Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Publication year range
1.
Biochem Biophys Res Commun ; 609: 111-118, 2022 06 18.
Article in English | MEDLINE | ID: mdl-35429678

ABSTRACT

Although GATA5 is vital in maintaining the function of endothelial cells, the relationship between GATA5 and angiogenesis, however, remains unclear. Our study aims to determine how endothelial GATA5 mediates angiogenesis. Using the ischemic hindlimb of mice with GATA5 overexpression in the endothelia (EC-Ad mice), we showed that GATA5 overexpression could improve blood perfusion and increase capillary density. Furthermore, we showed that overexpression of GATA5 can increase the protein and mRNA levels of angiopoietin-2 (Angpt2) and fetal liver kinase 1 (Flk1) in the endothelia of EC-Ad mice, while GATA5 knockdown can inhibit the VEGF-165-induced proliferation, tube formation, and migration of human umbilical vein endothelial cells (HUVECs). In addition, we observed a decrease in the Angpt2 and Flk1, and the matrix metalloproteinase (MMP) family proteins: MMP2 and MMP9 while GATA5 was decreased. Meanwhile, our study also demonstrated that the expression of cathepsin S (Cat S) decreases when GATA5 is downregulated. Immunoprecipitation assay indicated that GATA5 could bind to Cat S directly. Furthermore, GATA5 or Cat S overexpression can promote tube formation and migration of HUVECs, restore the Angpt2 and Flk1 expression levels in the GATA5 knockdown HUVECs, and upregulate MMP2 and MMP9 protein levels. In summary, our study demonstrated that endothelial GATA5 could mediate angiogenesis by inducing the expression of Cat S, which mediates the Angpt2/Flk1 and MMP2/9 signaling pathways.


Subject(s)
Angiopoietin-2 , Vascular Endothelial Growth Factor Receptor-2 , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Cathepsins , GATA5 Transcription Factor/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
J Hum Genet ; 67(9): 515-518, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35534675

ABSTRACT

Bicuspid aortic valve (BAV) is the most common congenital heart defect with a high index of heritability. Patients with BAV have different clinical courses and disease progression. Herein, we report three siblings with BAV and clinical differences. Their clinical presentations include moderate to severe aortic regurgitation, aortic stenosis, and ascending aortic aneurysm. Genetic investigation was carried out using Whole-Exome Sequencing for the three patients. We identified two non-synonymous variants in ROBO1 and GATA5 genes. The ROBO1: p.(Ser327Pro) variant is shared by the three BAV-affected siblings. The GATA5: p.(Gln3Arg) variant is shared only by the two brothers who presented BAV and ascending aortic aneurysm. Their sister, affected by BAV without aneurysm, does not harbor the GATA5: p.(Gln3Arg) variant. Both variants were absent in the patients' fourth brother who is clinically healthy with tricuspid aortic valve. To our knowledge, this is the first association of ROBO1 and GATA5 variants in familial BAV with a potential genotype-phenotype correlation. Our findings are suggestive of the implication of ROBO1 gene in BAV and the GATA5: p.(Gln3Arg) variant in ascending aortic aneurysm. Our family-based study further confirms the intrafamilial incomplete penetrance of BAV and the complex pattern of inheritance of the disease.


Subject(s)
Bicuspid Aortic Valve Disease , GATA5 Transcription Factor , Nerve Tissue Proteins , Receptors, Immunologic , Aortic Valve/abnormalities , Bicuspid Aortic Valve Disease/genetics , Female , GATA5 Transcription Factor/genetics , Humans , Male , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Roundabout Proteins
3.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563646

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.


