased contractility in the hypertrophied, but not normal RV, in a dose-dependent manner (P<0.01). CONCLUSIONS Patients and rats with PAH have an upregulation of the myocardial ET axis in RVH. This might be a compensatory mechanism to preserve RV contractility, as the afterload increases. ERAs use might potentially worsen RV function, and this could explain some of the peripheral edema noted clinically with these agents. Further studies are required to evaluate the effects of ERAs on the RV in patients with RVH and PAH.
Collapse
Affiliation(s)
- Jayan Nagendran
- Department of Medicine and Pulmonary Hypertension Program, University of Alberta and Mazankowski Alberta Heart Institute, 2C2 WMC, 8440-112 St, Edmonton, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Hamad EA, Zhu W, Chan TO, Myers V, Gao E, Li X, Zhang J, Song J, Zhang XQ, Cheung JY, Koch W, Feldman AM. Cardioprotection of controlled and cardiac-specific over-expression of A(2A)-adenosine receptor in the pressure overload. PLoS One 2012; 7:e39919. [PMID: 22792196 PMCID: PMC3391213 DOI: 10.1371/journal.pone.0039919] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 05/29/2012] [Indexed: 11/18/2022] Open
Abstract
Adenosine binds to three G protein-coupled receptors (R) located on the cardiomyocyte (A(1)-R, A(2A)-R and A(3)-R) and provides cardiac protection during both ischemic and load-induced stress. While the role of adenosine receptor-subtypes has been well defined in the setting of ischemia-reperfusion, far less is known regarding their roles in protecting the heart during other forms of cardiac stress. Because of its ability to increase cardiac contractility and heart rate, we hypothesized that enhanced signaling through A(2A)-R would protect the heart during the stress of transverse aortic constriction (TAC). Using a cardiac-specific and inducible promoter, we selectively over-expressed A(2A)-R in FVB mice. Echocardiograms were obtained at baseline, 2, 4, 8, 12, 14 weeks and hearts were harvested at 14 weeks, when WT mice developed a significant decrease in cardiac function, an increase in end systolic and diastolic dimensions, a higher heart weight to body weight ratio (HW/BW), and marked fibrosis when compared with sham-operated WT. More importantly, these changes were significantly attenuated by over expression of the A(2A)-R. Furthermore, WT mice also demonstrated marked increases in the hypertrophic genes β-myosin heavy chain (β-MHC), and atrial natriuretic factor (ANF)--changes that are mediated by activation of the transcription factor GATA-4. Levels of the mRNAs encoding β-MHC, ANP, and GATA-4 were significantly lower in myocardium from A(2A)-R TG mice after TAC when compared with WT and sham-operated controls. In addition, three inflammatory factors genes encoding cysteine dioxygenase, complement component 3, and serine peptidase inhibitor, member 3N, were enhanced in WT TAC mice, but their expression was suppressed in A(2A)-R TG mice. A(2A)-R over-expression is protective against pressure-induced heart failure secondary to TAC. These cardioprotective effects are associated with attenuation of GATA-4 expression and inflammatory factors. The A(2A)-R may provide a novel new target for pharmacologic therapy in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Eman A. Hamad
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Weizhong Zhu
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Tung O. Chan
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Valerie Myers
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Erhe Gao
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xue Li
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Jin Zhang
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Jianliang Song
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xue-Qian Zhang
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joseph Y. Cheung
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Walter Koch
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Arthur M. Feldman
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
19
|
Cardiac expression of ms1/STARS, a novel gene involved in cardiac development and disease, is regulated by GATA4. Mol Cell Biol 2012; 32:1830-43. [PMID: 22431517 DOI: 10.1128/mcb.06374-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ms1/STARS is a novel muscle-specific actin-binding protein that specifically modulates the myocardin-related transcription factor (MRTF)-serum response factor (SRF) regulatory axis within striated muscle. This ms1/STARS-dependent regulatory axis is of central importance within the cardiac gene regulatory network and has been implicated in cardiac development and postnatal cardiac function/homeostasis. The dysregulation of ms1/STARS is associated with and causative of pathological cardiac phenotypes, including cardiac hypertrophy and cardiomyopathy. In order to gain an understanding of the mechanisms governing ms1/STARS expression in the heart, we have coupled a comparative genomic in silico analysis with reporter, gain-of-function, and loss-of-function approaches. Through this integrated analysis, we have identified three evolutionarily conserved regions (ECRs), α, SINA, and DINA, that act as cis-regulatory modules and confer differential cardiac cell-specific activity. Two of these ECRs, α and DINA, displayed distinct regulatory sensitivity to the core cardiac transcription factor GATA4. Overall, our results demonstrate that within embryonic, neonatal, and adult hearts, GATA4 represses ms1/STARS expression with the pathologically associated depletion of GATA4 (type 1/type 2 diabetic models), resulting in ms1/STARS upregulation. This GATA4-dependent repression of ms1/STARS expression has major implications for MRTF-SRF signaling in the context of cardiac development and disease.
