1
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Nie S. Use of Frogs as a Model to Study the Etiology of HLHS. J Cardiovasc Dev Dis 2023; 10:51. [PMID: 36826547 PMCID: PMC9965361 DOI: 10.3390/jcdd10020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A frog is a classical model organism used to uncover processes and regulations of early vertebrate development, including heart development. Recently, we showed that a frog also represents a useful model to study a rare human congenital heart disease, hypoplastic left heart syndrome. In this review, we first summarized the cellular events and molecular regulations of vertebrate heart development, and the benefit of using a frog model to study congenital heart diseases. Next, we described the challenges in elucidating the etiology of hypoplastic left heart syndrome and discussed how a frog model may contribute to our understanding of the molecular and cellular bases of the disease. We concluded that a frog model offers its unique advantage in uncovering the cellular mechanisms of hypoplastic left heart syndrome; however, combining multiple model organisms, including frogs, is needed to gain a comprehensive understanding of the disease.
Collapse
Affiliation(s)
- Shuyi Nie
- School of Biological Sciences, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
3
|
Afouda BA. Towards Understanding the Gene-Specific Roles of GATA Factors in Heart Development: Does GATA4 Lead the Way? Int J Mol Sci 2022; 23:5255. [PMID: 35563646 PMCID: PMC9099915 DOI: 10.3390/ijms23095255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023] Open
Abstract
Transcription factors play crucial roles in the regulation of heart induction, formation, growth and morphogenesis. Zinc finger GATA transcription factors are among the critical regulators of these processes. GATA4, 5 and 6 genes are expressed in a partially overlapping manner in developing hearts, and GATA4 and 6 continue their expression in adult cardiac myocytes. Using different experimental models, GATA4, 5 and 6 were shown to work together not only to ensure specification of cardiac cells but also during subsequent heart development. The complex involvement of these related gene family members in those processes is demonstrated through the redundancy among them and crossregulation of each other. Our recent identification at the genome-wide level of genes specifically regulated by each of the three family members and our earlier discovery that gata4 and gata6 function upstream, while gata5 functions downstream of noncanonical Wnt signalling during cardiac differentiation, clearly demonstrate the functional differences among the cardiogenic GATA factors. Such suspected functional differences are worth exploring more widely. It appears that in the past few years, significant advances have indeed been made in providing a deeper understanding of the mechanisms by which each of these molecules function during heart development. In this review, I will therefore discuss current evidence of the role of individual cardiogenic GATA factors in the process of heart development and emphasize the emerging central role of GATA4.
Collapse
Affiliation(s)
- Boni A Afouda
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| |
Collapse
|
4
|
Zhang C, Zhao H, Song X, Wang J, Zhao S, Deng H, He L, Zhou X, Yin X, Zhang K, Zhang Y, Wu Z, Chen Q, Du J, Yu D, Zhang S, Deng W. Transcription factor GATA4 drives RNA polymerase III-directed transcription and transformed cell proliferation through a filamin A/GATA4/SP1 pathway. J Biol Chem 2022; 298:101581. [PMID: 35038452 PMCID: PMC8857480 DOI: 10.1016/j.jbc.2022.101581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 11/27/2022] Open
Abstract
RNA polymerase III (pol III) products play fundamental roles in a variety of cellular processes, including protein synthesis and cancer cell proliferation. In addition, dysregulation of pol III-directed transcription closely correlates with tumorigenesis. It is therefore of interest to identify novel pathways or factors governing pol III-directed transcription. Here, we show that transcription factor (TF) GATA binding protein 4 (GATA4) expression in SaOS2 cells was stimulated by the silencing of filamin A (FLNA), a repressor of pol III-directed transcription, suggesting that GATA4 is potentially associated with the regulation of pol III-directed transcription. Indeed, we show that GATA4 expression positively correlates with pol III-mediated transcription and tumor cell proliferation. Mechanistically, we found that GATA4 depletion inhibits the occupancies of the pol III transcription machinery factors at the loci of pol III target genes by reducing expression of both TFIIIB subunit TFIIB-related factor 1 and TFIIIC subunit general transcription factor 3C subunit 2 (GTF3C2). GATA4 has been shown to activate specificity factor 1 (Sp1) gene transcription by binding to the Sp1 gene promoter, and Sp1 has been confirmed to activate pol III gene transcription by directly binding to both Brf1 and Gtf3c2 gene promoters. Thus, the findings from this study suggest that GATA4 links FLNA and Sp1 signaling to form an FLNA/GATA4/Sp1 axis to modulate pol III-directed transcription and transformed cell proliferation. Taken together, these results provide novel insights into the regulatory mechanism of pol III-directed transcription.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Houliang Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoye Song
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Wang
- School of Materials and Metallurgy, Wuhan University of Science and Technology, Wuhan, China
| | - Shasha Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Liu He
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiangyu Zhou
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaomei Yin
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kewei Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yue Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Zhongyu Wu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qiyue Chen
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Jiannan Du
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Deen Yu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shihua Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| | - Wensheng Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
HOPX Plays a Critical Role in Antiretroviral Drugs Induced Epigenetic Modification and Cardiac Hypertrophy. Cells 2021; 10:cells10123458. [PMID: 34943964 PMCID: PMC8700328 DOI: 10.3390/cells10123458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/23/2022] Open
Abstract
People living with HIV (PLWH) have to take an antiretroviral therapy (ART) for life and show noncommunicable illnesses such as chronic inflammation, immune activation, and multiorgan dysregulation. Recent studies suggest that long-term use of ART induces comorbid conditions and is one of the leading causes of heart failure in PLWH. However, the molecular mechanism of antiretroviral drugs (ARVs) induced heart failure is unclear. To determine the mechanism of ARVs induced cardiac dysfunction, we performed global transcriptomic profiling of ARVs treated neonatal rat ventricular cardiomyocytes in culture. Differentially expressed genes were identified by RNA-sequencing. Our data show that ARVs treatment causes upregulation of several biological functions associated with cardiotoxicity, hypertrophy, and heart failure. Global gene expression data were validated in cardiac tissue isolated from HIV patients having a history of ART. Interestingly, we found that homeodomain-only protein homeobox (HOPX) expression was significantly increased in cardiomyocytes treated with ARVs and in the heart tissue of HIV patients. Furthermore, we found that HOPX plays a crucial role in ARVs mediated cellular hypertrophy. Mechanistically, we found that HOPX plays a critical role in epigenetic regulation, through deacetylation of histone, while the HDAC inhibitor, Trichostatin A, can restore the acetylation level of histone 3 in the presence of ARVs.
Collapse
|
6
|
Shi X, Jiang N, Mao J, Luo D, Liu Y. Mesenchymal stem cell‐derived exosomes for organ development and cell‐free therapy. NANO SELECT 2021. [DOI: 10.1002/nano.202000286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Xin Shi
- Center and School of Stomatology Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan P.R. China
- Laboratory of Biomimetic Nanomaterials Department of Orthodontics National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology Beijing P.R. China
| | - Nan Jiang
- Laboratory of Biomimetic Nanomaterials Department of Orthodontics National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology Beijing P.R. China
- Central Laboratory National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology Beijing P.R. China
| | - Jing Mao
- Center and School of Stomatology Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan P.R. China
| | - Dan Luo
- CAS Center for Excellence in Nanoscience Beijing Key Laboratory of Micro‐nano Energy and Sensor Beijing Institute of Nanoenergy and Nanosystems Chinese Academy of Sciences Beijing P.R. China
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials Department of Orthodontics National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology Beijing P.R. China
| |
Collapse
|
7
|
Luo L, Gong J, Zhang H, Qin J, Li C, Zhang J, Tang Y, Zhang Y, Chen J, Zhou Y, Tian Z, Liu Y, Liu M. Cartilage Endplate Stem Cells Transdifferentiate Into Nucleus Pulposus Cells via Autocrine Exosomes. Front Cell Dev Biol 2021; 9:648201. [PMID: 33748142 PMCID: PMC7970302 DOI: 10.3389/fcell.2021.648201] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Stem cells derived from cartilage endplate (CEP) cells (CESCs) repair intervertebral disc (IVD) injury; however, the mechanism remains unclear. Here, we evaluated whether CESCs could transdifferentiate into nucleus pulposus cells (NPCs) via autocrine exosomes and subsequently inhibit IVD degeneration. Exosomes derived from CESCs (CESC-Exos) were extracted and identified by ultra-high-speed centrifugation and transmission electron microscopy. The effects of exosomes on the invasion, migration, and differentiation of CESCs were assessed. The exosome-activating hypoxia-inducible factor (HIF)-1α/Wnt pathway was investigated using lenti-HIF-1α and Wnt agonists/inhibitors in cells and gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis in normal and degenerated human CEP tissue. The effects of GATA binding protein 4 (GATA4) on transforming growth factor (TGF)-β expression and on the invasion, migration, and transdifferentiation of CESCs were investigated using lenti-GATA4, TGF-β agonists, and inhibitors. Additionally, IVD repair was investigated by injecting CESCs overexpressing GATA4 into rats. The results indicated that CESC-Exos promoted the invasion, migration, and differentiation of CESCs by autocrine exosomes via the HIF-1α/Wnt pathway. Additionally, increased HIF-1α enhanced the activation of Wnt signaling and activated GATA4 expression. GATA4 effectively promoted TGF-β secretion and enhanced the invasion, migration, and transdifferentiation of CESCs into NPCs, resulting in promotion of rat IVD repair. CESCs were also converted into NPCs as endplate degeneration progressed in human samples. Overall, we found that CESC-Exos activated HIF-1α/Wnt signaling via autocrine mechanisms to increase the expression of GATA4 and TGF-β1, thereby promoting the migration of CESCs into the IVD and the transformation of CESCs into NPCs and inhibiting IVDD.