Subject(s)
GATA Transcription Factors , GATA4 Transcription Factor , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Heart , Myocytes, Cardiac/metabolism
4.
Circ Res ; 124(10): 1448-1461, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30894089

ABSTRACT

RATIONALE: Endothelial dysfunction is an important determinant risk factor for the development of hypertension and its complications. Thus, identification of potential therapeutic targets for preventing endothelial dysfunction has major clinical importance. Emerging evidence indicates that epigenetic modifications are closely associated with the regulation of endothelial function. Among them, HDAC (histone deacetylase)-mediated epigenetic processes in vascular homeostasis and cardiovascular disease have attracted much attention. SIRT6 (sirtuin 6) is one member of SIRTs (class III HDAC) that are highly conserved NAD+-dependent deacetylases. OBJECTIVE: This study was designed to elucidate the role of SIRT6 in the pathogenesis of hypertension, discover the new targets of SIRT6, and explore related mechanisms on the regulation of endothelial function. METHODS AND RESULTS: The levels of endothelial SIRT6 were significantly reduced in 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Utilizing genetically engineered endothelial-specific SIRT6 knockout (Cre+/SIRT6fl/fl) mice, we found that endothelial-specific deletion of SIRT6 significantly enhanced blood pressure, exacerbated endothelial dysfunction and cardiorenal injury in experimental hypertension. Functionally, SIRT6 has pleiotropic protective actions in endothelial cells, which include promoting endothelium-dependent vasodilatation and vascular NO bioavailability, reducing cellular permeability, ameliorating endothelial senescence and apoptosis, and facilitating autophagy. Mechanistically, SIRT6 induced the expression of GATA5 (GATA-binding protein 5), a novel regulator of blood pressure, through inhibiting Nkx3.2 (NK3 homeobox 2) transcription by deacetylating histone H3K9 (histone H3 lysine 9), thereby regulating GATA5-mediated signaling pathways to prevent endothelial injury. Finally, we provide direct evidence for the therapeutic potential of SIRT6 in desoxycorticosterone acetate/salt-induced hypertensive mice by overexpression of SIRT6 in vivo. CONCLUSIONS: This study for the first time demonstrates that SIRT6 prevents hypertension and its complications by maintaining endothelial function. Pharmacological targeting of SIRT6 may be an innovative therapeutic strategy for treating patients with hypertension.


Subject(s)
Endothelium, Vascular/physiology , Hypertension/prevention & control , Sirtuins/physiology , Acetylation , Angiotensin II , Animals , Desoxycorticosterone Acetate , Endothelium, Vascular/injuries , Epigenesis, Genetic , GATA5 Transcription Factor/metabolism , Histone Deacetylases , Histones/metabolism , Homeodomain Proteins/metabolism , Hypertension/chemically induced , Hypertension, Renal/metabolism , Kidney/injuries , Mice , Mice, Knockout , Nephritis/metabolism , Sirtuins/blood , Sirtuins/deficiency , Sirtuins/genetics , Sodium Chloride , Transcription Factors/metabolism , Vasoconstrictor Agents , Vasodilation
5.
J Biol Chem ; 294(8): 2732-2743, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30593510

ABSTRACT

Zebrafish gata4/5/6 genes encode transcription factors that lie on the apex of the regulatory hierarchy in primitive myelopoiesis. However, little is known about the roles of microRNAs in gata4/5/6-regulated processes. Performing RNA-Seq deep sequencing analysis of the expression changes of microRNAs in gata4/5/6-knockdown embryos, we identified miR-210-5p as a regulator of zebrafish primitive myelopoiesis. Knocking down gata4/5/6 (generating gata5/6 morphants) significantly increased miR-210-5p expression, whereas gata4/5/6 overexpression greatly reduced its expression. Consistent with inhibited primitive myelopoiesis in the gata5/6 morphants, miR-210-5p overexpression repressed primitive myelopoiesis, indicated by reduced numbers of granulocytes and macrophages. Moreover, knocking out miR-210 partially rescued the defective primitive myelopoiesis in zebrafish gata4/5/6-knockdown embryos. Furthermore, we show that the restrictive role of miR-210-5p in zebrafish primitive myelopoiesis is due to impaired differentiation of hemangioblast into myeloid progenitor cells. By comparing the set of genes with reduced expression levels in the gata5/6 morphants to the predicted target genes of miR-210-5p, we found that foxj1b and slc3a2a, encoding a forkhead box transcription factor and a solute carrier family 3 protein, respectively, are two direct downstream targets of miR-210-5p that mediate its inhibitory roles in zebrafish primitive myelopoiesis. In summary, our results reveal that miR-210-5p has an important role in the genetic network controlling zebrafish primitive myelopoiesis.