Collapse
|
20
|
Hu X, Li T, Zhang C, Liu Y, Xu M, Wang W, Jia Z, Ma K, Zhang Y, Zhou C. GATA4 regulates ANF expression synergistically with Sp1 in a cardiac hypertrophy model. J Cell Mol Med 2012; 15:1865-77. [PMID: 20874724 PMCID: PMC3918043 DOI: 10.1111/j.1582-4934.2010.01182.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cardiac hypertrophy in response to multiple stimuli has important physiological and pathological significances. GATA4 serves as a nuclear integrator of several signalling pathways during cardiac hypertrophy. Sp1 and Sp3 are also reported to be involved in this process. However, the mechanism by which GATA4 acts as a mediator, integrating these ubiquitously expressed transcriptional factors, is poorly understood. We found that the expression of GATA4 and Sp1 was up-regulated in the myocardium of a pressure overload hypertrophy rat model, as well in phenylephrine-induced (PE-induced) hypertrophic growth of neonatal cardiomyocytes. GST pull-down assays demonstrated that GATA4 could interact with Sp1 in vitro. Therefore, we proposed that GATA4 cooperates with Sp1 in regulating ANF expression, as its reactivation is closely linked with hypertrophy. Further studies demonstrated that GATA4 could activate the ANF promoter synergistically with Sp1 through direct interaction. In contrast, Sp3 exhibited antagonistic function, and overexpression of Sp3 repressed the transcriptional synergy between Sp1 and GATA4. We also found that Sp1 alone could activate the ANF promoter in cardiomyocytes, whereas Sp3 exerted negative effects on ANF expression. Bioinformatics analysis revealed novel Sp-binding sites on the ANF promoter. The recruitment of GATA4 and Sp1 on the ANF promoter was enhanced during phenylephrine-mediated hypertrophy, whereas the recruitment of Sp3 was reduced. The phosphorylation of GATA4 by ERK1/2 kinase could enhance the affinity between GATA4 and Sp1. Thus, our findings revealed the critical interaction of GATA4 and Sp1 in modulating ANF expression, indicating their involvement in cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoqing Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Suzuki YJ. Cell signaling pathways for the regulation of GATA4 transcription factor: Implications for cell growth and apoptosis. Cell Signal 2011; 23:1094-9. [PMID: 21376121 PMCID: PMC3078531 DOI: 10.1016/j.cellsig.2011.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 02/12/2011] [Accepted: 02/22/2011] [Indexed: 01/28/2023]
Abstract
GATA4 is a member of the GATA family of zinc finger transcription factor, which regulates gene transcription by binding to GATA elements. GATA4 was originally discovered as a regulator of cardiac development and subsequently identified as a major regulator of adult cardiac hypertrophy. GATA4 regulates gene expression of various genes, which are involved in cardiac development and cardiac hypertrophy and heart failure. In addition to the heart, GATA4 plays important roles in the reproductive system, gastrointestinal system, respiratory system and cancer. Positive and negative regulations of GATA4 therefore are important components of biologic functions. The activation of GATA4 occurs via various cell signaling events. Earlier studies have identified protein-protein interactions of GATA4 with other factors. The discovery of interactions of GATA4 with nuclear factor for activated T cells (NFAT) revealed the importance of calcium signaling in the activation of GATA4. GATA4 can also be phosphorylated by mitogen activated protein kinases and protein kinase A. Lysine modifications also occur on the GATA4 molecule including acetylation and sumoylation. Both reactive oxygen-dependent and -independent antioxidant-sensitive pathways for GATA4 activation have also been demonstrated. The GATA4 activity is also regulated by modulating the level of GATA4 expression via transcriptional as well as translational mechanisms. This work summarizes the current understanding of regulatory mechanisms for modulating GATA4 activity.
Collapse
Affiliation(s)
- Yuichiro J Suzuki
- Department of Pharmacology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA.
| |
Collapse
|
22
|
Barry SP, Townsend PA. What causes a broken heart--molecular insights into heart failure. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 284:113-79. [PMID: 20875630 DOI: 10.1016/s1937-6448(10)84003-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our understanding of the molecular processes which regulate cardiac function has grown immeasurably in recent years. Even with the advent of β-blockers, angiotensin inhibitors and calcium modulating agents, heart failure (HF) still remains a seriously debilitating and life-threatening condition. Here, we review the molecular changes which occur in the heart in response to increased load and the pathways which control cardiac hypertrophy, calcium homeostasis, and immune activation during HF. These can occur as a result of genetic mutation in the case of hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) or as a result of ischemic or hypertensive heart disease. In the majority of cases, calcineurin and CaMK respond to dysregulated calcium signaling and adrenergic drive is increased, each of which has a role to play in controlling blood pressure, heart rate, and left ventricular function. Many major pathways for pathological remodeling converge on a set of transcriptional regulators such as myocyte enhancer factor 2 (MEF2), nuclear factors of activated T cells (NFAT), and GATA4 and these are opposed by the action of the natriuretic peptides ANP and BNP. Epigenetic modification has emerged in recent years as a major influence cardiac physiology and histone acetyl transferases (HATs) and histone deacetylases (HDACs) are now known to both induce and antagonize hypertrophic growth. The newly emerging roles of microRNAs in regulating left ventricular dysfunction and fibrosis also has great potential for novel therapeutic intervention. Finally, we discuss the role of the immune system in mediating left ventricular dysfunction and fibrosis and ways this can be targeted in the setting of viral myocarditis.
Collapse
Affiliation(s)
- Seán P Barry
- Institute of Molecular Medicine, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | | |
Collapse
|
23
|
Synnergren J, Améen C, Lindahl A, Olsson B, Sartipy P. Expression of microRNAs and their target mRNAs in human stem cell-derived cardiomyocyte clusters and in heart tissue. Physiol Genomics 2010; 43:581-94. [PMID: 20841501 DOI: 10.1152/physiolgenomics.00074.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent studies have shown that microRNAs (miRNAs) act as posttranscriptional regulators and that they play important roles during heart development and in cardiac function. Thus, they may provide new means of altering stem cell fate and differentiation processes. However, information about the correlation between global miRNA and mRNA expression in cardiomyocyte clusters (CMCs) derived from human embryonic stem cells (hESC) and in fetal and adult heart tissue is lacking. In the present study the global miRNA and mRNA expression in hESC-derived CMCs and in fetal and adult heart tissue was investigated in parallel using microarrays. Target genes for the differentially expressed miRNAs were predicted using computational methods, and the concordance in miRNA expression and mRNA levels of potential target genes was determined across the experimental samples. The biology of the predicted target genes was further explored regarding their molecular functions and involvement in known regulatory pathways. A clear correlation between the global miRNA expression and corresponding target mRNA expression was observed. Using three different sources of cardiac tissue-like samples, we defined the similarities between in vitro hESC-derived CMCs and their in vivo counterparts. The results are in line with previously reported observations that miRNAs repress mRNA expression and additionally identify a number of novel miRNAs with potential important roles in human cardiac tissue. The concordant miRNA expression pattern observed among all the cardiac tissue-like samples analyzed here provide a starting point for future ambitious studies aiming towards assessment of the functional roles of specific miRNAs during cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Jane Synnergren
- School of Life Sciences, University of Skövde, Skövde, Sweden.
| | | | | | | | | |
Collapse
|
24
|
Lipskaia L, Ly H, Kawase Y, Hajjar RJ, Lompre AM. Treatment of heart failure by calcium cycling gene therapy. Future Cardiol 2010; 3:413-23. [PMID: 19804232 DOI: 10.2217/14796678.3.4.413] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Heart failure is a major cause of morbidity and mortality in Western countries. While progress in conventional treatment modalities is making steady and incremental gains to reduce this disease burden, there remains a need to explore new and potentially therapeutic approaches. Gene therapy, for example, was initially envisioned as a treatment strategy for inherited monogenic disorders. It is now apparent that gene therapy has broader potential, which also includes acquired polygenic diseases such as heart failure. Advances in the understanding of the molecular basis of conditions such as these, together with the evolution of increasingly efficient gene transfer technology, has placed congestive heart failure within the reach of gene-based therapy.