Collapse
Affiliation(s)
- Liwen Luo
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China.,Institute of Immunology, PLA, Army Medical University, Third Military Medical University, Chongqing, China
| | - Junfeng Gong
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Hongyu Zhang
- Department of Emergency, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinghao Qin
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Changqing Li
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yu Tang
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Yang Zhang
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Jian Chen
- Institute of Immunology, PLA, Army Medical University, Third Military Medical University, Chongqing, China
| | - Yue Zhou
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - Zhiqiang Tian
- Institute of Immunology, PLA, Army Medical University, Third Military Medical University, Chongqing, China.,State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
| | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| | - MingHan Liu
- Department of Orthopaedics, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
The role of HOPX in normal tissues and tumor progression. Biosci Rep 2020; 40:221873. [PMID: 31934721 PMCID: PMC6997107 DOI: 10.1042/bsr20191953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
The homeodomain-only protein homeobox (HOPX) as the smallest homeodomain protein, lacks certain conserved residues required for DNA binding. Through our literature search, we reviewed the current understandings of HOPX in normal tissues and tumor progression. HOPX was initially identified as a critical transcription factor in various normal tissues, which interacted with serum response factor (SRF) or other substance to regulate normal physiological function. However, HOPX is at a low expression or methylation level in tumors. These data indicated that HOPX may play a very important role in regulating differentiation phenotype and tumor suppressive function. We predicted the prognosis of HOPX in tumors from TCGA database and discussed the downstream genes of HOPX. To understand how HOPX is involved in the mechanisms between physical and pathological conditions could lead to novel therapeutic strategies for treatment.
Collapse
|
9
|
Hao C, Lu Z, Zhao Y, Chen Z, Shen C, Ma G, Chen L. Overexpression of GATA4 enhances the antiapoptotic effect of exosomes secreted from cardiac colony-forming unit fibroblasts via miRNA221-mediated targeting of the PTEN/PI3K/AKT signaling pathway. Stem Cell Res Ther 2020; 11:251. [PMID: 32586406 PMCID: PMC7318537 DOI: 10.1186/s13287-020-01759-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/25/2020] [Accepted: 06/04/2020] [Indexed: 01/04/2023] Open
Abstract
Background GATA4 is an early cardiac-specific transcription factor, and endogenous GATA4-positive cells play a critical role in cardioprotection after myocardial injury. As functional paracrine units of therapeutic cells, exosomes can partially reproduce the reparative properties of their parental cells. Here, we investigated the cardioprotective capabilities of exosomes derived from cardiac colony-forming unit fibroblasts (cCFU-Fs) overexpressing GATA4 (cCFU-FsGATA4) and the underlying mechanism through which these exosomes use microRNA (miRNA) delivery to regulate target proteins in myocardial infarction (MI). Methods Exosomes were harvested from cCFU-Fs by ultracentrifugation. miRNA arrays were performed to determine differential miRNA expression between exosomes derived from cCFU-FsGATA4 (GATA4-Exo) and control cCFU-Fs (NC-Exo). A dual-luciferase reporter assay confirmed that miR221 directly targets the 3′ untranslated region (UTR) of the phosphatase and tensin homolog on chromosome ten (PTEN) gene. Cardiac function and myocardial infarct size were evaluated by echocardiography and Masson trichrome staining, respectively. Results Compared with NC-Exo, GATA4-Exo increased the survival and reduced the apoptosis of H9c2 cells. Direct intramyocardial transplantation of GATA4-Exo at the border of the ischemic region following ligation of the left anterior descending (LAD) coronary artery significantly restored cardiac contractile function and reduced infarct size. Microarray analysis revealed significantly increased miR221 expression in GATA4-Exo. qPCR confirmed higher miR221 levels in H9c2 cells treated with GATA4-Exo than in those treated with NC-Exo. miR221 mimic-transfected H9c2 cells demonstrated a significantly higher survival rate following exposure to hypoxic conditions than those transfected with miR221 inhibitor. A dual-luciferase reporter gene assay confirmed the PTEN gene as a target of miR221. Western blot analysis showed that H9c2 cells treated with GATA4-Exo exhibited lower PTEN protein expression and higher p-Akt expression. Conclusion GATA4 overexpression enhances the protective effect of cCFU-F-derived exosomes on myocardial ischemic injury. In terms of the mechanism, it is at least partly due to the miR221 transferred by GATA4-Exo, which inhibits PTEN expression, activates the phosphatidylinositol 3 kinase (PI3K)/AKT signaling pathway, and subsequently alleviates apoptosis of myocardial cells (CMs).
Collapse
Affiliation(s)
- Chunshu Hao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Zhengri Lu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Zhong Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.
| |
Collapse
|
10
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
11
|
Xiang Q, Zhou D, He X, Fan J, Tang J, Qiu Z, Zhang Y, Qiu J, Xu Y, Lai G. The zinc finger protein GATA4 induces mesenchymal-to-epithelial transition and cellular senescence through the nuclear factor-κB pathway in hepatocellular carcinoma. J Gastroenterol Hepatol 2019; 34:2196-2205. [PMID: 30995348 DOI: 10.1111/jgh.14684] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM The high mortality and poor prognosis of hepatocellular carcinoma (HCC) have raised the public attention. Gene therapy is considered as a promising treatment option for cancer; thus, finding a new therapeutic target for HCC is urgently needed. GATA4 is a tumor suppressor gene in multiple cancers, but its role in HCC is unclear. In this study, we explored the function of GATA4 in HCC. METHODS Reverse transcription-polymerase chain reaction and quantitative polymerase chain reaction were used to detect the mRNA expression of GATA4 in HCC cells and tissues. Cell viability, transwell, colony formation, and flow cytometry assays were applied to examine different aspects of biological effects of GATA4 in vitro. Xenografts, immunohistochemistry, and terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assays were performed to evaluate the effect of GATA4 on tumorigenicity in vivo. Western blotting, immunofluorescence, and β-galactosidase staining were used to investigate the mechanism underlying the function of GATA4. RESULTS We found that GATA4 was silenced in 15/19 (79%) HCC tissues. Restoring the expression of GATA4 induced G0 /G1 phase arrest, promoted apoptosis, suppressed HCC proliferation in vitro, and inhibited HCC tumor growth in vivo. Our data further showed that the ectopic expression of GATA4 induced cellular senescence through regulating nuclear factor-κB and inducing mesenchymal-to-epithelial transition. CONCLUSIONS Our data demonstrated that by inducing cellular senescence and mesenchymal-to-epithelial transition, GATA4 plays a crucial role as a tumor suppressor in HCC. It may thus be a potential cancer therapeutic target for HCC.
Collapse
Affiliation(s)
- Qin Xiang
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dishu Zhou
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Xiaoqian He
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiangxia Fan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Tang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Qiu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingfu Qiu
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yongzhu Xu
- School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Chongqing Health Service Center, Chongqing, China
| | - Guoqi Lai
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Inhibition of microRNA-429 attenuates oxygen-glucose deprivation/reoxygenation-induced neuronal injury by promoting expression of GATA-binding protein 4. Neuroreport 2019; 29:723-730. [PMID: 29624520 DOI: 10.1097/wnr.0000000000001023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) have been documented as critical regulators in ischemia/reperfusion-induced neuronal death. A better understanding of miRNA-mediated molecular mechanisms in ischemia/reperfusion-induced neuronal death may provide therapeutic targets for cerebral ischemia/reperfusion injury. A growing body of evidence suggests that miR-429 is a apoptosis-related miRNA that is also induced by hypoxia. However, whether miR-429 is involved in regulating neuronal apoptosis during cerebral ischemia/reperfusion injury remains unclear. In this study, the effect of miR-429 on oxygen-glucose deprivation and reoxygenation (OGD/R)-induced neuronal injury was investigated in vitro. The results showed that miR-429 expression levels were upregulated in cultured neurons with OGD/R treatment. The downregulation of miR-429 significantly alleviated OGD/R-induced neuronal injury, whereas upregulation of miR-429 aggravated it. Bioinformatic analysis showed that miR-429 could directly target the 3'-untranslated region of GATA-binding protein 4 (GATA4), which was verified by dual-luciferase reporter assay. Moreover, we found that miR-429 negatively regulated GATA4 expression. Overexpression of GATA4 also significantly alleviated OGD/R-induced neuronal injury. However, knockdown of GATA4 partially reversed the protective effect induced by miR-429 downregulation. Overall, our data showed that downregulation of miR-429 protected neurons against OGD/R-induced injury by promoting GATA4 and suggested a potential therapeutic target for the treatment of cerebral ischemia/reperfusion injury.