Subject(s)
Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental , Gene Silencing , MicroRNAs/genetics , Myelopoiesis , RNA, Messenger/antagonists & inhibitors , Zebrafish Proteins/antagonists & inhibitors , Zebrafish/embryology , Animals , Embryo, Nonmammalian/metabolism , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Fusion Regulatory Protein 1, Heavy Chain/antagonists & inhibitors , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , GATA Transcription Factors/antagonists & inhibitors , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , GATA5 Transcription Factor/antagonists & inhibitors , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Gene Regulatory Networks , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Rev Cardiovasc Med ; 21(2): 253-261, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32706213

ABSTRACT

It is known that functional defects of GATA binding protein 5 (GATA5), an important member of GATA transcription factor family, could cause multiple congenital defects. However, the mechanisms of this transcription factor in cardiovascular diseases are still little known. Finding a genetic approach should help with understanding the possible roles of GATA5 in different cardiovascular diseases and purpose it as a possible therapeutic agent. Hence, this review is divided into three chapters to summarize the roles and main regulatory mechanisms of GATA5 in hypertension, arrhythmia and congenital heart disease, respectively. In each chapter, this review firstly introduces the roles of GATA5 mutations, and then discusses the main regulatory mechanisms of GATA5 in the corresponding diseases (Such as the endothelial dysfunction signaling pathway in the chapter of hypertension, GATA5-NaV1.5 signaling pathway in the chapter of arrhythmia, GATA5-HEY2 and GATA5-Nodal signaling pathway in the chapter of congenital heart disease). Additionally, based on these regulatory networks, it is also speculated that abnormal methylation of the GATA5 gene promoter may lead to cardiovascular diseases such as congenital heart disease. This conjecture is proposed to enrich the regulatory networks of GATA5 and provide a theoretical basis for diagnosis and treatment of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , GATA5 Transcription Factor/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Cardiovascular System/physiopathology , GATA5 Transcription Factor/genetics , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction
7.
J Cell Mol Med ; 23(4): 2536-2548, 2019 04.
Article in English | MEDLINE | ID: mdl-30672133

ABSTRACT

Evidence indicated that GATA5 may suppress hepatocellular carcinoma (HCC) cell malignant transformation, but the mechanism of how GATA5 affects cancer cell reprogramming to inhibit HCC malignant behaviour is still unclear. In this study, we report that the expression of ß-catenin and reprogramming genes p-Oct4, Nanog, Klf4, c-myc and EpCAM was significantly higher in HCC tissues compared to normal liver tissues. In contrast, the expression of GATA5 was significantly lower in HCC tissues compared to normal liver tissues. Transfection of CDH-GATA5 vectors into HCC cells (HLE, Bel 7402 and PLC/PRF/5 cells) increased the GATA5 expression and decreased the expression of ß-catenin and reprogramming genes p-Oct4, Nanog, Klf4, c-myc and EpCAM. Increased GATA5 expression by transfection with its expression vectors was also able to inhibit the cell growth, colony formation and capability of migration, invasion, while promoting apoptosis in HCC cells. Results revealed that GATA5 co-localization with ß-catenin in the cytoplasm, preventing ß-catenin from entering the nucleus. Treatment with the specific Wnt/ß-catenin pathway inhibitor salinomycin was able to reduce the expression of ß-catenin and reprogramming genes. Salinomycin exerted a similar influence as GATA5, and siRNA-GATA5 restored ß-catenin and reprogramming gene expression. This study demonstrates that an increase in the expression of GATA5 inhibits the expression of ß-catenin and reprogramming genes and suppresses tumour growth, colony formation, metastasis and invasion, while promoting apoptosis in HCC cells. The mechanism of GATA5 inhibiting the malignant behaviours of HCC cells may involve in the disruption of the Wnt/ß-catenin pathway and the reduction of reprogramming gene expression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , GATA5 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , beta Catenin/genetics , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Case-Control Studies , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Female , GATA5 Transcription Factor/antagonists & inhibitors , GATA5 Transcription Factor/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrans/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
8.
Eur J Clin Invest ; 48(12): e13027, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30229885