Collapse
Affiliation(s)
- Larissa Lipskaia
- INSERM U621, Université Pierre et Marie Curie-CHU Pitié-Salpétriêre, Paris, France
| | | | | | | | | |
Collapse
|
25
|
Sood AK, Armaiz-Pena GN, Halder J, Nick AM, Stone RL, Hu W, Carroll AR, Spannuth WA, Deavers MT, Allen JK, Han LY, Kamat AA, Shahzad MMK, McIntyre BW, Diaz-Montero CM, Jennings NB, Lin YG, Merritt WM, DeGeest K, Vivas-Mejia PE, Lopez-Berestein G, Schaller MD, Cole SW, Lutgendorf SK. Adrenergic modulation of focal adhesion kinase protects human ovarian cancer cells from anoikis. J Clin Invest 2010; 120:1515-23. [PMID: 20389021 DOI: 10.1172/jci40802] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 02/03/2010] [Indexed: 12/11/2022] Open
Abstract
Chronic stress is associated with hormonal changes that are known to affect multiple systems, including the immune and endocrine systems, but the effects of stress on cancer growth and progression are not fully understood. Here, we demonstrate that human ovarian cancer cells exposed to either norepinephrine or epinephrine exhibit lower levels of anoikis, the process by which cells enter apoptosis when separated from ECM and neighboring cells. In an orthotopic mouse model of human ovarian cancer, restraint stress and the associated increases in norepinephrine and epinephrine protected the tumor cells from anoikis and promoted their growth by activating focal adhesion kinase (FAK). These effects involved phosphorylation of FAKY397, which was itself associated with actin-dependent Src interaction with membrane-associated FAK. Importantly, in human ovarian cancer patients, behavioral states related to greater adrenergic activity were associated with higher levels of pFAKY397, which was in turn linked to substantially accelerated mortality. These data suggest that FAK modulation by stress hormones, especially norepinephrine and epinephrine, can contribute to tumor progression in patients with ovarian cancer and may point to potential new therapeutic targets for cancer management.
Collapse
Affiliation(s)
- Anil K Sood
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Xu M, Ji H, Dai DZ, Tang XY, Dai Y. Protective effect of the endothelin antagonist CPU0213 against isoprenaline-induced heart failure by suppressing abnormal expression of leptin, calcineurin and SERCA2a in rats. J Pharm Pharmacol 2010; 60:739-45. [DOI: 10.1211/jpp.60.6.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Abstract
Heart failure (HF) may be produced by sustained β-adrenoceptor stimulation by causing changes in the expression of endothelin-1 (ET-1), the leptin system, calcineurin and sarcoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) underlying cardiac dysfunction. The aim of this study was to verify whether isoprenaline (ISO)-induced HF is attributed to changes in the above molecular markers, and whether the dual ET-receptor antagonist CPU0213 could reverse the cardiac dysfunction caused by ISO treatment, focusing on these molecular markers. HF was induced in rats by administration of ISO (2 mgkg−1 s.c.) for 10 days. CPU0213 (30 mgkg−1 s.c.) and propranolol (4 mgkg−1 s.c.) were administered on days 7–10. HF developed after 10 days' ISO administration and was manifest as impaired cardiac performance, increased heart weight index, oxidative stress, elevated serum enzymes, and disordered expression of the endothelin system, leptin system, calcineurin and SERCA2a. All these abnormalities were significantly reversed by CPU0213, and the effectiveness of this ET-receptor antagonist was comparable to that of propranolol. Thus, antagonism of ET receptors by CPU0213 normalizes these changes in molecular markers, alleviating HF.
Collapse
Affiliation(s)
- Ming Xu
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Hui Ji
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - De-Zai Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xiao-Yun Tang
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yin Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
27
|
Yoshida Y, Morimoto T, Takaya T, Kawamura T, Sunagawa Y, Wada H, Fujita M, Shimatsu A, Kita T, Hasegawa K. Aldosterone Signaling Associates With p300/GATA4 Transcriptional Pathway During the Hypertrophic Response of Cardiomyocytes. Circ J 2010; 74:156-62. [DOI: 10.1253/circj.cj-09-0050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yoshinori Yoshida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Tatsuya Morimoto
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Tomohide Takaya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Teruhisa Kawamura
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Yoichi Sunagawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Hiromichi Wada
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| | - Masatoshi Fujita
- Human Health Sciences, Graduate School of Medicine, Kyoto University
| | - Akira Shimatsu
- Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Toru Kita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University
| | - Koji Hasegawa
- Division of Translational Research, Kyoto Medical Center, National Hospital Organization
| |
Collapse
|
28
|
Tanaka H. [Cooperative research and educational center for breeding and standardization of medicinal plants and drug development between Japan and China]. YAKUGAKU ZASSHI 2009; 129:393-400. [PMID: 19336992 DOI: 10.1248/yakushi.129.393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A project "Cooperative research and educational center for breeding and standardization of medicinal plants and drug development between Japan and China" was launched in 2006 with the support of National Natural Science Foundation of China (NSFC) and Japan Society for the Promotion of Science (JSPS). The project focuses on medicinal plants that are important sources of traditional medicines and modern drugs. Many of these plants exist in Asian countries and have been utilized in health care to maintain quality of life. Recently, the short supply of some important medicinal plants, like Glycyrrhiza sp. and Ephedra sp. has become an issue in Japan and China. We believe that cooperative research by many specialist researchers is a prerequisite for overcoming this problem and for establishing a supply of these resources with their highly added values. Furthermore, with our group's participating researchers utilizing state-of-the-art drug evaluation methods in pharmacological and pharmacutical sciences, the development of new drugs and treatment candidates using the collaborative resources of this project, which include distinguished natural chemists, is anticipated. Another important aim of the project is to foster young scientists. In the project, we are encouraging them to continue their studies to produce new findings regarding traditional medicines and drug development.