Collapse
|
13
|
Niderla-BieliŃska J, Jankowska-Steifer E, Flaht-Zabost A, Gula G, Czarnowska E, Ratajska A. Proepicardium: Current Understanding of its Structure, Induction, and Fate. Anat Rec (Hoboken) 2018; 302:893-903. [PMID: 30421563 DOI: 10.1002/ar.24028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 08/20/2018] [Accepted: 08/30/2018] [Indexed: 12/24/2022]
Abstract
The proepicardium (PE) is a transitory extracardiac embryonic structure which plays a crucial role in cardiac morphogenesis and delivers various cell lineages to the developing heart. The PE arises from the lateral plate mesoderm (LPM) and is present in all vertebrate species. During development, mesothelial cells of the PE reach the naked myocardium either as free-floating aggregates in the form of vesicles or via a tissue bridge; subsequently, they attach to the myocardium and, finally, form the third layer of a mature heart-the epicardium. After undergoing epithelial-to-mesenchymal transition (EMT) some of the epicardial cells migrate into the myocardial wall and differentiate into fibroblasts, smooth muscle cells, and possibly other cell types. Despite many recent findings, the molecular pathways that control not only proepicardial induction and differentiation but also epicardial formation and epicardial cell fate are poorly understood. Knowledge about these events is essential because molecular mechanisms that occur during embryonic development have been shown to be reactivated in pathological conditions, for example, after myocardial infarction, during hypertensive heart disease or other cardiovascular diseases. Therefore, in this review we intended to summarize the current knowledge about PE formation and structure, as well as proepicardial cell fate in animals commonly used as models for studies on heart development. Anat Rec, 302:893-903, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | | | - Grzegorz Gula
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland.,The Postgraduate School of Molecular Medicine (SMM), Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
14
|
Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functionally significant, novel GATA4
variants are frequently associated with Tetralogy of Fallot. Hum Mutat 2018; 39:1957-1972. [DOI: 10.1002/humu.23620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Ritu Dixit
- Cytogenetics Laboratory; Department of Zoology; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Chitra Narasimhan
- Department of Pediatric Cardiology; Sri Jayadeva Institute of Cardiovascular Sciences and Research; Bengaluru Karnataka India
| | - Vijyalakshmi I. Balekundri
- Super Speciality Hospital; Prime Minister Swasth Suraksha Yojana (PMSSY); Bengaluru Medical College and Research Institute; Bengaluru Karnataka India
| | - Damyanti Agrawal
- Department of Cardio-vascular and Thoracic Surgery; Institute of Medical Science; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Ashok Kumar
- Department of Pediatrics; Institute of Medical Sciences; Banaras Hindu University; Varanasi Uttar Pradesh India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory; Department of Zoology; Banaras Hindu University; Varanasi Uttar Pradesh India
| |
Collapse
|
15
|
Liu X, Li Z, Wang B, Zhu H, Liu Y, Qi J, Zhang Q. GATA4 is a transcriptional regulator of R-spondin1 in Japanese flounder (Paralichthys olivaceus). Gene 2018; 648:68-75. [PMID: 29331483 DOI: 10.1016/j.gene.2018.01.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/21/2017] [Accepted: 01/09/2018] [Indexed: 10/18/2022]
Abstract
GATA4 is a well-known transcription factor of the GATA family implicated in regulation of sex determination and gonadal development in mammals. In this study, we cloned the full-length cDNA of Paralichthys olivaceus gata4 (Po-gata4). Phylogenetic, gene structure, and synteny analysis showed that Po-GATA4 is homologous to GATA4 of teleost and tetrapod. Po-gata4 transcripts were detected in Sertoli cells, spermatogonia, oogonia and oocytes, with higher transcript levels overall in the testis than the ovary. The promoter region of P. olivaceus R-spondin1was found to contain a GATA4-binding motif. Results of CBA (cleaved amplified polymorphic sequence-based binding assay) indicated that GATA4 could indeed bind to the promoter sequence of R-spondin1. Moreover, human GATA4 recombinant protein could upregulate R-spondin1 in P. olivaceus ovary cells and FBCs (flounder brain cell line). In FBCs, overexpression of Po-gata4 resulted in elevated transcript levels of R-spondin1. Taken together, our results indicate that Po-GATA4 is involved in gonadal development by regulating R-spondin1 expression.
Collapse
Affiliation(s)
- Xiumei Liu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China
| | - Zan Li
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China
| | - Bo Wang
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China
| | - He Zhu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China
| | - Yuezhong Liu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China
| | - Jie Qi
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China.
| | - Quanqi Zhang
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 266003 Qingdao, Shandong, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, PR China
| |
Collapse
|
16
|
Han Q, Xu X, Li J, Wang J, Bai L, Wang A, Wang W, Zhang B. GATA4 is highly expressed in childhood acute lymphoblastic leukemia, promotes cell proliferation and inhibits apoptosis by activating BCL2 and MDM2. Mol Med Rep 2017; 16:6290-6298. [PMID: 28849107 DOI: 10.3892/mmr.2017.7369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 06/16/2017] [Indexed: 11/05/2022] Open
Abstract
Members of the GATA‑binding factor protein family, including GATA1, GATA2 and GATA3, serve an inhibiting role in leukemia. The present study demonstrated that GATA4 was upregulated in children with acute lymphoblastic leukemia (ALL). Results from a number of functional experiments, including cell proliferation analysis, cell cycle analysis, cell apoptosis assay and Transwell migration and invasion analyses, have suggested that high expression of GATA4 may facilitate proliferation and metastasis, and suppress apoptosis in ALL cells. Chromatin immunoprecipitation assay and luciferase reporter assay revealed that GATA4 was a transcription factor that activated mouse double minute 2 homolog (MDM2) and B cell lymphoma 2 (BCL2) expression in ALL cells. BCL2 is a key anti‑apoptosis protein that was demonstrated to suppress cell apoptosis. In addition, GATA4 was revealed to regulate p53 through the transcriptional activation of MDM2, subsequently influencing cell cycle and apoptosis. Results from the present study suggested that GATA4 may be a key marker in ALL diagnosis and a potential target of molecular therapy.
Collapse
Affiliation(s)
- Qiuguo Han
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Xin Xu
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Jing Li
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Jinggang Wang
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Li Bai
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Aihong Wang
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Wei Wang
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163000, P.R. China
| | - Bo Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
17
|
Lin HY, Zeng D, Liang YK, Wei XL, Chen CF. GATA3 and TRPS1 are distinct biomarkers and prognostic factors in breast cancer: database mining for GATA family members in malignancies. Oncotarget 2017; 8:34750-34761. [PMID: 28423734 PMCID: PMC5471008 DOI: 10.18632/oncotarget.16160] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/13/2017] [Indexed: 02/05/2023] Open
Abstract
GATA transcription factors are zinc finger DNA binding proteins that activate transcription during development and cell differentiation. To date, 7 members of GATA family have been reported. However, the expression patterns and the exact roles of distinct GATA family members contributing to tumorigenesis and progression of breast cancer (BC) remain to be elucidated. Here, we studied the expression of GATA transcripts in a variety of tumor types compared with the normal controls using the ONCOMINE and GOBO databases, along with their corresponding expression profiles in an array of cancer cell lines through CCLE analysis. Based on Kaplan-Meier plotter, we further investigated the prognostic values of GATA members specifically high expressed in BC patients. It was found that, when compared with normal tissues, GATA3 and TRPS1 were distinctly high expressed in BC patients among all GATA members. GATA3 expression was significantly associated with ESR1, while TRPS1 was correlated with ERBB2. In survival analysis, GATA3 and TRPS1 mRNA high expressions were correlated to better survival in BC patients, and TRPS1 high expression was significantly associated with longer RFS in patients who have received chemotherapy. These results suggest that GATA3 and TRPS1 are distinct biomarkers and essential prognostic factors for breast cancer.