ABSTRACT

BACKGROUND: Bicuspid aortic valve (BAV) is a heterogeneous and still not fully understood condition, with diverse genetic aetiology and associated phenotypes ranging from aortic stenosis or regurgitation to aneurysm and dissection. Several genes have been associated with the presence of BAV, notably some members of the GATA family of transcription factors that play important roles in cardiac embryogenesis. METHODS: A case-control study with 122 unrelated and ethnically matched patients with bicuspid and 154 with tricuspid aortic valve was performed. All exons of GATA4, GATA5, and GATA6 genes were sequenced searching for variants. Frequencies were compared and functional effects of rare variants were analysed by informatic prediction tools. RESULTS: Four rare and potentially pathogenic variants were identified in only five sporadic cases: a missense p.Arg202Gln (rs782614097) in GATA5 and three synonymous variants, p.Cys274= (rs55980825) and p.His302= (rs201516339) in GATA4, and p.Asn458= (rs143026087) in GATA6. Minor alleles of p.His302=, p.Arg202Gln and rs3764962 are enriched in BAV patients compared to ExAC database (P = 0.01, 0.03, and 0.01 respectively). In addition, a common polymorphism in GATA5 (p.Asp203=, rs41305803) is associated with BAV showing a protective effect in carriers of the minor allele (OR [95%CI] = 0.45[0.25-0.81]; P = 0.004). CONCLUSION: This study associates additional genetic variants in GATA4 and GATA5 with BAV, supporting the implication of these genes in the development of this valvulopathy. The discovery of all the genetic factors involved will contribute to a better understanding of the process and, therefore, to detect a genetic predisposition and even to the identification of therapeutic targets.


Subject(s)
Aortic Valve/abnormalities , GATA4 Transcription Factor/genetics , GATA5 Transcription Factor/genetics , Heart Valve Diseases/genetics , Aged , Bicuspid Aortic Valve Disease , Case-Control Studies , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genotype , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics
9.
Dig Dis Sci ; 63(11): 2889-2897, 2018 11.
Article in English | MEDLINE | ID: mdl-30083861

ABSTRACT

BACKGROUND: GATA factors, which constitute a family of transcription regulatory proteins, participate in gastrointestinal development. Trefoil factor 1 (TFF1) plays a crucial role in mucosal defense and healing, and evidence suggests that GATA-5 mediated its regulation. Gastric cancer is a multiple-step process triggered by Helicobacter pylori and is characterized by accumulation of molecular and epigenetic alteration. The aim of this study was to evaluate the effect of H. pylori infection on the regulation of GATA-5 and TFF1 in vitro and in vivo. RESULTS: Infected cells exhibited upregulation of GATA-5 and TFF1 after 48 h. An increase in GATA-5 and TFF1 mRNA levels was also found in mice samples after 6 and 12 months of infection, respectively. In human samples, we found an association between H. pylori infection and GATA-5 upregulation. In fact, among H. pylori-infected patients, hypermethylation was observed in 45.5% of pediatric samples, in 62.6% of chronic gastritis samples, and in 63% of gastric cancer samples. Regarding TFF1, the expression levels were similar in pediatrics and adults patients, and were independent of H. pylori infection, and the expression of these factors was downregulated in gastric cancer samples. GATA-5 promoter methylation was associated with a decrease in TFF1 mRNA levels. CONCLUSIONS: Our results suggest that the upregulation of GATA-5 and TFF1 observed in vitro and in vivo may be correlated with a protective effect of the mucosa in response to infection. The epigenetic inactivation of GATA-5 observed in human biopsies from infected patients may suggest that this alteration is an early event occurring in association with H. pylori infection.


Subject(s)
GATA5 Transcription Factor/metabolism , Gastritis/metabolism , Helicobacter Infections/metabolism , Stomach Neoplasms/metabolism , Trefoil Factor-1/metabolism , Adult , Aged , Animals , Child , Child, Preschool , DNA Methylation , Epithelial Cells/metabolism , Female , Gastritis/microbiology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Promoter Regions, Genetic , Stomach Neoplasms/microbiology , Young Adult
10.
J Mol Cell Cardiol ; 102: 74-82, 2017 01.
Article in English | MEDLINE | ID: mdl-27894866