Collapse
Affiliation(s)
- Hiroyuki Tanaka
- Graduate School of Pharmaceutical Sciences, Kyushu University, Japan.
| |
Collapse
|
29
|
Lipopolysaccharide induces cellular hypertrophy through calcineurin/NFAT-3 signaling pathway in H9c2 myocardiac cells. Mol Cell Biochem 2008; 313:167-78. [PMID: 18398669 DOI: 10.1007/s11010-008-9754-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 03/28/2008] [Indexed: 01/19/2023]
Abstract
Evidences suggest that lipopolysaccharide (LPS) participates in the inflammatory response in the cardiovascular system; however, it is unknown if LPS is sufficient to cause the cardiac hypertrophy. In the present study, we treated H9c2 myocardiac cells with LPS to explore whether LPS causes cardiac hypertrophy, and to identify the precise molecular and cellular mechanisms behind hypertrophic responses. Here we show that LPS challenge induces pathological hypertrophic responses such as the increase in cell size, the reorganization of actin filaments, and the upregulation of hypertrophy markers including atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) in H9c2 cells. LPS treatment significantly promotes the activation of GATA-4 and the nuclear translocation of NFAT-3, which act as transcription factors mediating the development of cardiac hypertrophy. After administration of inhibitors including U0126 (ERK1/2 inhibitor), SB203580 (p38 MAPK inhibitor), SP600125 (JNK1/2 inhibitor), CsA (calcineurin inhibitor), FK506 (calcineurin inhibitor), and QNZ (NFkappaB inhibitor), LPS-induced hypertrophic characteristic features, such as increases in cell size, actin fibers, and levels of ANP and BNP, and the nuclear localization of NFAT-3 are markedly inhibited only by calcineurin inhibitors, CsA and FK506. Collectively, these results suggest that LPS leads to myocardiac hypertrophy through calcineurin/NFAT-3 signaling pathway in H9c2 cells. Our findings further provide a link between the LPS-induced inflammatory response and the calcineurin/NFAT-3 signaling pathway that mediates the development of cardiac hypertrophy.
Collapse
|
30
|
Diedrichs H, Hagemeister J, Chi M, Boelck B, Müller-Ehmsen J, Schneider CA. Activation of the calcineurin/NFAT signalling cascade starts early in human hypertrophic myocardium. J Int Med Res 2008; 35:803-18. [PMID: 18034994 DOI: 10.1177/147323000703500609] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cardiac hypertrophy is an independent risk factor for heart failure. Recent studies on gene regulation of proteins have involved intracellular Ca2+ homeostasis. The Ca2+-sensitive phosphatase, calcineurin, is one potential regulator of the hypertrophic response, so we aimed to investigate the calcineurin-dependent signal pathway at different stages of hypertrophy in human myocardium. We found the calcineurin pathway to be significantly activated in hypertrophic compared with non-hypertrophic myocardium as demonstrated by increased calcineurin activity and expression of calcineurin A-beta and B, and GATA-4, and a shift of phosphorylated cytoplasmic NFAT-3 into the nucleus as dephosphorylated nuclear NFAT-3. There was a tendency for these changes to be more pronounced in the decompensated compared with the compensated hypertrophic myocardium. The present study provides evidence for significant activation of the Ca2+-triggered calcineurin pathway in hypertrophic humans. Already present in compensated hypertrophy it showed a tendency to a further increase following transition to decompensated hypertrophy.
Collapse
Affiliation(s)
- H Diedrichs
- Laboratory of Muscle Research and Molecular Cardiology, Department of Internal Medicine, University of Cologne, Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Yiu GK, Toker A. NFAT induces breast cancer cell invasion by promoting the induction of cyclooxygenase-2. J Biol Chem 2006; 281:12210-7. [PMID: 16505480 DOI: 10.1074/jbc.m600184200] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors plays a fundamental role in the transcriptional regulation of the immune response. However, NFATs are ubiquitously expressed, and recent evidence points to their important functions in human epithelial cells and carcinomas. Specifically, NFAT has been shown to be active in human breast and colon carcinoma cells and to promote their invasion through Matrigel. The mechanisms by which NFAT promotes invasion have not been defined. To identify NFAT target genes that induce carcinoma invasion, we have established stable breast cancer cell lines that inducibly express transcriptionally active NFAT. Gene expression profiling by cDNA microarray of cells induced to express NFAT revealed up-regulation of cyclooxygenase-2 (COX-2). Increased NFAT expression and activity induced COX-2 expression as well as prostaglandin E2 synthesis. This induction was more prominent when NFAT was activated by phorbol 12-myristate 13-acetate and calcium ionophore ionomycin and was blocked by the NFAT antagonist cyclosporin A. Breast cancer cells with elevated COX-2 expression showed increased invasion through Matrigel, and this was reduced in cells treated with COX-2 inhibitors. Conversely, loss of NFAT1 protein expression using small interfering RNA led to a reduction in COX-2 transcription and reduced invasion. Similarly, Matrigel invasion was reduced in cells in which COX-2 expression was reduced using specific siRNA. These findings demonstrate that NFAT promotes breast cancer cell invasion through the induction of COX-2 and the synthesis of prostaglandins.
Collapse
Affiliation(s)
- Gary K Yiu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
33
|
Zhu Z, Zhu S, Liu D, Yu Z, Yang Y, van der Giet M, Tepel M. GATA4-mediated cardiac hypertrophy induced by d-myo-inositol 1,4,5-tris-phosphate. Biochem Biophys Res Commun 2005; 338:1236-40. [PMID: 16259952 DOI: 10.1016/j.bbrc.2005.10.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Accepted: 10/17/2005] [Indexed: 11/16/2022]
Abstract
We evaluated the effects of d-myo-inositol 1,4,5-tris-phosphate on cardiac hypertrophy. d-myo-inositol 1,4,5-tris-phosphate augmented cardiac hypertrophy as evidenced by its effects on DNA synthesis, protein synthesis, and expression of immediate-early genes c-myc and c-fos, beta-myosin heavy chain, and alpha-actin. The administration of d-myo-inositol 1,4,5-tris-phosphate increased the expression of nuclear factor of activated T-cells and cardiac-restricted zinc finger transcription factor (GATA4). Real-time quantitative RT-PCR showed that d-myo-inositol 1,4,5-tris-phosphate-induced GATA4 mRNA was significantly enhanced even in the presence of the calcineurin inhibitor, cyclosporine A. The effect of d-myo-inositol 1,4,5-tris-phosphate was blocked after inhibition of inositol-trisphosphate receptors but not after inhibition of c-Raf/mitogen-activated protein kinase kinase (MEK)/mitogen-activated protein kinase (ERK) or p38 mitogen-activated protein kinase pathways. The study shows that d-myo-inositol 1,4,5-tris-phosphate-induced cardiac hypertrophy is mediated by GATA4 but independent from the calcineurin pathway.