Collapse
Affiliation(s)
- Hao-Yu Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - De Zeng
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- ChangJiang Scholar's Laboratory of Shantou University Medical College, Shantou, China
| | - Yuan-Ke Liang
- ChangJiang Scholar's Laboratory of Shantou University Medical College, Shantou, China
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of SUMC, Shantou, China
| | - Chun-Fa Chen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
18
|
Chen Z, Zhang S, Guo C, Li J, Sang W. Downregulation of miR-200c protects cardiomyocytes from hypoxia-induced apoptosis by targeting GATA-4. Int J Mol Med 2017; 39:1589-1596. [PMID: 28440427 DOI: 10.3892/ijmm.2017.2959] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/22/2017] [Indexed: 11/05/2022] Open
Abstract
Hypoxia-induced cardiomyocyte apoptosis plays an important role in the development of ischemic heart disease. MicroRNAs (miRNAs or miRs) are emerging as critical regulators of hypoxia-induced cardiomyocyte apoptosis. miR-200c is an miRNA that has been reported to be related to apoptosis in various pathological processes; however, its role in hypoxia‑induced cardiomyocyte apoptosis remains unclear. In the present study, we aimed to investigate the potential role and underlying mechanism of miR-200c in regulating hypoxia‑induced cardiomyocyte apoptosis. We found that miR-200c was significantly upregulated by hypoxia in cardiomyocytes, as detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The lactate dehydrogenase, MTT, Annexin V/propidium iodide apoptosis and caspase-3 activity assays showed that downregulation of miR-200c markedly improved cell survival and suppressed the apoptosis of cardiomyocytes in response to hypoxia. Bioinformatics analysis and the dual-luciferase reporter assay demonstrated that miR-200c directly targeted the 3'-untranslated region of GATA-4, an important transcription factor for cardiomyocyte survival. RT-qPCR and western blot analysis showed that suppression of miR-200c significantly increased GATA-4 expression. Furthermore, downregulation of miR-200c upregulated the expression of the anti-apoptotic gene Bcl-2. However, the protective effects against hypoxia induced by the downregulation of miR‑200c were significantly abolished by GATA-4 knockdown. Taken together, our results suggest that downregulation of miR-200c protects cardiomyocytes from hypoxia-induced apoptosis by targeting GATA-4, providing a potential therapeutic molecular target for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Zhigang Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Shaoli Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Changlei Guo
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jianhua Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Wenfeng Sang
- Department of Internal Medicine Nursing, College of Nursing, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
19
|
Constanzo JD, Deng M, Rindhe S, Tang KJ, Zhang CC, Scaglioni PP. Pias1 is essential for erythroid and vascular development in the mouse embryo. Dev Biol 2016; 415:98-110. [PMID: 27155222 DOI: 10.1016/j.ydbio.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 01/26/2023]
Abstract
The protein inhibitor of activated STAT-1 (PIAS1) is one of the few known SUMO E3 ligases. PIAS1 has been implicated in several biological processes including repression of innate immunity and DNA repair. However, PIAS1 function during development and tissue differentiation has not been studied. Here, we report that Pias1 is required for proper embryonic development. Approximately 90% of Pias1 null embryos die in utero between E10.5 and E12.5. We found significant apoptosis within the yolk sac (YS) blood vessels and concomitant loss of red blood cells (RBCs) resulting in profound anemia. In addition, Pias1 loss impairs YS angiogenesis and results in defective capillary plexus formation and blood vessel occlusions. Moreover, heart development is impaired as a result of loss of myocardium muscle mass. Accordingly, we found that Pias1 expression in primary myoblasts enhances the induction of cardiac muscle genes MyoD, Myogenin and Myomaker. PIAS1 protein regulation of cardiac gene transcription is dependent on transcription factors Myocardin and Gata-4. Finally, endothelial cell specific inactivation of Pias1 in vivo impairs YS erythrogenesis, angiogenesis and recapitulates loss of myocardium muscle mass. However, these defects are not sufficient to recapitulate the lethal phenotype of Pias1 null embryos. These findings highlight Pias1 as an essential gene for YS erythropoiesis and vasculogenesis in vivo.
Collapse
Affiliation(s)
- Jerfiz D Constanzo
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mi Deng
- Departments of Physiology, and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Smita Rindhe
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ke-Jing Tang
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pulmonary Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Cheng-Cheng Zhang
- Departments of Physiology, and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pier Paolo Scaglioni
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
20
|
Liu J, Zhang W, Du X, Jiang J, Wang C, Wang X, Zhang Q, He Y. Molecular characterization and functional analysis of the GATA4 in tongue sole (Cynoglossus semilaevis). Comp Biochem Physiol B Biochem Mol Biol 2016; 193:1-8. [DOI: 10.1016/j.cbpb.2015.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 11/23/2015] [Accepted: 12/02/2015] [Indexed: 01/11/2023]
|
21
|
Yuan W, Liu Y, Lok JB, Stoltzfus JD, Gasser RB, Lei W, Fang R, Zhao J, Hu M. Exploring features and function of Ss-riok-3, an enigmatic kinase gene from Strongyloides stercoralis. Parasit Vectors 2014; 7:561. [PMID: 25477034 PMCID: PMC4265397 DOI: 10.1186/s13071-014-0561-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/23/2014] [Indexed: 02/01/2023] Open
Abstract
Background Right open reading frame protein kinase 3 (RIOK-3) belongs to the atypical kinase family. Unlike the other two members, RIOK-1 and RIOK-2, which are conserved from Archaea to humans, RIOK-3 occurs only in multicellular organisms. Studies on HeLa cells indicate that human RIOK-3 is a component of the 40S small ribosome subunit and supports cancer cell growth and survival. However, almost nothing is known about the function of RIOK-3. We explored the functional role of RIOK-3 encoding gene from Strongyloides stercoralis, a parasitic nematode of humans and dogs. Methods To analyze the gene and promoter structure of Ss-riok-3, RACE-PCR and Genome-walker PCR were performed to isolate the full length cDNA, gDNA and promoter region of Ss-riok-3. RNA-seq was conducted to assess the transcript abundance of Ss-riok-3 in different stages of S. stercoralis. Transgenesis was employed to determine the anatomic expression patterns of Ss-riok-3. Results The RIOK-3 protein-encoding gene (designated Ss-riok-3) of S. stercoralis was characterized. The full-length complementary and genomic DNAs of the RIOK-3 encoding gene (riok-3) were isolated from this nematode. The cDNA of Ss-riok-3 is 1,757 bp in length, including a 23 bp 5’-UTR, a 36 bp 3’-UTR and a 1,698 bp coding region encoding a protein of 565 amino acids (aa) containing a RIO kinase domain. RNA sequencing (RNA-seq) analysis revealed that Ss-riok-3 is transcribed in all developmental stages of S. stercoralis assessed, with transcripts being particularly abundant in parasitic females. Gene structure analysis revealed that Ss-riok-3 contains no intron. The putative promoter contains conserved promoter elements, including four TATA, two GATA, one inverse GATA and one inverse CAAT boxes. The promoter of Ss-riok-3 drives GFP expression in the head neuron, intestine and body wall muscle of transgenic S. stercoralis larvae, and the TATA boxes present in the 3’-UTR of the gene immediately upstream of Ss-riok-3 initiate transcription. Conclusions The characterization of the RIOK-3 encoding gene from S. stercoralis provides a sound foundation for investigating in detail its function in the development and reproduction of this important pathogen. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0561-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wang Yuan
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Yingying Liu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - James B Lok
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Jonathan D Stoltzfus
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA. .,Department of Biology, Hollins University, Roanoke, VI, 24020, USA.
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner of Flemington Road and Park Drive, Parkville, VI, 3010, Australia.
| | - Weiqiang Lei
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| |
Collapse
|
22
|
Jun JH, Jun NH, Shim JK, Shin EJ, Kwak YL. Erythropoietin protects myocardium against ischemia-reperfusion injury under moderate hyperglycemia. Eur J Pharmacol 2014; 745:1-9. [PMID: 25446919 DOI: 10.1016/j.ejphar.2014.09.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 01/26/2023]
Abstract
Erythropoietin (EPO), an essential hormone for erythropoiesis, provides protection against myocardial ischemia/reperfusion (I/R) injury. Hyperglycemia during acute myocardial infarction aggravates organ damage and attenuates the efficacies of various protective measures. This study aimed to investigate the protective role of EPO against myocardial I/R injury under a clinically relevant moderate hyperglycemic condition and its associated mechanisms. Eighty-two Sprague-Dawley rats were randomly assigned to six groups: normoglycemia-Sham, normoglycemia-I/R-control-saline (IRC), normoglycemia-I/R-EPO (IRE), hyperglycemia-Sham, hyperglycemia-IRC, and hyperglycemia-IRE. The rats received 1.2 g/kg dextrose or same volume of normal saline depending on the group. I/R was induced by a 30 min period of ischemia followed by reperfusion for 4 h. For 1 h before I/R injury, intravenous 4000 IU/kg of EPO was administered. EPO pretreatment significantly reduced the number of apoptotic cells and the infarct size compared with those of the control groups. EPO increased GATA-4 phosphorylation and acetylation against I/R in hyperglycemic myocardium. It also enhanced ERK induced GATA-4 post-translational modifications such as increased GATA-4 phosphorylation and acetylation, and decreased GATA-4 ubiquitination following hypoxia-reoxygenation in H9c2 cells in hyperglycemic medium. Increased GATA-4 stability by EPO diminished I/R-related down-regulation of Bcl-2 and reduction of caspase-3 activities in hyperglycemic myocardium. In conclusion, EPO pretreatment before I/R injury conveyed significant myocardial protection under moderate hyperglycemic condition through mechanisms involved in reduction of caspase-3 activity and up-regulation of Bcl-2 in association with enhanced ERK-induced GATA-4 stability.