ABSTRACT

Aberrant expression of the sodium channel gene (SCN5A) has been proposed to disrupt cardiac action potential and cause human cardiac arrhythmias, but the mechanisms of SCN5A gene regulation and dysregulation still remain largely unexplored. To gain insight into the transcriptional regulatory networks of SCN5A, we surveyed the promoter and first intronic regions of the SCN5A gene, predicting the presence of several binding sites for GATA transcription factors (TFs). Consistent with this prediction, chromatin immunoprecipitation (ChIP) and sequential ChIP (Re-ChIP) assays show co-occupancy of cardiac GATA TFs GATA4 and GATA5 on promoter and intron 1 SCN5A regions in fresh-frozen human left ventricle samples. Gene reporter experiments show GATA4 and GATA5 synergism in the activation of the SCN5A promoter, and its dependence on predicted GATA binding sites. GATA4 and GATA6 mRNAs are robustly expressed in fresh-frozen human left ventricle samples as measured by highly sensitive droplet digital PCR (ddPCR). GATA5 mRNA is marginally but still clearly detected in the same samples. Importantly, GATA4 mRNA levels are strongly and positively correlated with SCN5A transcript levels in the human heart. Together, our findings uncover a novel mechanism of GATA TFs in the regulation of the SCN5A gene in human heart tissue. Our studies suggest that GATA5 but especially GATA4 are main contributors to SCN5A gene expression, thus providing a new paradigm of SCN5A expression regulation that may shed new light into the understanding of cardiac disease.


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Transcription, Genetic , Animals , Binding Sites , Cell Line , GATA5 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Mutation , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats
11.
Dev Biol ; 418(1): 17-27, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27554166

ABSTRACT

The Mesp family of transcription factors have been implicated in the early formation and migration of the cardiac lineage, although the precise molecular mechanisms underlying this process remain unknown. In this study we examine the function of Mesp family members in zebrafish cardiac development and find that Mespaa is remarkably efficient at promoting cardiac fates in normally non-cardiogenic cells. However, Mespaa is dispensable for normal cardiac formation. Despite no overt defects in cardiovascular specification, we find a consistent defect in cardiac laterality in mespaa null embryos. This is further exacerbated by the depletion of other mesp paralogues, highlighting a conserved role for the mesp family in left-right asymmetry, distinct from a function in cardiac specification. Despite an early requirement for mespaa to promote cardiogenesis, cells over-expressing mespaa are found to both exhibit unique cellular behaviors and activate the transcription of gata5 only after the completion of gastrulation. We propose that while mespaa remains capable of driving cardiac progenitor formation in zebrafish, it may not play an essential role in the cardiac regulatory network. Furthermore, the late activation of migration and cardiac gene transcription in mespaa over-expressing cells challenges previous studies on the timing of these events and provides intriguing questions for future study.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Heart/embryology , Myocytes, Cardiac/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning/genetics , Cell Differentiation , GATA5 Transcription Factor/biosynthesis , GATA5 Transcription Factor/genetics , Gastrulation/physiology , Morpholinos/genetics , Zebrafish Proteins/biosynthesis
12.
Dev Biol ; 418(1): 179-188, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27542689

ABSTRACT

The cellular turnover of adult tissues and injury-induced repair proceed through an exquisite integration of proliferation, differentiation, and survival signals that involve stem/progenitor cell populations, their progeny, and differentiated tissues. GATA factors are DNA binding proteins that control stem cells and the development of tissues by activating or repressing transcription. Here we examined the role of GATA transcription factors in Schmidtea mediterranea, a freshwater planarian that provides an excellent model to investigate gene function in adult stem cells, regeneration, and differentiation. Smed-gata4/5/6, the homolog of the three mammalian GATA-4,-5,-6 factors is expressed at high levels in differentiated gut cells but also at lower levels in neoblast populations, the planarian stem cells. Smed-gata4/5/6 knock-down results in broad differentiation defects, especially in response to injury. These defects are not restricted to the intestinal lineage. In particular, at late time points during the response to injury, loss of Smed-gata4/5/6 leads to decreased neoblast proliferation and to gene expression changes in several neoblast subpopulations. Thus, Smed-gata4/5/6 plays a key evolutionary conserved role in intestinal differentiation in planarians. These data further support a model in which defects in the intestinal lineage can indirectly affect other differentiation pathways in planarians.