Collapse
Affiliation(s)
- Zhiming Zhu
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing Hypertension Institute, Chongqing, PR China.
| | | | | | | | | | | | | |
Collapse
|
34
|
Winitsky SO, Gopal TV, Hassanzadeh S, Takahashi H, Gryder D, Rogawski MA, Takeda K, Yu ZX, Xu YH, Epstein ND. Adult murine skeletal muscle contains cells that can differentiate into beating cardiomyocytes in vitro. PLoS Biol 2005; 3:e87. [PMID: 15757365 PMCID: PMC1064849 DOI: 10.1371/journal.pbio.0030087] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 01/06/2005] [Indexed: 12/30/2022] Open
Abstract
It has long been held as scientific fact that soon after birth, cardiomyocytes cease dividing, thus explaining the limited restoration of cardiac function after a heart attack. Recent demonstrations of cardiac myocyte differentiation observed in vitro or after in vivo transplantation of adult stem cells from blood, fat, skeletal muscle, or heart have challenged this view. Analysis of these studies has been complicated by the large disparity in the magnitude of effects seen by different groups and obscured by the recently appreciated process of in vivo stem-cell fusion. We now show a novel population of nonsatellite cells in adult murine skeletal muscle that progress under standard primary cell-culture conditions to autonomously beating cardiomyocytes. Their differentiation into beating cardiomyocytes is characterized here by video microscopy, confocal-detected calcium transients, electron microscopy, immunofluorescent cardiac-specific markers, and single-cell patch recordings of cardiac action potentials. Within 2 d after tail-vein injection of these marked cells into a mouse model of acute infarction, the marked cells are visible in the heart. By 6 d they begin to differentiate without fusing to recipient cardiac cells. Three months later, the tagged cells are visible as striated heart muscle restricted to the region of the cardiac infarct. A population of primitive cells from adult murine skeletal muscle can develop into beating cardiomyocytes in vitro and can contribute to the repair of damaged heart in vivo
Collapse
Affiliation(s)
- Steve O Winitsky
- 1Molecular Physiology Section, Laboratory of Molecular CardiologyNational Heart, Lung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Thiru V Gopal
- 1Molecular Physiology Section, Laboratory of Molecular CardiologyNational Heart, Lung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Shahin Hassanzadeh
- 1Molecular Physiology Section, Laboratory of Molecular CardiologyNational Heart, Lung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Hiroshi Takahashi
- 1Molecular Physiology Section, Laboratory of Molecular CardiologyNational Heart, Lung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Divina Gryder
- 2Epilepsy Research Section, National Institute of Neurological Disorders and StrokeBethesda, MarylandUnited States of America
| | - Michael A Rogawski
- 2Epilepsy Research Section, National Institute of Neurological Disorders and StrokeBethesda, MarylandUnited States of America
| | - Kazuyo Takeda
- 3Laboratory of Molecular Cardiology, National HeartLung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Zu X Yu
- 4Pathology Section, National HeartLung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Yu H Xu
- 5Electron Microscopy Core Facility, National HeartLung, and Blood Institute, Bethesda, MarylandUnited States of America
| | - Neal D Epstein
- 1Molecular Physiology Section, Laboratory of Molecular CardiologyNational Heart, Lung, and Blood Institute, Bethesda, MarylandUnited States of America
| |
Collapse
|
35
|
Sanna B, Bueno OF, Dai YS, Wilkins BJ, Molkentin JD. Direct and indirect interactions between calcineurin-NFAT and MEK1-extracellular signal-regulated kinase 1/2 signaling pathways regulate cardiac gene expression and cellular growth. Mol Cell Biol 2005; 25:865-78. [PMID: 15657416 PMCID: PMC544001 DOI: 10.1128/mcb.25.3.865-878.2005] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MEK1, a member of the mitogen-activated protein kinase (MAPK) cascade that directly activates extracellular signal-regulated kinase (ERK), induces cardiac hypertrophy in transgenic mice. Calcineurin is a calcium-regulated protein phosphatase that also functions as a positive regulator of cardiac hypertrophic growth through a direct mechanism involving activation of nuclear factor of activated T-cell (NFAT) transcription factors. Here we determined that calcineurin-NFAT and MEK1-ERK1/2 signaling pathways are interdependent in cardiomyocytes, where they directly coregulate the hypertrophic growth response. For example, genetic deletion of the calcineurin Abeta gene reduced the hypertrophic response elicited by an activated MEK1 transgene in the heart, while inhibition of calcineurin or NFAT in cultured neonatal cardiomyocytes also blunted the hypertrophic response driven by activated MEK1. Conversely, targeted inhibition of MEK1-ERK1/2 signaling in cultured cardiomyocytes attenuated the hypertrophic growth response directed by activated calcineurin. However, targeted inhibition of MEK1-ERK1/2 signaling did not directly affect calcineurin-NFAT activation, nor was MEK1-ERK1/2 activation altered by targeted inhibition of calcineurin-NFAT. Mechanistically, we show that MEK1-ERK1/2 signaling augments NFAT transcriptional activity independent of calcineurin, independent of changes in NFAT nuclear localization, and independent of alterations in NFAT transactivation potential. In contrast, MEK1-ERK1/2 signaling enhances NFAT-dependent gene expression through an indirect mechanism involving induction of cardiac AP-1 activity, which functions as a necessary NFAT-interacting partner. As a second mechanism, MEK1-ERK1/2 and calcineurin-NFAT proteins form a complex in cardiac myocytes, resulting in direct phosphorylation of NFATc3 within its C terminus. MEK1-ERK1/2-mediated phosphorylation of NFATc3 directly augmented its DNA binding activity, while inhibition of MEK1-ERK1/2 signaling reduced NFATc3 DNA binding activity. Collectively, these results indicate that calcineurin-NFAT and MEK1-ERK1/2 pathways constitute a codependent signaling module in cardiomyocytes that coordinately regulates the growth response through two distinct mechanisms.