Collapse
Affiliation(s)
- Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Na-Hyung Jun
- Department of Anesthesiology and Pain Medicine, National Health Insurance Corporation Ilsan Hospital, Goyang, Republic of Korea
| | - Jae-Kwang Shim
- Department of Anesthesiology and Pain Medicine, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Shin
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Lan Kwak
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Sakata H, Maéno M. Nkx2.5 is involved in myeloid cell differentiation at anterior ventral blood islands in the Xenopus embryo. Dev Growth Differ 2014; 56:544-54. [PMID: 25283688 DOI: 10.1111/dgd.12155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 11/28/2022]
Abstract
We have shown previously that two populations of myeloid cells emerge in the anterior and posterior ventral blood islands (aVBI and pVBI) at the different stages in Xenopus laevis embryo. In order to elucidate the regulatory mechanism of myeloid cell differentiation in the aVBI, we examined the role of Nkx2.5, an essential transcription factor for heart differentiation, in regulation of the myeloid cell differentiation in this region. Knockdown of endogenous Nkx2.5 by introducing MO into the dorsal marginal zone (DMZ) suppressed the expression of MHCα as well as that of mpo and spib in the resultant embryos and in DMZ explants made from the injected embryos. Expression of c/ebpα was less affected in the embryos injected with Nkx2.5 MO. The effect of Nkx2.5 MO in myeloid cell differentiation was recovered by coinjection of nkx2.5 or c/ebpα mRNA, indicating that Nkx2.5 functions at the same or the upper level of C/EBPα for the specification of myeloid cells. An attempt to identify transcription factors for myeloid cell differentiation in ventral marginal zone (VMZ) explants demonstrated that coinjection of two transcription factors out of three factors, namely C/EBPα, Nkx2.5 and GATA4, was sufficient to induce a certain amount of mpo expression. We suggest that C/EBPα is an unequivocal factor for myeloid cell differentiation in the aVBI and that Nkx2.5 and GATA4 cooperate with C/EBPα for promotion of myeloid cell differentiation.
Collapse
Affiliation(s)
- Hiroyuki Sakata
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata, 950-2181, Japan
| | | |
Collapse
|
24
|
Takagi K, Moriguchi T, Miki Y, Nakamura Y, Watanabe M, Ishida T, Yamamoto M, Sasano H, Suzuki T. GATA4 immunolocalization in breast carcinoma as a potent prognostic predictor. Cancer Sci 2014; 105:600-7. [PMID: 24862985 PMCID: PMC4317835 DOI: 10.1111/cas.12382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/23/2022] Open
Abstract
Transcriptional GATA factors are known lineage selector genes and regulate a variety of biological processes including specification and differentiation of tissues. In the present study, we examined expression profiles of six GATA factor genes in invasive ductal carcinomas (IDC) of the breast using microarray analysis (n = 20) and found that GATA4 expression was closely correlated with recurrence in patients. Because the significance of GATA4 has remained largely unknown in breast carcinoma, we further immunolocalized GATA4 in ductal carcinoma in situ (DCIS) of the breast (n = 48) and IDC (n = 163). GATA4 immunoreactivity was detected in the nuclei of carcinoma cells and was positive in 27% of DCIS and 31% of IDC cases. GATA4 status was significantly associated with nuclear grade and van Nuys classification in DCIS and was positively associated with distant metastasis, histological grade and HER2 status, but negatively correlated with progesterone receptor labeling index in IDC. Subsequent multivariate analysis demonstrated that GATA4 status was an independent prognostic factor for both disease-free and breast cancer-specific survival of IDC patients. All of these results indicate that GATA4 plays important roles in the progression of breast carcinoma from an early stage and that immunohistochemical GATA4 status is considered a potent prognostic factor in human breast cancer patients.
Collapse
Affiliation(s)
- Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lessons from the heart: mirroring electrophysiological characteristics during cardiac development to in vitro differentiation of stem cell derived cardiomyocytes. J Mol Cell Cardiol 2013; 67:12-25. [PMID: 24370890 DOI: 10.1016/j.yjmcc.2013.12.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 11/14/2013] [Accepted: 12/13/2013] [Indexed: 01/12/2023]
Abstract
The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.
Collapse
|
26
|
Jun JH, Shin EJ, Kim JH, Kim SO, Shim JK, Kwak YL. Erythropoietin prevents hypoxia-induced GATA-4 ubiquitination via phosphorylation of serine 105 of GATA-4. Biol Pharm Bull 2013; 36:1126-33. [PMID: 23811561 DOI: 10.1248/bpb.b13-00100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Erythropoietin (EPO), an essential hormone for erythropoiesis, can provide protection against myocardial ischemia/reperfusion (I/R) injury and hypoxic apoptosis. GATA-4 is a zinc finger transcription factor, and its activation and post-translational modification are essential components in the transcriptional response to hypoxia. GATA-4 has also been reported to play a role in the cellular mechanisms of EPO-induced myocardial protection against I/R injury. In this study, we aimed to investigate the influence of EPO on GATA-4 protein stability and post-translational modification under hypoxic conditions without reperfusion. EPO induced cell viability under long-term hypoxia. EPO significantly increased phosphorylation of GATA-4 via the extracellular signal-regulated kinase (ERK) signaling pathway and reduced hypoxia-induced GATA-4 ubiquitination, which enhanced GATA-4 stability under hypoxia. ERK activation by over-expression of constitutively active mitogen-activated protein kinase 1 (MEK1) strongly increased GATA-4 phosphorylation and its protein levels and decreased GATA-4 ubiquitination under hypoxia. Despite ERK activation, GATA-4 ubiquitination was not affected under hypoxia in a GATA-4-S105A mutant. Under hypoxic condition without reperfusion, EPO-induced ERK activation was associated with post-translational modification of GATA-4, mediated by enhancement of phosphorylation of GATA-4 at Ser-105. Subsequent attenuation of GATA-4 ubiquitination led to increases in GATA-4 protein stability, which resulted in increased cell viability under hypoxia.
Collapse
Affiliation(s)
- Ji Hae Jun
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120–752, Republic of Korea
| | | | | | | | | | | |
Collapse
|
27
|
Yu B, Gong M, He Z, Wang YG, Millard RW, Ashraf M, Xu M. Enhanced mesenchymal stem cell survival induced by GATA-4 overexpression is partially mediated by regulation of the miR-15 family. Int J Biochem Cell Biol 2013; 45:2724-35. [PMID: 24070634 DOI: 10.1016/j.biocel.2013.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/12/2013] [Accepted: 09/16/2013] [Indexed: 01/12/2023]
Abstract
UNLABELLED We reported previously that pre-programming mesenchymal stem cells with the GATA-4 gene increases significantly cell survival in an ischemic environment. In this study, we tested whether regulation of microRNAs and their target proteins was associated with the cytoprotective effects of GATA-4. METHODS AND RESULTS Mesenchymal stem cells were harvested from adult rat bone marrow and transduced with GATA-4 (MSC(GATA-4)) using the murine stem cell virus retroviral expression system. Cells transfected with empty vector (MSC(Null)) were used as controls. Quantitative real-time PCR data showed that the expression levels of miR-15 family members (miR-15b, miR-16, and miR-195) were significantly down-regulated in MSC(GATA-4). The protein expression of Bcl-w (Bcl-2-like-2), an anti-apoptotic Bcl-2 family protein, was increased in MSC(GATA-4). Hypoxic culture (low glucose and low oxygen) induced the release of lactate dehydrogenase from mesenchymal stem cells and reduced cell survival. Compared to MSC(Null), MSC(GATA-4) showed less lactate dehydrogenase release and greater cell survival following 72 h hypoxia exposure. The mitochondrial membrane potential, detected with the dye JC-1, was well maintained, and mitochondrial membrane permeability, expressed as caspase 3 and 7 activities in response to the ischemic environment was lower in MSC(GATA-4). Moreover, transfection with miR-195 significantly down-regulated Bcl-w expression in mesenchymal stem cells through a binding site in the 3'-UTR of the Bcl-w mRNA and reduced mesenchymal stem cell resistance to ischemic injury. CONCLUSIONS The overexpression of GATA-4 in mesenchymal stem cells down-regulates miR-15 family members, causing increased resistance to ischemia through the up-regulation of anti-apoptotic proteins in the Bcl-2 family.
Collapse
Affiliation(s)
- Bin Yu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Suzuki M, Kobayashi-Osaki M, Tsutsumi S, Pan X, Ohmori S, Takai J, Moriguchi T, Ohneda O, Ohneda K, Shimizu R, Kanki Y, Kodama T, Aburatani H, Yamamoto M. GATA factor switching from GATA2 to GATA1 contributes to erythroid differentiation. Genes Cells 2013; 18:921-33. [PMID: 23911012 DOI: 10.1111/gtc.12086] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/16/2013] [Indexed: 11/30/2022]
Abstract
Transcription factor GATA2 is highly expressed in hematopoietic stem cells and progenitors, whereas its expression declines after erythroid commitment of progenitors. In contrast, the start of GATA1 expression coincides with the erythroid commitment and increases along with the erythroid differentiation. We refer this dynamic transition of GATA factor expression to as the 'GATA factor switching'. Here, we examined contribution of the GATA factor switching to the erythroid differentiation. In Gata1-knockdown embryos that concomitantly express Gata2-GFP reporter, high-level expression of GFP reporter was detected in accumulated immature hematopoietic cells with impaired differentiation, demonstrating that GATA1 represses Gata2 gene expression in hematopoietic progenitors in vivo. We have conducted chromatin immunoprecipitation (ChIP) on microarray analyses of GATA2 and GATA1, and results indicate that the GATA1-binding sites widely overlap with the sites pre-occupied by GATA2 before the GATA1 expression. Importantly, erythroid genes harboring GATA boxes bound by both GATA1 and GATA2 tend to be expressed in immature erythroid cells, whereas those harboring GATA boxes to which GATA1 binds highly but GATA2 binds only weakly are important for the mature erythroid cell function. Our results thus support the contention that preceding binding of GATA2 helps the following binding of GATA1 and thereby secures smooth expression of the transient-phase genes.
Collapse
Affiliation(s)
- Mikiko Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan; Center for Radioisotope Sciences, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan; Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Viger RS, Taniguchi H, Robert NM, Tremblay JJ. The 25th Volume: Role of the GATA Family of Transcription Factors in Andrology. ACTA ACUST UNITED AC 2013; 25:441-52. [PMID: 15223831 DOI: 10.1002/j.1939-4640.2004.tb02813.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Robert S Viger
- Ontogeny-Reproduction Research Unit, CHUL Research Centre, and Centre de Recherche en Biologie de la Reproduction, Department of Obstetrics and Gynecology, Faculty of Medicine, Université Laval, Ste-Foy, Québec, Canada.
| | | | | | | |
Collapse
|
30
|
Lai F, Singh A, King ML. Xenopus Nanos1 is required to prevent endoderm gene expression and apoptosis in primordial germ cells. Development 2012; 139:1476-86. [PMID: 22399685 DOI: 10.1242/dev.079608] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nanos is expressed in multipotent cells, stem cells and primordial germ cells (PGCs) of organisms as diverse as jellyfish and humans. It functions together with Pumilio to translationally repress targeted mRNAs. Here we show by loss-of-function experiments that Xenopus Nanos1 is required to preserve PGC fate. Morpholino knockdown of maternal Nanos1 resulted in a striking decrease in PGCs and a loss of germ cells from the gonads. Lineage tracing and TUNEL staining reveal that Nanos1-deficient PGCs fail to migrate out of the endoderm. They appear to undergo apoptosis rather than convert to normal endoderm. Whereas normal PGCs do not become transcriptionally active until neurula, Nanos1-depleted PGCs prematurely exhibit a hyperphosphorylated RNA polymerase II C-terminal domain at the midblastula transition. Furthermore, they inappropriately express somatic genes characteristic of endoderm regulated by maternal VegT, including Xsox17α, Bix4, Mixer, GATA4 and Edd. We further demonstrate that Pumilio specifically binds VegT RNA in vitro and represses, along with Nanos1, VegT translation within PGCs. Repressed VegT RNA in wild-type PGCs is significantly less stable than VegT in Nanos1-depleted PGCs. Our data indicate that maternal VegT RNA is an authentic target of Nanos1/Pumilio translational repression. We propose that Nanos1 functions to translationally repress RNAs that normally specify endoderm and promote apoptosis, thus preserving the germline.
Collapse
Affiliation(s)
- Fangfang Lai
- Department of Cell Biology, University of Miami School of Medicine, 1011 NW 15th St, Miami, FL 33136, USA
| | | | | |
Collapse
|
31
|
Li J, Chen W, Wang D, Zhou L, Sakai F, Guan G, Nagahama Y. GATA4 is involved in the gonadal development and maturation of the teleost fish tilapia, Oreochromis niloticus. J Reprod Dev 2011; 58:237-42. [PMID: 22186677 DOI: 10.1262/jrd.11-131s] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GATA4, a member of the GATA family, is a well-known transcription factor implicated in the regulation of sex determination and sexual differentiation in mammals. However, little is known about the possible role of GATA4 in fish reproduction. In the present study, a full-length GATA4 cDNA from the tilapia was cloned and characterized. The tilapia GATA4 gene contained an open reading frame (ORF) of 1179 nucleotides encoding a protein of 392 amino acids. Sequence alignment revealed that the tilapia GATA4 protein shared higher homology (ranging from 63.1 to 74.6%) with other vertebrates. RT-PCR analysis indicated that the GATA4 gene is expressed in the ovary, testis, liver, intestine and heart in adult tilapia. In situ hybridization was performed to examine the temporal and spatial expression patterns of GATA4 during tilapia gonadal differentiation and development. In the undifferentiated gonad, GATA4 was expressed in the somatic cells of both sexes. Subsequently, GATA4 expression persisted in the differentiated, juvenile and adult ovary and testis in tilapia. Our data indicate for the first time that GATA4 is not only necessary for the onset of gonadal differentiation, but also important for gonadal development and maturation.
Collapse
Affiliation(s)
- Jianzhong Li
- Key Lab of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Molekulargenetische Grundlagen des Vorhofseptumdefekts. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2011. [DOI: 10.1007/s00398-011-0866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
33
|
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
|
34
|
Chen M, Lin YQ, Xie SL, Wang JF. Mitogen-activated protein kinase in endothelin-1-induced cardiac differentiation of mouse embryonic stem cells. J Cell Biochem 2011; 111:1619-28. [PMID: 21053276 DOI: 10.1002/jcb.22895] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelin-1(ET-1) is a potent vasoconstrictor involved in the development of cardiovascular diseases and is an important regulator of heart development. However, the role of ET-1 in cardiac differentiation of mouse embryonic stem cells (mESCs) and the underlying molecular mechanisms remain poorly understood. In the present study, we showed that ET-1 significantly up-regulated gene expression of the cardiac specific transcriptional factors Nkx2.5, GATA4, and conduction system specific marker CX40, with no affect on the gene expression of α-MHC and β-MHC in cardiac differentiation of mESCs. The percentage of beating embryoid bodies (EB) and the Troponin T (TnT) positive area in total EBs was unchanged following ET-1 treatment, while the percentage of spindle cells that stained positively with TnT was increased in the presence of ET-1. Further investigation indicated that the percentage of beating EBs and the TnT positive area were decreased by the extracellular signal-related kinases (ERK)-1/2 inhibitor U0126 and the p38 inhibitor SB203580, but not by the Jun amino-terminal kinases (JNK) inhibitor SP600125. Inhibition of ERK1/2, p38, and JNK pathways also blocked the up-regulation of Nkx2.5 and GATA4 by ET-1, however only inhibition of the ERK1/2 pathway had negatively effects on the increase in CX40 expression in response to ET-1. ET-1 induced an increase in the percentage of spindle cells was also inhibited by U0126. Our results suggest that ET-1 plays a significant role in the cardiac differentiation of mESCs, especially in those cells committed to the conduction system, with the ERK1/2 pathway playing a critical role in this process.
Collapse
Affiliation(s)
- Ming Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | |
Collapse
|
35
|
|
36
|
Kawaguchi N, Smith AJ, Waring CD, Hasan MK, Miyamoto S, Matsuoka R, Ellison GM. c-kitpos GATA-4 high rat cardiac stem cells foster adult cardiomyocyte survival through IGF-1 paracrine signalling. PLoS One 2010; 5:e14297. [PMID: 21179204 PMCID: PMC3001457 DOI: 10.1371/journal.pone.0014297] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 11/17/2010] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Resident c-kit positive (c-kitpos) cardiac stem cells (CSCs) could be considered the most appropriate cell type for myocardial regeneration therapies. However, much is still unknown regarding their biological properties and potential. METHODOLOGY/PRINCIPAL FINDINGS We produced clones of high and low expressing GATA-4 CSCs from long-term bulk-cultured c-kitpos CSCs isolated from adult rat hearts. When c-kitpos GATA-4 high expressing clonal CSCs (cCSCs) were co-cultured with adult rat ventricular cardiomyocytes, we observed increased survival and contractility of the cardiomyocytes, compared to cardiomyocytes cultured alone, co-cultured with fibroblasts or c-kitpos GATA-4 low expressing cCSCs. When analysed by ELISA, the concentration of IGF-1 was significantly increased in the c-kitpos GATA-4 high cCSC/cardiomyocyte co-cultures and there was a significant correlation between IGF-1 concentration and cardiomyocyte survival. We showed the activation of the IGF-1 receptor and its downstream molecular targets in cardiomyocytes co-cultured with c-kitpos GATA-4 high cCSCs but not in cardiomyocytes that were cultured alone, co-cultured with fibroblasts or c-kitpos GATA-4 low cCSCs. Addition of a blocking antibody specific to the IGF-1 receptor inhibited the survival of cardiomyocytes and prevented the activation of its signalling in cardiomyocytes in the c-kitpos GATA-4 high cCSC/cardiomyocyte co-culture system. IGF-1 supplementation or IGF-1 high conditioned medium taken from the co-culture of c-kitpos GATA-4 high cCSCs plus cardiomyocytes did extend the survival and contractility of cardiomyocytes cultured alone and cardiomyocytes co-cultured with c-kitpos GATA-4 low cCSCs. CONCLUSION/SIGNIFICANCE c-kitpos GATA-4 high cCSCs exert a paracrine survival effect on cardiomyocytes through induction of the IGF-1R and signalling pathway.
Collapse
Affiliation(s)
- Nanako Kawaguchi
- International Research and Educational Institute for Integrated Medical Sciences (IREIIMS), Tokyo Women's Medical University, Tokyo, Japan
| | - Andrew J. Smith
- Stem Cell and Molecular Physiology Laboratory, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Cheryl D. Waring
- Stem Cell and Molecular Physiology Laboratory, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Md Kamrul Hasan
- International Research and Educational Institute for Integrated Medical Sciences (IREIIMS), Tokyo Women's Medical University, Tokyo, Japan
| | - Shinka Miyamoto
- International Research and Educational Institute for Integrated Medical Sciences (IREIIMS), Tokyo Women's Medical University, Tokyo, Japan
- Department of Cardiovascular Surgery, Heart Institute of Japan, Tokyo Women's Medical University, Tokyo, Japan
| | - Rumiko Matsuoka
- International Research and Educational Institute for Integrated Medical Sciences (IREIIMS), Tokyo Women's Medical University, Tokyo, Japan
| | - Georgina M. Ellison
- Stem Cell and Molecular Physiology Laboratory, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
Trivedi CM, Zhu W, Wang Q, Jia C, Kee HJ, Li L, Hannenhalli S, Epstein JA. Hopx and Hdac2 interact to modulate Gata4 acetylation and embryonic cardiac myocyte proliferation. Dev Cell 2010; 19:450-9. [PMID: 20833366 DOI: 10.1016/j.devcel.2010.08.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 07/22/2010] [Accepted: 08/20/2010] [Indexed: 10/19/2022]
Abstract
Regulation of chromatin structure via histone modification has recently received intense attention. Here, we demonstrate that the chromatin-modifying enzyme histone deacetylase 2 (Hdac2) functions with a small homeodomain factor, Hopx, to mediate deacetylation of Gata4, which is expressed by cardiac progenitor cells and plays critical roles in the regulation of cardiogenesis. In the absence of Hopx and Hdac2 in mouse embryos, Gata4 hyperacetylation is associated with a marked increase in cardiac myocyte proliferation, upregulation of Gata4 target genes, and perinatal lethality. Hdac2 physically interacts with Gata4, and this interaction is stabilized by Hopx. The ability of Gata4 to transactivate cell cycle genes is impaired by Hopx/Hdac2-mediated deacetylation, and this effect is abrogated by loss of Hdac2-Gata4 interaction. These results suggest that Gata4 is a nonhistone target of Hdac2-mediated deacetylation and that Hdac2, Hopx, and Gata4 coordinately regulate cardiac myocyte proliferation during embryonic development.
Collapse
Affiliation(s)
- Chinmay M Trivedi
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Witte DP, Aronow BJ, Harmony JAK. Understanding Cardiac Development Through the Perspective of Gene Regulation and Gene Manipulation. ACTA ACUST UNITED AC 2010. [DOI: 10.1080/15513819609169282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
39
|
Miyamoto S, Kawaguchi N, Ellison GM, Matsuoka R, Shin'oka T, Kurosawa H. Characterization of long-term cultured c-kit+ cardiac stem cells derived from adult rat hearts. Stem Cells Dev 2010; 19:105-16. [PMID: 19580375 DOI: 10.1089/scd.2009.0041] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Previous studies have revealed c-kit-positive (c-kit(+)) cardiac stem cells (CSCs) in the adult mammalian heart and these cells could be a suitable cell source for heart regeneration therapy. However, these cells have not been fully evaluated in terms of characterization and effect of long-term culture, which is necessary for their safe and optimal usage. Therefore, we isolated c-kit(+) CSCs from adult rat hearts to characterize these cells and investigate stability over long-term culture. We performed isolations of c-kit(+) CSCs 11 times and passaged them 40 times in a bulk culture system; we termed these cultures, bulk culture CSCs (CSC-BC). c-kit(+) CSCs expressed stemness genes and exhibited stem cell properties of single cell-derived clone formation, cardiosphere generation, and potential to differentiate into the three main cardiac lineages: cardiomyocyte, smooth muscle, and endothelial cells in vitro. Over long-term culture, some CSC-BC up-regulated GATA-4 expression, which resulted in enhanced cardiomyocyte differentiation, suggesting that the GATA-4 high c-kit(+) CSCs have potent cardiac regenerative potential. We also observed the spontaneous differentiation into cells other than cardiac lineages, such as adipocyte and skeletal myocyte. This effect of long-term culture on the c-kit(+) CSCs has not been previously reported. Interestingly, when c-kit(+) CSCs were co-cultured with adult rat cardiomyocytes, we found increased cardiomyocyte survival, and the growth factors, insulin-like growth factor 1 (IGF-1) and vascular endothelial growth factor (VEGF), appeared to be responsible factors. The present study suggests that c-kit(+) CSCs have great therapeutic potential yet should be further investigated and optimized as a cell source for regenerative therapies prior to transplantation.
Collapse
Affiliation(s)
- Shinka Miyamoto
- Department of Cardiovascular Surgery, Heart Institute of Japan, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Posch MG, Perrot A, Berger F, Ozcelik C. Molecular genetics of congenital atrial septal defects. Clin Res Cardiol 2010; 99:137-47. [PMID: 20012542 PMCID: PMC2830584 DOI: 10.1007/s00392-009-0095-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 11/19/2009] [Indexed: 11/24/2022]
Abstract
Congenital heart defects (CHD) are the most common developmental errors in humans, affecting 8 out of 1,000 newborns. Clinical diagnosis and treatment of CHD has dramatically improved in the last decades. Hence, the majority of CHD patients are now reaching reproductive age. While the risk of familial recurrence has been evaluated in various population studies, little is known about the genetic pathogenesis of CHD. In recent years significant progress has been made in uncovering genetic processes during cardiac development. Data from human genetic studies in CHD patients indicate that the genetic aetiology was presumably underestimated in the past. Inherited mutations in genes encoding cardiac transcription factors and sarcomeric proteins were found as an underlying cause for familial recurrence of non-syndromic CHD in humans, in particular cardiac septal defects. Notably, the cardiac phenotypes most frequently seen in mutation carriers are ostium secundum atrial septal defects (ASDII). This review outlines experimental approaches employed for the detection of CHD-related genes in humans and summarizes recent findings in molecular genetics of congenital cardiac septal defects with an emphasis on ASDII.
Collapse
Affiliation(s)
- Maximilian G Posch
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Germany.
| | | | | | | |
Collapse
|
41
|
c-Jun NH2-terminal kinase is required for lineage-specific differentiation but not stem cell self-renewal. Mol Cell Biol 2010; 30:1329-40. [PMID: 20065035 DOI: 10.1128/mcb.00795-09] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The c-Jun NH(2)-terminal kinase (JNK) is implicated in proliferation. Mice with a deficiency of either the Jnk1 or the Jnk2 genes are viable, but a compound deficiency of both Jnk1 and Jnk2 causes early embryonic lethality. Studies using conditional gene ablation and chemical genetic approaches demonstrate that the combined loss of JNK1 and JNK2 protein kinase function results in rapid senescence. To test whether this role of JNK was required for stem cell proliferation, we isolated embryonic stem (ES) cells from wild-type and JNK-deficient mice. We found that Jnk1(-/-) Jnk2(-/-) ES cells underwent self-renewal, but these cells proliferated more rapidly than wild-type ES cells and exhibited major defects in lineage-specific differentiation. Together, these data demonstrate that JNK is not required for proliferation or self-renewal of ES cells, but JNK plays a key role in the differentiation of ES cells.
Collapse
|
42
|
Miazga CM, McLaughlin KA. Coordinating the timing of cardiac precursor development during gastrulation: A new role for Notch signaling. Dev Biol 2009; 333:285-96. [DOI: 10.1016/j.ydbio.2009.06.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 06/16/2009] [Accepted: 06/27/2009] [Indexed: 10/20/2022]
|
43
|
Loh XJ, Gong J, Sakuragi M, Kitajima T, Liu M, Li J, Ito Y. Surface Coating with a Thermoresponsive Copolymer for the Culture and Non-Enzymatic Recovery of Mouse Embryonic Stem Cells. Macromol Biosci 2009; 9:1069-79. [DOI: 10.1002/mabi.200900081] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Tbx5-mediated expression of Ca2+/calmodulin-dependent protein kinase II is necessary for zebrafish cardiac and pectoral fin morphogenesis. Dev Biol 2009; 330:175-84. [DOI: 10.1016/j.ydbio.2009.03.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 03/24/2009] [Accepted: 03/26/2009] [Indexed: 01/30/2023]
|
45
|
Lowry JA, Gamsjaeger R, Thong SY, Hung W, Kwan AH, Broitman-Maduro G, Matthews JM, Maduro M, Mackay JP. Structural analysis of MED-1 reveals unexpected diversity in the mechanism of DNA recognition by GATA-type zinc finger domains. J Biol Chem 2008; 284:5827-35. [PMID: 19095651 DOI: 10.1074/jbc.m808712200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MED-1 is a member of a group of divergent GATA-type zinc finger proteins recently identified in several species of Caenorhabditis. The med genes are transcriptional regulators that are involved in the specification of the mesoderm and endoderm precursor cells in nematodes. Unlike other GATA-type zinc fingers that recognize the consensus sequence (A/C/T)GATA(A/G), the MED-1 zinc finger (MED1zf) binds the larger and atypical site GTATACT(T/C)(3). We have examined the basis for this unusual DNA specificity using a range of biochemical and biophysical approaches. Most strikingly, we show that although the core of the MED1zf structure is similar to that of GATA-1, the basic tail C-terminal to the zinc finger unexpectedly adopts an alpha-helical structure upon binding DNA. This additional helix appears to contact the major groove of the DNA, making contacts that explain the extended DNA consensus sequence observed for MED1zf. Our data expand the versatility of DNA recognition by GATA-type zinc fingers and perhaps shed new light on the DNA-binding properties of mammalian GATA factors.
Collapse
Affiliation(s)
- Jason A Lowry
- School of Molecular and Microbial Biosciences, University of Sydney, New South Wales 2006, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yagi Y, Ito Y, Kuhara S, Tashiro K. Cephalic hedgehog expression is regulated directly by Sox17 in endoderm development of Xenopus laevis. Cytotechnology 2008; 57:151-9. [PMID: 19003160 DOI: 10.1007/s10616-008-9127-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Accepted: 01/18/2008] [Indexed: 11/28/2022] Open
Abstract
In early development of animals, hedgehog (Hh) genes function as morphogen in the axis determination and the organ formation. In Xenopus, three hedgehog genes, sonic (shh), banded (bhh), and cephalic (chh), were identified and might organize various tissues and organs in embryogenesis. Here, we report the spatial and temporal regulation of Xchh which is expressed in endoderm cells differentiating to digestive organs. Xchh expression in endoderm was inhibited by ectopic expression of the dominant-negative activin receptor, tAR. Moreover, a maternally inherited transcription factor VegT and its downstream regulators activated Xchh expression. These indicates that Xchh is regulated by the factor involved in the cascade originated from VegT via activin/nodal signals. Using the Sox17alpha-VP16-GR construct, we showed that Xchh expression might be induced directly by transcription factor Sox17.
Collapse
Affiliation(s)
- Yumihiko Yagi
- Graduate School of Systems Life Sciences, Kyushu University, Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | | | | | | |
Collapse
|
47
|
Chen JF, Wang S, Wu Q, Cao D, Nguyen T, Chen Y, Wang DZ. Myocardin marks the earliest cardiac gene expression and plays an important role in heart development. Anat Rec (Hoboken) 2008; 291:1200-11. [PMID: 18780304 DOI: 10.1002/ar.20756] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Myocardin belongs to the SAP domain family of transcription factors and is expressed specifically in cardiac and smooth muscle during embryogenesis and in adulthood. Myocardin functions as a transcriptional coactivator of SRF and is sufficient and necessary for smooth muscle gene expression. However, the in vivo function of myocardin during cardiogenesis is not completely understood. Here we clone myocardin from chick embryonic hearts and show that myocardin protein sequences are highly conserved cross species. Detailed studies of chick myocardin expression reveal that myocardin is expressed in cardiac and smooth muscle lineage during early embryogenesis, similar to that found in mouse. Interestingly, the expression of myocardin in the heart was found enriched in the outflow tract and the sinoatrial segments shortly after the formation of linear heart tube. Such expression pattern is also maintained in later developing embryos, suggesting that myocardin may play a unique role in the formation of those cardiac modules. Similar to its mouse counterpart, chick myocardin is able to activate cardiac and smooth muscle promoter reporter genes and induce smooth muscle gene expression in nonmuscle cells. Ectopic overexpression of myocardin enlarged the embryonic chick heart. Conversely, repression of the endogenous chick myocardin using antisense oligonucleotides or a dominant negative mutant form of myocardin inhibited cardiogenesis. Together, our data place myocardin as one of the earliest cardiac marker genes for cardiogenesis and support the idea that myocardin plays an essential role in cardiac gene expression and cardiogenesis.
Collapse
Affiliation(s)
- Jian-Fu Chen
- Carolina Cardiovascular Biology Center, Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina, 27599-7126, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Ito Y, Seno S, Nakamura H, Fukui A, Asashima M. XHAPLN3 plays a key role in cardiogenesis by maintaining the hyaluronan matrix around heart anlage. Dev Biol 2008; 319:34-45. [DOI: 10.1016/j.ydbio.2008.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 02/22/2008] [Accepted: 03/31/2008] [Indexed: 11/28/2022]
|
49
|
Hepatocyte nuclear factor 4alpha contributes to thyroid hormone homeostasis by cooperatively regulating the type 1 iodothyronine deiodinase gene with GATA4 and Kruppel-like transcription factor 9. Mol Cell Biol 2008; 28:3917-31. [PMID: 18426912 DOI: 10.1128/mcb.02154-07] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type 1 iodothyronine deiodinase (Dio1), a selenoenzyme catalyzing the bioactivation of thyroid hormone, is highly expressed in the liver. Dio1 mRNA and enzyme activity levels are markedly reduced in the livers of hepatocyte nuclear factor 4alpha (HNF4alpha)-null mice, thus accounting for its liver-specific expression. Consistent with this deficiency, serum T4 and rT3 concentrations are elevated in these mice compared with those in HNF4alpha-floxed control littermates; however, serum T3 levels are unchanged. Promoter analysis of the mouse Dio1 gene demonstrated that HNF4alpha plays a key role in the transactivation of the mouse Dio1 gene. Deletion and substitution mutation analyses demonstrated that a proximal HNF4alpha site (direct repeat 1 [TGGACAAAGGTGC]; HNF4alpha-RE) is crucial for transactivation of the mouse Dio1 gene by HNF4alpha. Mouse Dio1 is also stimulated by thyroid hormone signaling, but a direct role for thyroid hormone receptor action has not been reported. We also showed that thyroid hormone-inducible Krüppel-like factor 9 (KLF9) stimulates the mouse Dio1 promoter very efficiently through two CACCC sequences that are located on either side of HNF4alpha-RE. Furthermore, KLF9 functions together with HNF4alpha and GATA4 to synergistically activate the mouse Dio1 promoter, suggesting that Dio1 is regulated by thyroid hormone in the mouse through an indirect mechanism requiring prior KLF9 induction. In addition, we showed that physical interactions between the C-terminal zinc finger domain (Cf) of GATA4 and activation function 2 of HNF4alpha and between the basic domain adjacent to Cf of GATA4 and a C-terminal domain of KLF9 are both required for this synergistic response. Taken together, these results suggest that HNF4alpha regulates thyroid hormone homeostasis through transcriptional regulation of the mouse Dio1 gene with GATA4 and KLF9.
Collapse
|
50
|
Luzzani C, Cardillo SB, Bermúdez Moretti M, Correa García S. New insights into the regulation of the Saccharomyces cerevisiae UGA4 gene: two parallel pathways participate in carbon-regulated transcription. MICROBIOLOGY-SGM 2008; 153:3677-3684. [PMID: 17975075 DOI: 10.1099/mic.0.2007/010231-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The Saccharomyces cerevisiae UGA4 gene, which encodes the gamma-aminobutyric acid (GABA) and delta-aminolaevulinic acid (ALA) permease, is well known to be regulated by the nitrogen source. Its expression levels are low in the presence of a rich nitrogen source but are higher when a poor nitrogen source is used. In addition, GABA can induce UGA4 expression when cells are grown with proline but not when they are grown with ammonium. Although vast amounts of evidence have been gathered about UGA4 regulation by nitrogen, little is known about its regulation by the carbon source. Using glucose and acetate as rich and poor carbon source respectively, this work aimed to shed light on hitherto unclear aspects of the regulation of this gene. In poor nitrogen conditions, cells grown with acetate were found to have higher UGA4 basal expression levels than those grown with glucose, and did not show UGA4 induction in response to GABA. Analysis of the expression and subcellular localization of the transcription factors that regulate UGA4 as well as partial deletions and site-directed mutations of the UGA4 promoter region suggested that there are two parallel pathways that act in regulating this gene by the carbon source. Furthermore, the results demonstrate the existence of a new factor operating in UGA4 regulation.
Collapse
Affiliation(s)
- Carlos Luzzani
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 2. C1428EGA, Buenos Aires, Argentina
| | - Sabrina Beatriz Cardillo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 2. C1428EGA, Buenos Aires, Argentina
| | - Mariana Bermúdez Moretti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 2. C1428EGA, Buenos Aires, Argentina
| | - Susana Correa García
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 2. C1428EGA, Buenos Aires, Argentina
| |
Collapse
|