Subject(s)
GATA4 Transcription Factor/genetics , GATA5 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Intestines/cytology , Planarians/embryology , Regeneration/genetics , Regeneration/physiology , Stem Cells/cytology , Animals , Cell Proliferation/genetics , GATA4 Transcription Factor/biosynthesis , GATA5 Transcription Factor/biosynthesis , GATA6 Transcription Factor/biosynthesis , Intestinal Mucosa/metabolism , Planarians/genetics , RNA Interference , RNA, Small Interfering/genetics
13.
Hum Genet ; 136(3): 339-346, 2017 03.
Article in English | MEDLINE | ID: mdl-28180938

ABSTRACT

GATA5 belongs to the GATA family of transcription factors characterized by highly evolutionarily conserved zinc-finger DNA-binding domains. Mouse models have implicated a role of GATA5 during mammalian embryogenesis, including proper heart development and gender-specific regulation of female genitourinary tract formation. Previous studies have found an association of heterozygous missense alterations in GATA5 with a broad variety of heart diseases; however, the clinical relevance of the identified susceptibility variants has remained unclear. Here, we report on a girl with hydrops fetalis, congenital heart defects, clitoromegaly and postnatally increased 17-hydroxyprogesterone levels. By trio whole-exome sequencing, we identified compound heterozygous missense mutations, p.Ser19Trp and p.Arg202Gln, in GATA5 as putative disease-causing alterations. The identified mutations fail to rescue the cardia bifida phenotype in a zebrafish model, mislocalize to subnuclear foci when transiently transfected in HEK293 cells and possess less transcriptional activity. In addition to demonstrating the pathogenicity of identified mutations, our findings show that GATA5 mutations, in addition to heart diseases, can result in congenital abnormalities of the female genitourinary tract in humans.


Subject(s)
GATA5 Transcription Factor/genetics , Genitalia, Female/abnormalities , Heart Defects, Congenital/genetics , Heterozygote , Hydrops Fetalis/genetics , Mutation , Animals , Female , HEK293 Cells , Heart/embryology , Humans , Infant, Newborn , Male , Pedigree , Zebrafish/embryology
14.
Development ; 141(20): 3978-87, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25294942

ABSTRACT

The relative timing of SHH and BMP signals controls whether presomitic mesoderm (PSM) cells will adopt either a chondrogenic or lateral plate mesoderm fate. Here we document that SHH-mediated induction of Nkx3.2 maintains the competence of somitic cells to initiate chondrogenesis in response to subsequent BMP signals by repressing BMP-dependent induction of GATA genes. Conversely, administration of BMP signals to PSM or forced expression of GATA family members in chick PSM explants blocks induction of hedgehog-dependent gene expression. We demonstrate that GATA factors can interact with Gli factors and can recruit the transcriptional co-factor FOG1 (ZFPM1) to the regulatory region of the mouse Gli1 gene, repressing the induction of Gli1 by SHH by binding to both GATA and Gli binding sites. Knockdown of FOG1 reverses the ability of GATA factors to repress Gli1 expression. Our findings uncover a novel role for GATA transcription factors as repressors of hedgehog signaling, and document that NKX3.2 maintains the ability of sclerotomal cells to express SHH transcriptional targets in the presence of BMP signals by repressing the induction of Gata4/5/6.


Subject(s)
Bone Morphogenetic Proteins/metabolism , GATA4 Transcription Factor/metabolism , GATA5 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chondrocytes/cytology , Gene Expression Profiling , Kruppel-Like Transcription Factors/metabolism , Mice , NIH 3T3 Cells , Nuclear Proteins/metabolism , Zinc Finger Protein GLI1
15.
Biochim Biophys Acta Gen Subj ; 1861(7): 1691-1701, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28285006