Collapse
Affiliation(s)
- Bastiano Sanna
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC7020, Cincinnati, OH 45229-3039, USA
| | | | | | | | | |
Collapse
|
36
|
Monticelli S, Solymar DC, Rao A. Role of NFAT proteins in IL13 gene transcription in mast cells. J Biol Chem 2004; 279:36210-8. [PMID: 15229217 DOI: 10.1074/jbc.m406354200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Th2 and mast cells are participants in the asthmatic response to allergens, and both cell types produce the cytokines interleukin (IL)-4 and IL-13. IL-13 in particular is both necessary and sufficient for experimental models of asthma. The transcription factor NFAT plays a central role in cytokine transcriptional regulation in both cell types. Here, we analyze the molecular basis of IL13 gene transcription in Th2 and mast cells. We show that NFAT1 is the major NFAT protein involved in regulating IL13 transcription in mast cells. Although NFAT2 is correctly expressed and regulated in mast cells, it does not contribute to IL13 gene transcription as shown by analysis of cells lacking NFAT2 and cells expressing a constitutively active version of NFAT2. The difference between NFAT1 and NFAT2 appears to be due to a preferential synergistic interaction of NFAT1 with GATA proteins at the IL13 promoter. We suggest that mast cells lack a co-activator protein that stabilizes the binding of NFAT2 to the IL13 promoter by interacting either with NFAT2 itself or with a DNA-bound complex of NFAT2 and GATA proteins.
Collapse
Affiliation(s)
- Silvia Monticelli
- Department of Pathology, Harvard Medical School, and CBR Institute for Biomedical Research, Boston, MA 02115, USA
| | | | | |
Collapse
|
37
|
|
38
|
Aries A, Paradis P, Lefebvre C, Schwartz RJ, Nemer M. Essential role of GATA-4 in cell survival and drug-induced cardiotoxicity. Proc Natl Acad Sci U S A 2004; 101:6975-80. [PMID: 15100413 PMCID: PMC406451 DOI: 10.1073/pnas.0401833101] [Citation(s) in RCA: 221] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In recent years, significant progress has been made in understanding cardiomyocyte differentiation. However, little is known about the regulation of myocyte survival despite the fact that myocyte apoptosis is a leading cause of heart failure. Here we report that transcription factor GATA-4 is a survival factor for differentiated, postnatal cardiomyocytes and an upstream activator of the antiapoptotic gene Bcl-X. An early event in the cardiotoxic effect of the antitumor drug doxorubicin is GATA-4 depletion, which in turn causes cardiomyocyte apoptosis. Mouse heterozygotes for a null Gata4 allele have enhanced susceptibility to doxorubicin cardiotoxicity. Genetic or pharmacologic enhancement of GATA-4 prevents cardiomyocyte apoptosis and drug-induced cardiotoxicity. The results indicate that GATA-4 is an antiapoptotic factor required for the adaptive stress response of the adult heart. Modulation of survival/apoptosis genes by tissue-specific transcription factors may be a general paradigm that can be exploited effectively for cell-specific regulation of apoptosis in disease states.
Collapse
Affiliation(s)
- Anne Aries
- Laboratory of Cardiac Growth and Differentiation, Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada H2W 1R7
| | | | | | | | | |
Collapse
|
39
|
Abstract
The Ras subfamily of 21-kDa ("small") guanine nucleotide binding proteins [which includes Ha-Ras, Ki(A)-Ras, Ki(B)-Ras, and N-Ras] is universally important in regulating intracellular signaling events in mammalian cells and controls their growth, proliferation, senescence, differentiation, and survival. These Ras isoforms act as membrane-associated biological switches that transduce signals from transmembrane receptors, thus potentially activating a variety of downstream signaling proteins. These include ultimately two Ser/Thr protein kinase families, the extracellular signal-regulated kinases 1/2 (ERK1/2) and Akt (or protein kinase B). Activation of ERK1/2 has been associated with cardiac myocyte hypertrophy (ie, increased cell size and myofibrillogenesis, with concurrent transcriptional changes to a fetal pattern of gene expression), whereas activation of Akt is associated with the increased protein accretion in hypertrophy. Both ERK1/2 and Akt may promote myocyte survival. In the intact heart in vivo and in primary cultures of cardiac myocytes, mechanical strain induces hypertrophy, a process known as mechanotransduction, which may involve Ras, ERK1/2, and Akt. In this study, general and cardiospecific aspects of the regulation of Ras and Akt will be described. The various mechanisms through which mechanical strain might initiate Ras- or Akt-dependent signaling will be discussed. The overall conclusion is that although an involvement of Ras and Akt in mechanotransduction is likely, more work (particularly focusing on mechanoreception) needs to be undertaken before it is unequivocally established.
Collapse
Affiliation(s)
- Peter H Sugden
- National Heart and Lung Institute Division, Faculty of Medicine, Imperial College London, Flowers Building (4th Floor), Armstrong Road, London SW7 2AZ, UK.
| |
Collapse
|
40
|
Kedzierski RM, Grayburn PA, Kisanuki YY, Williams CS, Hammer RE, Richardson JA, Schneider MD, Yanagisawa M. Cardiomyocyte-specific endothelin A receptor knockout mice have normal cardiac function and an unaltered hypertrophic response to angiotensin II and isoproterenol. Mol Cell Biol 2003; 23:8226-32. [PMID: 14585980 PMCID: PMC262340 DOI: 10.1128/mcb.23.22.8226-8232.2003] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Even though endothelin is recognized as an important vasoregulatory molecule, the roles of endothelin receptors in specific cell types are not yet fully understood. Mice with a null mutation in endothelin A receptor gene (ET(A)) or in the gene of its ligand (endothelin 1) die neonatally due to craniofacial and cardiac abnormalities. This early lethality has in the past hindered studies on the role of endothelin in cardiovascular physiology and pathophysiology. To overcome this obstacle, we utilized the cre/loxP technology to generate mice in which the ET(A) gene could be deleted specifically in cardiomyocytes. The cre recombinase transgene driven by the alpha-myosin heavy-chain promoter deleted the floxed ET(A) allele specifically in the hearts of these mice, resulting in a 78% reduction in cardiac ET(A) mRNA level compared to wild-type controls. Cardiomyocyte-specific ET(A) knockout animals are viable and exhibit normal growth, cardiac anatomy, and cardiac contractility, as assessed by echocardiography. In addition, these animals exhibit hypertrophic and contractile responses to 10-day infusion of angiotensin II or isoproterenol similar to those observed in control animals. These results indicate that in adult mice cardiac ET(A) receptors are not necessary for either baseline cardiac function or stress-induced response to angiotensin II or isoproterenol.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Base Sequence
- Cardiomegaly/etiology
- DNA/genetics
- Female
- Heart/physiology
- Isoproterenol/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Endothelin A/deficiency
- Receptor, Endothelin A/genetics
- Receptor, Endothelin A/physiology
- Receptor, Endothelin B/genetics
- Receptor, Endothelin B/physiology
Collapse
Affiliation(s)
- Rafal M Kedzierski
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lakshmikuttyamma A, Selvakumar P, Kakkar R, Kanthan R, Wang R, Sharma RK. Activation of calcineurin expression in ischemia-reperfused rat heart and in human ischemic myocardium. J Cell Biochem 2003; 90:987-97. [PMID: 14624458 DOI: 10.1002/jcb.10722] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Calcineurin (CaN) has been reported as a critical mediator for cardiac hypertrophy and cardiac myocyte apoptosis. In the present study, we investigated the activity and expression of CaN and the effect of calpain in rat heart after ischemia and reperfusion. Rat ischemic heart showed significant increase in CaN activity. Western blot analysis of normal rat heart extract with a polyclonal antibody raised against bovine CaN indicated a prominent immunoreactive band of 60 kDa (CaN A). In ischemic-reperfused hearts, the expression of CaN A was significantly low and immunoreactivity was observed in proteolytic bands of 46 kDa. This may be due to the proteolytic degradation of CaN A in ischemic tissues by m-calpain. We also noticed in vitro proteolysis of bovine cardiac CaN A by m-calpain. Immunohistochemical studies showed strong staining of immunoreactivity in rat hearts that had gone under 30 min ischemia followed by 30 min reperfusion similar to that found in human ischemic heart. Ischemia is associated with multiple alterations in the extracellular and intracellular signaling of cardiomyocytes and may act as an inducer of apoptosis. The increase in CaN activity and strong immunostaining observed in ischemic/perfused rat heart may be due to the calpain-mediated proteolysis of this phosphatase.
Collapse
Affiliation(s)
- Ashakumary Lakshmikuttyamma
- Department of Pathology, College of Medicine and Cancer Research Unit, Health Research Division, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, Canada S7N 4H4
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Mesangial cells (MCs) play a central role in the physiology and pathophysiology of endothelin-1 (ET-1) in the kidney. MCs release ET-1 in response to a variety of factors, many of which are elevated in glomerular injury. MCs also express ET receptors, activation of which leads to a complex signaling cascade with resultant stimulation of MC hypertrophy, proliferation, contraction, and extracellular matrix accumulation. MC ET-1 interacts with other important regulatory factors, including arachidonate metabolites, nitric oxide, and angiotensin II. Excessive stimulation of ET-1 production by, and activity in, MC is likely of pathogenic importance in glomerular damage in the setting of diabetes, hypertension, and glomerulonephritis. The recent introduction of ET antagonists, and possibly ET-converting enzyme inhibitors, into the clinical arena establishes the potential for new therapies for those diseases characterized by increased MC ET-1 actions. This review will examine our present understanding of how ET-1 is involved in mesangial function in health and disease. In addition, we will discuss the status of clinical trials using ET antagonists, which have only been conducted in nonrenal disease, as a background for advocating their use in diseases characterized by excessive MC-derived ET-1.
Collapse
Affiliation(s)
- Andrey Sorokin
- Division of Nephrology, Medical College of Wisconsin, Milwaukee 53226, USA
| | | |
Collapse
|
43
|
Hogan PG, Chen L, Nardone J, Rao A. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev 2003; 17:2205-32. [PMID: 12975316 DOI: 10.1101/gad.1102703] [Citation(s) in RCA: 1546] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Patrick G Hogan
- The Center for Blood Research, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
44
|
Pikkarainen S, Tokola H, Majalahti-Palviainen T, Kerkela R, Hautala N, Bhalla SS, Charron F, Nemer M, Vuolteenaho O, Ruskoaho H. GATA-4 is a nuclear mediator of mechanical stretch-activated hypertrophic program. J Biol Chem 2003; 278:23807-16. [PMID: 12704188 DOI: 10.1074/jbc.m302719200] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In overloaded heart the cardiomyocytes adapt to increased mechanical and neurohumoral stress by activation of hypertrophic program, resulting in morphological changes of individual cells and specific changes in gene expression. Accumulating evidence suggests an important role for the zinc finger transcription factor GATA-4 in hypertrophic agonist-induced cardiac hypertrophy. However, its role in stretch-induced cardiomyocyte hypertrophy is not known. We employed an in vitro mechanical stretch model of cultured cardiomyocytes and used rat B-type natriuretic peptide promoter as stretch-sensitive reporter gene. Stretch transiently increased GATA-4 DNA binding activity and transcript levels, which was followed by increases in the expression of B-type natriuretic peptide as well as atrial natriuretic peptide and skeletal alpha-actin genes. The stretch inducibility mapped primarily to the proximal 520 bp of the B-type natriuretic peptide promoter. Mutational studies showed that the tandem GATA consensus sites of the proximal promoter in combination with an Nkx-2.5 binding element are critical for stretch-activated B-type natriuretic peptide transcription. Inhibition of GATA-4 protein production by adenovirus-mediated transfer of GATA-4 antisense cDNA blocked stretch-induced increases in B-type natriuretic peptide transcript levels and the sarcomere reorganization. The proportion of myocytes with assembled sarcomeres in control adenovirus-infected cultures increased from 14 to 59% in response to stretch, whereas the values for GATA-4 antisense-treated cells were 6 and 13%, respectively. These results show that activation of GATA-4, in cooperation with a factor binding on Nkx-2.5 binding element, is essential for mechanical stretch-induced cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Sampsa Pikkarainen
- Department of Pharmacology, Biocenter Oulu, University of Oulu, PO Box 5000, FIN-90014 University of Oulu, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Porter GA, Makuck RF, Rivkees SA. Intracellular calcium plays an essential role in cardiac development. Dev Dyn 2003; 227:280-90. [PMID: 12761855 DOI: 10.1002/dvdy.10307] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intracellular calcium signaling plays an essential role in cardiac physiology and modulates cardiac gene expression. However, the role that intracellular calcium signaling plays during cardiac development is not known. To address this issue, we examined the effects of altered intracellular calcium levels on cardiac morphogenesis. In acutely cultured mouse embryos, L-type calcium channel blockade decreased resting intracellular calcium levels and inhibited calcium transients. Embryos cultured at embryonic day (E) 7.5-8.5 in the presence of the L-type calcium channel blockers nifedipine and verapamil developed hearts that had a large left ventricle, lacked a right ventricle and had a long, thin outflow tract. If embryos were cultured at E7.5, calcium channel blockade also induced an abnormal, anterior cardiac loop. These alterations in development were not due to altered cardiac function, as heart rates at the end of the culture period were not affected by calcium channel blockade and blood flow was observed. Treatment with nifedipine altered the mRNA expression of the transcription factor Gata4, which was absent in the developing ventricles, and the sarcomeric protein Mylpc (myosin light chain 2V), which was decreased distal to the left ventricle and was absent at the site of the developing right ventricle. In contrast, the expression pattern of other cardiac transcription factor (Hand1, Hand2, Mef2c, Nkx2-5) and cytoskeletal protein (Myhca, Tagln) mRNA did not change with calcium channel blockade. These data demonstrate that proper intracellular calcium signaling is essential for normal cardiac looping, gene expression, and organ development.
Collapse
Affiliation(s)
- George A Porter
- Yale Child Health Research Center and Department of Pediatrics, Division of Cardiology, Yale University School of Medicine, New Haven, Connecticut 06520-8064, USA.
| | | | | |
Collapse
|
46
|
Abstract
Different cell types, equipped with unique structure and function, synthesize different sets of proteins on the basis of different patterns of gene expression, even though their genomes are identical. Cardiac transcription factors have been reported to control a cardiac gene program and thus to play a crucial role in transcriptional regulation during embryogenesis. Recently, postnatal roles of cardiac transcription factors have been extensively investigated. Consistent with the direct transactivation of numerous cardiac genes reactivated in response to hypertrophic stimulation, cardiac transcription factors are profoundly involved in the generation of cardiac hypertrophy or in cardioprotection from cytotoxic stress in the adult heart. In this review, the regulation of a cardiac gene program by cardiac transcription factors is summarized, with an emphasis on their potential role in the generation of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hiroshi Akazawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | |
Collapse
|
47
|
Abstract
Over the past year, vertebrate GATA factors have been found to participate directly in several signal-transduction pathways. Smad3, phosphorylated by TGF-beta signalling, interacts with GATA3 to induce differentiation of T helper cells. Hypertrophic stimuli act through RhoA GTPase and ROCK kinase to activate GATA4 in cardiac myocytes. In the liver, GATA4 is elevated by BMP and FGF signalling, and is able to bind to chromatin targets. Invertebrate GATA factors play a central role in specifying the mesendoderm.
Collapse
Affiliation(s)
- Roger K Patient
- Institute of Genetics, University of Nottingham, Queen's Medical Centre, UK
| | | |
Collapse
|
48
|
Abstract
Brain natriuretic peptide (BNP) is a cardiac hormone constitutively expressed in the adult heart. We previously showed that the human BNP (hBNP) proximal promoter region from -127 to -40 confers myocyte-specific expression. The proximal hBNP promoter contains several putative cis elements. Here we tested whether the proximal GATA element plays a role in basal and inducible regulation of the hBNP promoter. The hBNP promoter was coupled to a luciferase reporter gene (1818hBNPLuc) and transferred into neonatal ventricular myocytes (NVM), and luciferase activity was measured as an index of hBNP promoter activity. Mutation of the putative GATA element at -85 of the hBNP promoter [1818(mGATA)hBNPLuc] reduced activity by 97%. To study transactivation of the hBNP promoter, we co-transfected 1818hBNPLuc with the GATA-4 expression vector. GATA-4 activated 1818hBNPLuc, and this effect was eliminated by mutation of the proximal GATA element. Electrophoretic mobility shift assay showed that an oligonucleotide containing the hBNP GATA motif bound to cardiomyocyte nuclear protein, which was competed for by a consensus GATA oligonucleotide but not a mutated hBNP GATA element. The beta-adrenergic agonist isoproterenol and its second messenger cAMP stimulated hBNP promoter activity and binding of nuclear protein to the proximal GATA element. Thus the GATA element in the proximal hBNP promoter is involved in both basal and inducible transcriptional regulation in cardiac myocytes.
Collapse
Affiliation(s)
- Quan He
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
49
|
Liang Q, Molkentin JD. Divergent signaling pathways converge on GATA4 to regulate cardiac hypertrophic gene expression. J Mol Cell Cardiol 2002; 34:611-6. [PMID: 12054848 DOI: 10.1006/jmcc.2002.2011] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
50
|
Abstract
The heart is a dynamic organ capable of adapting its size and architecture in response to alterations in workload associated with developmental maturation, physiological stimulation and pathological diseases. Such alterations in heart size typically result from the hypertrophic growth of individual myocytes, but not myocyte cellular proliferation. In recent years, a great deal of investigation has gone toward elucidating the molecular signalling machinery that underlies the hypertrophic response and manner in which increased cardiac load promotes alterations in gene expression. To this end, the Ca(2+)-calmodulin-activated phosphatase calcineurin has been proposed as a necessary component of the multi-pathway hypertrophy program in the heart. Despite initial controversy over this hypothesis due to disparate results from pharmacological inhibitory studies in animal models of hypertrophy, compelling data from genetic models with calcineurin inhibition now exist. This review will summarize many of these studies and will attempt to address a number of unanswered issues. In particular, specific downstream mediators of calcineurin signalling will be discussed, as well as the need to identify calcineurin's temporal activation profile, transcriptional targets and cross-communication with other reactive signalling pathways in the heart. Finally, we will present evidence suggesting that calcineurin, as a Ca(2+)-responsive enzyme, may function as an internal load sensor in cardiac myocytes, matching output demands to hypertrophic growth.
Collapse
Affiliation(s)
- Benjamin J Wilkins
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, Children's Hospital Medical Center, Cincinnati, OH, USA
| | | |
Collapse
|