ABSTRACT

BACKGROUND: SUMOylation is a critical regulatory protein modification in eukaryotic cells and plays a pivotal role in cardiac development and disease. Several cardiac transcription factors are modified by SUMO, but little is known about the impact of SUMOylation on their function during cardiac development. METHODS: We used a zebrafish model to address the impact of SUMOylation on GATA5, an essential transcription factor in zebrafish cardiac development. GATA5 SUMOylation was probed by western blot, the subcellular localization and transcriptional activity of GATA5 mutants were examined by immunostaining and luciferase reporter assay. The in vivo function of GATA5 SUMOylation was evaluated by gata5 mutants mRNA microinjection and in situ hybridization in gata5 morphants and ubc9 mutants. RESULTS: Firstly, we identified GATA5 as a SUMO substrate, and lysine 324 (K324) and lysine 360 (K360) as two major modification sites. Conversion of lysine to arginine at these two sites did not affect subcellular localization, but did affect the transcriptional activity of GATA5. Secondly, in vivo experiments demonstrated that the wild type (WT) and K324R mutant of gata5 could rescue impaired cardiac precursor differentiation, while the K360R mutant of gata5 drastically lost this potency in gata5 morphant. Furthermore, in SUMOylation-deficient ubc9 mutants, the abnormal expression pattern displayed by the early markers of cardiac development (nkx2.5 and mef2cb) could be restored using a sumo-gata5 fusion, but not with a WT gata5. CONCLUSION: GATA5 SUMOylation is indispensable for early zebrafish cardiac development. GENERAL SIGNIFICANCE: Our studies highlight the potential importance of transcription factor SUMOylation in cardiac development.


Subject(s)
GATA5 Transcription Factor/physiology , Heart/embryology , Sumoylation , Zebrafish/embryology , Active Transport, Cell Nucleus , Animals , Cell Differentiation , Cells, Cultured
16.
Dev Biol ; 408(1): 56-65, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26460096

ABSTRACT

Pten is a multifunctional tumor suppressor. Deletions and mutations in the Pten gene have been associated with multiple forms of human cancers. Pten is a central regulator of several signaling pathways that influences multiple cellular functions. One such function is in cell motility and migration, although the precise mechanism remains unknown. In this study, we deleted Pten in the embryonic lung epithelium using Gata5-cre mice. Absence of Pten blocked branching morphogenesis and ERK and AKT phosphorylation at E12.5. In an explant model, Pten(Δ/Δ) mesenchyme-free embryonic lung endoderm failed to branch. Inhibition of budding in Pten(Δ/Δ) explants was associated with major changes in cell migration, while cell proliferation was not affected. We further examined the role of ERK and AKT in branching morphogenesis by conditional, endodermal-specific mutants which blocked ERK or AKT phosphorylation. MEK(DM/+); Gata5-cre (blocking of ERK phosphorylation) lung showed more severe phenotype in branching morphogenesis. The inhibition of budding was also associated with disruption of cell migration. Thus, the mechanisms by which Pten is required for early endodermal morphogenesis may involve ERK, but not AKT, mediated cell migration.


Subject(s)
Endoderm/embryology , Endoderm/enzymology , Lung/embryology , MAP Kinase Signaling System , Morphogenesis , PTEN Phosphohydrolase/metabolism , Animals , Cell Movement , Epithelium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GATA5 Transcription Factor/metabolism , Gene Deletion , Integrases/metabolism , Mice , Models, Biological , Organ Specificity , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
17.
Hum Genet ; 135(8): 895-906, 2016 08.
Article in English | MEDLINE | ID: mdl-27225266

ABSTRACT

Gastric cancer incidence varies considerably among populations, even those with comparable rates of Helicobacter pylori infection. To test the hypothesis that genetic variation plays a role in gastric disease, we assessed the relationship between genotypes and gastric histopathology in a Colombian study population, using a genotyping array of immune-related single nucleotide polymorphisms (SNPs). Two synonymous SNPs (rs6061243 and rs6587239) were associated with progression of premalignant gastric lesions in a dominant-effects model after correction for multiple comparisons (p = 2.63E-07 and p = 7.97E-07, respectively); effect sizes were ß = -0.863 and ß = -0.815, respectively, where ß is an estimate of effect on histopathology scores, which ranged from 1 (normal) to 5 (dysplasia). In our replication cohort, a second Colombian population, both SNPs were associated with histopathology when additively modeled (ß = -0.256, 95 % CI = -0.47, -0.039; and ß = -0.239, 95 % CI = -0.45, -0.024), and rs6587239 was significantly associated in a dominant-effects model (ß = -0.330, 95 % CI = -0.66, 0.00). Because promoter methylation of GATA5 has previously been associated with gastric cancer, we also tested for the association of methylation status with more advanced histopathology scores in our samples and found a significant relationship (p = 0.001). A multivariate regression model revealed that the effects of both the promoter methylation and the exonic SNPs in GATA5 were independent. A SNP-by-methylation interaction term was also significant. This interaction between GATA5 variants and GATA5 promoter methylation indicates that the association of either factor with gastric disease progression is modified by the other.


Subject(s)
DNA Methylation/genetics , Epigenomics , GATA5 Transcription Factor/genetics , Helicobacter Infections/genetics , Stomach Neoplasms/genetics , Adult , Female , Genetic Association Studies , Genotype , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Risk Factors , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
18.
Development ; 140(18): 3787-98, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23903195

ABSTRACT

Biphasic control of WNT signaling is essential during cardiogenesis, but how the pathway switches from promoting cardiac mesoderm to restricting cardiomyocyte progenitor fate is unknown. We identified genes expressed in lateral mesoderm that are dysregulated in zebrafish when both gata5 and gata6 are depleted, causing a block to cardiomyocyte specification. This screen identified tmem88a, which is expressed in the early cardiac progenitor field and was previously implicated in WNT modulation by overexpression studies. Depletion of tmem88a results in a profound cardiomyopathy, secondary to impaired cardiomyocyte specification. In tmem88a morphants, activation of the WNT pathway exacerbates the cardiomyocyte deficiency, whereas WNT inhibition rescues progenitor cells and cardiogenesis. We conclude that specification of cardiac fate downstream of gata5/6 involves activation of the tmem88a gene to constrain WNT signaling and expand the number of cardiac progenitors. Tmem88a is a novel component of the regulatory mechanism controlling the second phase of biphasic WNT activity essential for embryonic cardiogenesis.


Subject(s)
Body Patterning , GATA Transcription Factors/metabolism , GATA5 Transcription Factor/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Stem Cells/metabolism , Wnt Signaling Pathway , Zebrafish Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cardiomyopathies/embryology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Genetic Association Studies , Membrane Proteins/genetics , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/metabolism , Morpholinos/pharmacology , Mutation/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organogenesis/drug effects , Organogenesis/genetics , Phenotype , RNA Transport/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
20.
Clin Chem Lab Med ; 54(12): 1971-1980, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27171388

ABSTRACT

BACKGROUND: Aberrant hypermethylation of tumour suppressor genes (TSGs) occurring in hepatocellular carcinoma (HCC) could provide a mean of molecular characterisation of this cancer. The aim of this study was to investigate promoter methylation and gene expression of selected TSGs in HCC to identify candidate genes for further validation as potential biomarkers. METHODS: Methylation-specific multiplex ligation-dependent probe amplification method was used to measure the methylation status of 25 TSGs in 49 HCC samples and 36 corresponding non-cancerous liver tissue samples. Relative expression of the differentially methylated genes was assessed at the mRNA level using quantitative PCR. RESULTS: We observed a significantly higher methylation in genes WT1, PAX5, PAX6, PYCARD and GATA5 in HCC compared with control samples. The expression of PAX5 was significantly decreased by methylation; conversely methylation of WT1 was associated with higher mRNA levels. Methylation of GATA5 was significantly associated with overall survival and methylation of WT1 and PAX5 significantly varied between patients with ALBI score 1 vs. 2+3. Moreover, PAX5 was significantly more methylated in patients with tumour grade 2+3 vs. grade 1, and methylation of the PAX5 correlated with the patient's age at the time of diagnosis. CONCLUSIONS: HCC evince aberrant promoter methylation of WT1, PAX5, PAX6, PYCARD and GATA5 genes. Correlation between GATA5, WT1 and PAX5 methylation and clinical/histological parameters is suggestive of applicability of these markers in non-invasive (epi)genetic testing in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Methylation , GATA5 Transcription Factor/genetics , Genes, Tumor Suppressor , Liver Neoplasms/genetics , PAX5 Transcription Factor/genetics , WT1 Proteins/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL