IMR Press / RCM / Volume 21 / Issue 2 / DOI: 10.31083/j.rcm.2020.02.5104
Open Access Review
Potential roles of GATA binding protein 5 in cardiovascular diseases
Show Less
1 Department of Medicine, Shandong University School of Medicine, Jinan, Shandong, 250014, P. R. China
2 The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, P. R. China
3 Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, P. R. China
4 Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, P. R. China
*Correspondence: yanbo@mail.jnmc.edu.cn (Bo Yan)
Rev. Cardiovasc. Med. 2020, 21(2), 253–261; https://doi.org/10.31083/j.rcm.2020.02.5104
Submitted: 10 December 2019 | Revised: 20 March 2020 | Accepted: 30 March 2020 | Published: 30 June 2020
Copyright: © 2020 Song and Yan Published by IMR Press.
This is an open access article under the CC BY 4.0 license http://creativecommons.org/licenses/by/4.0/.
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.

Keywords
GATA5
hypertension
arrhythmia
congenital heart disease
1. Introduction

Rapid economic development has promoted the transformation of the human disease model from communicable disease to non-communicable disease (Du et al., 2019). Cardiovascular disease and cancer are the two most common chronic non-communicable human diseases and are often interdependent (Regulska et al., 2019). Coronary heart disease is one of the major cardiovascular diseases affecting the global population and has been shown to be the leading cause of death in both developed and developing countries (Malakar et al., 2019). In China, the prevalence and mortality of cardiovascular diseases have increased significantly in the past 20 years and are likely to continue to do so into the future (Shen and Ge, 2018). Therefore, it is imperative to further actively explore the mechanism of cardiovascular diseases to stem the ongoing expansion of the cardiovascular diseases burden.

The GATA transcription factor family is divided into two subfamilies GATA1/2/3 and GATA4/5/6. GATA1/2/3 is necessary for the differentiation of mesoderm and ectoderm (including the central nervous and hematopoiesis systems). GATA4/5/6 is involved in the differentiation and development of endoderm and mesoderm, including cardiovascular embryogenesis (Fujiwara, 2017; Lentjes et al., 2016). In vertebrates, each of the six GATA proteins has two zinc finger DNA binding domains, Cys-X2-C-X17-Cys-X2-Cys (ZNI and ZNII), which recognize the sequences (A/T)GATA(A/G) (Lentjes et al., 2016).

GATA binding protein 5 (GATA5) is an important member of the GATA family and regulates many biological processes, such as cell metabolism (Bruun et al., 2014; Zibrova et al., 2017), coronary artery development (Wang et al., 2017; Ying et al., 2017), vascular inflammation, oxidative stress and endothelial function (Messaoudi et al., 2015; Wang et al., 2010) by affecting the expression of bone morphogenic protein (BMP)-4, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), Friend of GATA-2 (FOG-2) and other cytokines. GATA5 deficiency decreases the activity of AMPK and up-regulates the expression of BMP-4, interleukin-6 and intercellular cell adhesion molecule-1 (ICAM-1) (Messaoudi et al., 2015). AMPK promotes the regeneration of coronary artery and exerts a cardioprotective effect under stress conditions such as myocardial ischemia/hypoxia and ischemia/reperfusion (Wang et al., 2017; Zibrova et al., 2017). However, the functional defect of GATA5 decreases AMPK activity (Messaoudi et al., 2015), which weakens the protective effect of AMPK on myocardial cells. This affects the occurrence and development of cardiovascular diseases (Wang et al., 2010). BMP-4 is the earliest marker of atherosclerosis and is considered to be a mechanically sensitive autocrine cytokine that plays an important role in promoting inflammation, atherosclerosis and hypertension (Hong et al., 2016). GATA5 deficiency can lead to hypertension by inducing the production of BMP-4. (Hong et al., 2016; Koga et al., 2013; Messaoudi et al., 2015; Zhang et al., 2014). Therefore, altered GATA5 levels may interfere with the cardiac gene regulatory networks, increasing susceptibility to cardiovascular diseases. Animal experiments and human studies have shown that GATA5 plays an indispensable role in the occurrence and development of cardiovascular diseases such as congenital heart disease, atrial fibrillation and hypertension (Lentjes et al., 2016; Messaoudi et al., 2015; Shi et al., 2014; Wang et al., 2013).

Finding a genetic approach should help with understanding the possible role of GATA5 in different cardiovascular diseases and purpose it as a possible therapeutic agent. Therefore, this review summarizes the roles and main molecular mechanisms of GATA5 in the occurrence and development of hypertension, arrhythmia and congenital heart disease (CHD) (Fig.1). Additionally, based on these regulatory networks, a hypothesis is proposed as a basis for further research.

Figure 1.

The summary figure of GATA5. GATA5, GATA binding protein 5; CHD, congenital heart disease; SIRT6, Sirtuin 6; Nav1. 5, voltage-gated sodium channel 1. 5; HEY2, Hes-related family basic helix-loop-helix (bHLH) transcription factor with YRPW motif 2; Nodal, Nodal growth differentiation factor; PKA, protein kinase A; AMPK, adenosine 5'-monophosphate (AMP)-activated protein kinase; NF-κB, nuclear factor κB.

2. GATA5 and cardiovascular diseases
2.1 Hypertension

Hypertension, with 31 % to 68 % heritability and a global prevalence rate of 25 % to 30 %, is a complex disease determined by both environmental factors and genetic factors (Messaoudi et al., 2015). Hypertension is considered to be a major risk factor for stroke, chronic kidney disease and coronary heart disease, as well as a leading cause of global morbidity and mortality (Guo et al., 2019). Although the findings from the three consortia, Global BP Genetics(GLOBAL BPgen), Cohorts for Heart and Aging Research Genome Epidemiology(CHARGE) and Asian Genetic Epidemiology Network-Blood Pressure (AGEN-BP), represented an important advance in hypertension research, the collective effect of all identified hypertension genes explains only a small fraction (2%) of hypertension heritability (Messaoudi et al., 2015).

The stability of blood pressure is regulated by multiple physiological systems (endocrine, sympathetic, cardiovascular and kidney) (Rafii et al., 2016). GATA5 may balance blood pressure in the kidney and possibly arterial vessels in other organs (Barry et al., 2019). The dysfunction of GATA5 in endothelial cells disrupts this balance by interfering with multiple systems involved in the regulation of blood pressure. Firstly, the absence of GATA5 in endothelial cells promotes renal inflammation and glomerular lesions, which may contribute to the development of hypertension (Messaoudi et al., 2015). Although the mechanism is not clear, it may be related to the secretion of cytokines such as BMP-4 (Tominaga et al., 2011) or the alteration of podocyte mediated by endothelial cells (Daehn et al., 2014; Jefferson and Shankland, 2014). Secondly, in mice, lack of GATA5 can lead to elevated blood pressure, age-dependent terminal organ damage and endothelial dysfunction, which are all characteristics of human hypertension (Messaoudi et al., 2015). Finally, the variations (rs6061245 and rs6587239) of GATA5 sequence are significantly associated with anti-hypertensive medication (Table 1). Rs6587239 is located in the region rich in splicing regulatory sites, which may affect the RNA splicing and the expression of GATA5 (Messaoudi et al., 2015). Therefore, GATA5 could be a susceptible gene for human hypertension.

10.31083/j.rcm.2020.02.5104.t0001 Table 1 The table of GATA5 mutations and the known associated cardiovascular phenotypes.
References Mutations Types of mutations Cardiovascular phenotypes
Messaoudi et al., 2015 rs6061245 Not mentioned hypertensive
rs6587239 Not mentioned hypertensive
Wang et al., 2013 p. W200G loss-of function AF
Gu et al., 2012 p. Y138F loss-of function AF
p. C210G loss-of function AF
Yang et al., 2012 p. G184V loss-of function AF
p. K218T loss-of function AF
p. A266P loss-of function AF
Shi et al., 2014 p. Y16D loss-of function BAV
p. T252P loss-of function BAV
Kassab et al., 2016 p. T289A missense variant AVC malformation
p. L233P missense variant CoA
p. G166S missense variant SV/TOF
p. Y142H loss-of function DORV
p. T67P missense variant AS, CoA, VSD, PDA, AVC malformation
p. G63A missense variant PS
p. R61S missense variant VSD
p. S19W missense variant ASD, TOF
Wei et al., 2013a p. R187G loss-of function TOF, ASD, PDA
p. H207R loss-of function TOF
Wei et al., 2013b p. L199V loss-of function VSD

AF, atrial fibrillation; BAV, bicuspid aortic valve; AVC, atrio-ventricular canal; CoA, coarctation of the aorta; SV, single ventricle; TOF, tetralogy of fallot; DORV, double outlet right ventricle; AS, aortic stenosis; VSD, ventricular septal defect; PDA, persistent ductus arteriosus; PS, pulmonary stenosis; ASD, atrial septal defect; PDA, patent ductus arteriosus

2.1.1 GATA5 deficiency can lead to endothelial dysfunction and hypertension

The latest data show that GATA5, expressed in adult microvascular endothelial cells, is a new blood pressure regulator that regulates many genes and signaling pathways (such as inflammatory pathway) required for endothelial homeostasis and blood pressure regulation (Messaoudi et al., 2015).

Firstly, GATA5 deficiency can lead to oxidative stress and vascular endothelial dysfunction by reducing the activities of protein kinase A (PKA) and AMPK. In terms of mechanism, decreased AMPK activity caused by GATA5 deficiency can increase the activity of 26 S proteasome by changing the phosphorylation status of proteasome subunits and/or interactions between 19 S and 20 S proteasomes. An activated 26 S proteasome exposes the nuclear localization signal of a nuclear factor κB (NF-κB) dimer by degrading the inhibitor of NF-κB α (IκB-α). This allows NF-κB to translocate to the nucleus and induce the transcription of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits (Messaoudi et al., 2015; Wang et al., 2010). Besides, decreased PKA activity caused by GATA5 deficiency can also increase the activity of NADPH oxidase by directly down-regulating the expression of BMP-4 (Hong et al., 2016; Koga et al., 2013; Zhang et al., 2014). These result in oxidative stress and vascular endothelial dysfunction and ultimately promote the occurrence and development of hypertension (Fig.2).

Figure 2.

Roles of GATA5 in hypertension. GATA5 deficiency can lead to vascular endothelial dysfunction and hypertension by reducing the activities of PKA and AMPK. GATA5, GATA binding protein 5; AMPK, adenosine 5'-monophosphate (AMP)-activated protein kinase; PKA, protein kinase A; NF-κB, nuclear factor κB; IκB-α, inhibitor of NF-κB α; NADPH, nicotinamide adenine dinucleotide phosphate; BMP-4, bone morphogenic protein-4; ROS, reactive oxygen species; IL6, interleukin-6; ICAM-1, intercellular cell adhesion molecule-1; SIRT6, Sirtuin 6; H3K9, histone H3 lysine 9; Nkx3-2, NK3 homeobox 2; PARP-1, poly [ADP ribose] polymerase-1; Bcl-6, B-cell lymphoma-6; VCAM-1, vascular cell adhesion molecule-1; MCP-1, monocyte chemotactic protein-1; MCP-3, monocyte chemotactic protein-3.

Secondly, the functional deficiency of GATA5 up-regulates the expression of pro-inflammatory molecules (such as IL6 and ICAM-1) and decreases vascular nitric oxide (NO) bioavailability, which are expected to contribute to endothelial dysfunction and participate in maintaining the hypertensive phenotype (Messaoudi et al., 2015) (Fig.2).

Finally, poly [ADP ribose] polymerase-1 (PARP-1) inhibits the expression of B-cell lymphoma-6 protein (Bcl-6) by binding to Bcl-6 intron 1. When PARP-1 is phosphorylated by AMPK, Bcl-6 separates from PARP-1 and resumes its expression. Bcl-6 prevents endothelial dysfunction and hypertension by inhibiting the activity of NF-κB and consequently down-regulates the expression of inflammatory genes, including vascular cell adhesion molecule-1, monocyte chemotactic protein (MCP)-1 and MCP-3. However, GATA5 deficiency can inhibit this signal pathway by reducing the activity of AMPK, thus promoting oxidative stress and vascular endothelial dysfunction, eventually leading to hypertension (Gongol et al., 2013) (Fig.2).

2.1.2 Sirtuin 6 (SIRT6) regulates a GATA5-mediated signaling pathway

The epigenetic process mediated by histone deacetylase (HDAC) has attracted much attention in the fields of vascular homeostasis and cardiovascular disease. SIRT6 is a member of the sirtuin family of class III HDACs and is a highly conserved NAD+ dependent deacetylase (Liu et al., 2017; Pillai et al., 2014). Endothelial dysfunction is an important determinant risk factor for the development of hypertension and its complications. SIRT6 is also a key regulator of endothelial function, which inhibits the transcription of the NK3 homeobox 2 (Nkx3-2) by deacetylating histone H3 lysine 9 (H3K9). SIRT6 then induces a GATA5-mediated signaling pathway to prevent endothelial dysfunction and hypertension. Dysfunction of SIRT6 can lead to endothelial dysfunction, hypertension, heart and kidney injury by mediating a GATA5-mediated signaling pathway (Guo et al., 2019) (Fig.2).

2.2 Arrhythmia

Cardiovascular disease is the most common cause of death in the world, with sudden deaths caused by arrhythmias accounting for 50 % of these deaths (Moreau and Chahine, 2018). Atrial fibrillation (AF) is the most common form of arrhythmia observed in clinical practice, as well as being the most common persistent arrhythmia and the main cause of arrhythmia-related hospitalization. It accounts for about 1/3 of hospitalizations due to various arrhythmias. However, AF is a genetically heterogeneous disease with the genetic determinants of most patients still unclear (Gu et al., 2012; Wang et al., 2013). Recently, six significant GATA5 heterozygous mismeaning mutations (p.W200G; p.Y138F; p.C210G; p.G184V, p.K218T and p.A266P) were found in 358 unrelated families with AF (Gu et al., 2012; Wang et al., 2013; Yang et al., 2012) (Table 1), This suggests that the dysfunction of GATA5 may be associated with arrhythmias (including AF).

It has previously been confirmed that mutations of GATA4, GATA6 and NK2 homeobox 5(NKX2-5) have a causal relationship with AF (Yang et al., 2012). GATA4, GATA5 and GATA6 belong to the same GATA subfamily and during cardiac development, especially the process of synergistic regulation of the target gene NKX2-5, their expression and function overlap with each other (Zhang et al., 2007). Thus, GATA5 may promote the formation of a pulmonary myocardial sleeve and shift the pulmonary myocardium to a sinoatrial node-like phenotype by reducing NKX2-5 when dominant negative mutation occurs, hence creating an atrial electrophysiological substrate liable to AF (Yang et al., 2012; Wang et al., 2013).

Additionally, GATA4 and GATA5 bind to the same binding region of the sodium voltage-gated channel alpha subunit 5 (SCN5A) gene promoter and co-activate the transcription of SCN5A gene (Tarradas et al., 2017). Mutations in the SCN5A gene cause a variety of arrhythmias, including Brudaga syndrome, long QT syndrome 3, sick sinus syndrome, cardiac conduction disease, progressive cardiac conduction defect and AF (Han et al., 2018). The SCN5A gene (chromosome 3p21) encodes the alpha subunit of the voltage-gated sodium channel 1.5 (Nav1.5) (Verkerk et al., 2018). Nav1.5 is a giant complex composed of one alpha subunit, four auxiliary beta subunits and many chaperone proteins. It primarily mediates inward sodium current (Savio-Galimberti et al. 2018), which is the main component in the fast depolarization phase after which the excitation-contraction coupling cascade and proper conduction of the electrical impulse is subsequently initiated within the heart (Li et al., 2018). Therefore, GATA5 could regulate cardiac electrophysiological function through a GATA5/SCN5A/NaV1.5 signal pathway and the deficiency of GATA5 function may lead to various arrhythmias (such as AF).

2.3 Congenital heart disease

CHD is the most common congenital defect, accounting for 25 % of the total number of structural and functional defects in newborns and 1 % of all newborns (Lage et al., 2012). Severe CHD can result in decreased exercise ability, poor quality of life, pulmonary hypertension, infective endocarditis, thromboembolism, eisenmenger syndrome, heart failure, arrhythmia and even death (Bouma and Mulder, 2017; Brida et al., 2019; El-Assaad et al., 2017; Papamichalis et al., 2019). Although more than 100 gene mutations have been confirmed to be causally related to CHD (Kalisch-Smith et al., 2019), little is known about the specific molecular genetic mechanisms of these CHD susceptible genes.

2.3.1 GATA5 and heart development

Previous studies have confirmed that GATA transcription factors regulate many cardiac genes, such as atrial natriuretic peptide, brain natriuretic peptide, myocyte enhancer factor 2C, NKX2-5, BMP-4, myosin heavy chain 6 cardiac muscle-α and myosin heavy chain 7 cardiac muscle-β (Laforest and Nemer, 2011). GATA5, a member of the GATA family, can coordinate with other factors to regulate the expression of cardiac genes, such as atrial natriuretic factor, connexin 40 and α-actin (Park et al., 2009). GATA5 is a dosage-sensitive regulator of early heart development and acts earlier than GATA4 to regulate the development of heart precursors (Haworth et al., 2008). Moreover, GATA5 also plays an indispensable role in early myocardial differentiation, endocardial cell differentiation, endocardial cushion differentiation and valvular formation (Laforest et al., 2011; Reiter et al., 2001), efficiently directing cardiac fate from embryonic stem cells (Turbendian et al., 2013). In addition, valve development is a complex process, which involves the differentiation and expansion of endocardial cells and their migration after epithelial-to-mesenchymal transformation to form endocardial cushions within the outflow tract and at the atrioventricular canal. GATA5 is an important regulator of valve development, its dysfunction will lead to defective valve morphogenesis and bicuspid aortic valve (Laforest et al., 2011). In zebrafish, GATA5 dysfunction can result in embryonic lethality due to defects in endocardial and myocardial differentiation migration (Heicklen-Klein, et al., 2004).

2.3.2 GATA5 loss-of-function mutations and CHD

Zinc finger transcription factors encoded by the GATA5 gene are necessary for cardiovascular development and structural remodeling. A large number of GATA5 mutations are related to various congenital cardiovascular malformations, including aortic stenosis, pulmonary stenosis, double outlet of right ventricle, tetralogy of Fallot (TOF), bicuspid aortic valve, ventricular septal defect and atrial septal defect (Zhang et al., 2015) (Table 1).

Shi et al sequenced the splice junction sites and coding regions of GATA5 gene in 110 unrelated patients with bicuspid aortic valve and 200 unrelated healthy subjects. Two new heterozygous mutations (p.Y16D and p.T252P) were found in two families with autosomal dominant inheritance of bicuspid aortic valve. Both of them significantly reduced the transcriptional activity of GATA5. This is the first study on the relationship between increased susceptibility to bicuspid aortic valve and GATA5 loss-of-function mutations (Shi et al., 2014). Kassab et al screened the coding exons of GATA5 in 185 patients with different forms of CHD and 150 healthy individuals. Eight missense variants (p.T289A, p.L233P, p.G166S, p.Y142H, p.T67P, p.G63A, p.R61S and p.S19W) were identified. It is confirmed for the first time that human biallelic GATA5 mutations are directly related to the phenotypes of pulmonary stenosis, ventricular septal defect and double outflow right ventricle (Kassab et al., 2016). Wei et al sequenced all the coding regions of GATA5 gene in 130 unrelated TOF patients and 200 unrelated individuals. Two new heterozygous mutations (p.R187G and p.H207R) were found in two families with autosomal dominantly inherited TOF, which significantly decreased the transcriptional activity of GATA5. This is the first report on the link of functionally compromised GATA5 to human TOF (Wei et al., 2013a). Wei et al sequenced the whole coding region of GATA5 gene in 120 unrelated patients with ventricular septal defect and 200 unrelated control individuals. A novel heterozygous mutation (p.L199V) was identified in a patient, which significantly decreased the transcriptional activity of GATA5. this is the first report on the association between ventricular septal defect and GATA5 loss-of-function mutations (Wei et al., 2013b).

2.3.3 GATA5/Connexin 36.7(cx36.7)/NKX2-5/HEY2 Signaling Pathway

As a coactivator of GATA5, NKX2-5 is also involved in the normal cardiovascular development of vertebrates, such as regulation of the number of cardiac precursor cells, development of the conduction system and valve formation (Anderson et al., 2018). GATA4, GATA5 and GATA6 maintain the expression of NKX2-5 by activating cx36.7 (Miyagi et al., 2016), the expression of NKX2-5 can directly up-regulate the expression of the Hes-related family basic helix-loop-helix (bHLH) transcription factor with YRPW motif 2 (HEY2), SCN5A and other genes (Anderson et al., 2018). SCN5A is closely related to arrhythmia (Han et al., 2018). Of note, HEY2 is a member of the bHLH transcription inhibitor family, the non-synonymous mutations of HEY2 are related to cardiac malformations, such as congenital ventricular septal defect (Fardoun et al., 2019). Therefore, GATA5 participates in the regulation of various congenital heart diseases through a GATA5/cx36.7/NKX2-5/HEY2 signaling pathway (Fig. 3).

Figure 3.

Roles of GATA5 in congenital heart disease. GATA5 participates in heart development via GATA5/cx36. 7/NKX2-5/HEY2 and GATA5/cx36. 7/NKX2-5/GDF1/Nodal signal pathway. GATA5, GATA binding protein 5; cx36. 7, Connexin 36. 7; NKX2-5, NK2 homeobox 5; HEY2, Hes-related family basic helix-loop-helix (bHLH) transcription factor with YRPW motif 2; GDF1, growth differentiation factor 1; Nodal, Nodal growth differentiation factor.

2.3.4 GATA5/cx36.7/NKX2-5/GDF1/ Nodal Signaling Pathway

As mentioned in section 2.3.3, GATA5 maintains the expression of NKX2-5 by activating cx36.7 (Miyagi et al., 2016). NKX2-5 then binds to the growth differentiation factor 1 (GDF1) promoter and activates its expression in cardiac and non-cardiac cells (Gao et al., 2019). Notably, GDF1 directly interacts with Nodal growth differentiation factor (Nodal) to form a GDF1-Nodal heterodimer, which significantly increases the activity of Nodal (Tanaka et al., 2007). Nodal and its family members are important signal molecules for heart development (Barnes et al., 2016; David and Massagué, 2018; Mohapatra et al., 2009). Therefore, a GATA5/cx36.7/NKX2-5/GDF1/Nodal signal pathway provides another way for GATA5 to participate in the occurrence of various congenital heart diseases (Fig.3).

3. Prospects

Functional defects of the GATA5 could lead to the occurrence and development of hypertension, arrhythmia, CHD and many other human diseases through many different signaling pathways. CHD can be caused by environmental factors, oligogenic factors, and/or gene-environment interaction. However, until now, only one-third of CHD cases can be explained by a simple genetic cause. Although compelling evidence that environmental factors can lead to CHD has existed for nearly 80 years, it has not been studied in detail (Kalisch-Smith et al., 2019). It has recently been reported that acute alcohol exposure may ultimately lead to fetal alcohol spectrum disorder (including CHD) by directly affecting the methylation of critical developmental genes (Kalisch-Smith et al., 2019; Ungerer et al., 2013). DNA methylation is one of the most well known epigenetic processes (De Jong et al., 2009). The abnormal methylation of the CpG island in the promoter region of a tumor suppressor gene is the main mechanism of tumor suppressor gene silencing and one of the earliest and most frequent changes in cancer (Belinsky, 2004a, 2005b). GATA5 has been confirmed to be a tumor suppressor gene (Lyon et al., 2007). Abnormal methylation of a GATA5 gene promoter and low level expression of the GATA5 gene have been observed in cancer cells of different types of human cancers, such as lung, pancreatic, esophageal, gastric, colorectal, and retinoblastoma cancers (Akiyama et al., 2003; Guo et al., 2004a, 2006b; Hellebrekers et al., 2009; Livide et al., 2012; Wen et al., 2010).

Therefore, the authors believe that it will be very meaningful to actively explore whether environmental factors lead to the occurrence and development of cardiovascular diseases such as CHD through abnormal methylation of a GATA5 gene promoter. The reasons include: (1) Aberrant methylation of a GATA5 gene promoter can lead to the occurrence and development of various cancers (Akiyama et al., 2003; Guo et al., 2004a, 2006b; Hellebrekers et al., 2009; Livide et al., 2012; Lyon et al., 2007; Wen et al., 2010). (2) Cardiovascular disease and cancer are the two most common chronic human diseases and based on their common etiology and molecular background, they are often interdependent (Regulska et al., 2019). GATA5 mediates the occurrence and development of cardiovascular disease and cancer. For example, the Nodal signaling pathway regulated by GATA5 is not only related to a series of congenital defects, including CHD (Barnes and Black, 2016), but is also involved in maintaining the survival, proliferation, invasion and self-renewal of cancer cell lines (Kalyan et al., 2017; Kirsammer et al., 2014; Wei and Wang, 2018). (3) Epigenetic modifications such as DNA methylation represent the molecular substrate for detrimental environmental stimuli and may lead to cardiovascular disease initiation including CHD (Lorenzen et al., 2012; Vallaster et al., 2012). (4) CHD is primarily caused by non-genetic factors, but the mechanisms associated with environmental factors are still not clear (Kalisch-Smith et al., 2019; Ungerer et al., 2013). This field has great research potential. (5) Gene targeted therapy will inevitably expose the complex in vivo regulatory networks and both its therapeutic effects and side effects are currently unpredictable. But, the interventions of environmental factors are controllable, safe and easy to popularize, to the benefit of many people.

Authors' contributions

Zhipeng Song wrote the paper. Bo Yan reviewed and edited the manuscript. All of the authors read and approved the final manuscript.

Acknowledgments

The authors acknowledge funding support from the National Natural Science Foundation of China (81870279). The authors would also like to extend their thanks to the peer reviewers and editors.

Conflict of interest

The authors declare no conf1licts of interests.

References
[1]
Akiyama, Y., Watkins, N., Suzuki, H., Jair, K. W., van Engeland, M., Esteller, M., Sakai, H., Ren, C. Y., Yuasa, Y., Herman, J. G. and Baylin, S. B. (2003) GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer. Molecular and Cellular Biology 23, 8429-8439. 10.1128/mcb.23.23.8429-8439.200314612389https://www.ncbi.nlm.nih.gov/pubmed/14612389
[2]
Anderson, D. J., Kaplan, D. I., Bell, K. M., Koutsis, K., Haynes, J. M., Mills, R. J., Phelan, D. G., Qian, E. L., Leitoguinho, A. R., and Arasaratnam, D. (2018) NKX2-5 regulates human cardiomyogenesis via a HEY2 dependent transcriptional network. Nature Communications 9, 1373. 10.1038/s41467-018-03714-x29636455https://www.ncbi.nlm.nih.gov/pubmed/29636455
[3]
Barnes, R. M., Black, B. L. (2016) Nodal Signaling and Congenital Heart Defects. In, Nakanishi, T., et al. (eds.) Etiology and Morphogenesis of Congenital Heart Disease (pp. 183-192). Tokyo: Springer.
[4]
Barry, D. M., McMillan, E. A., Kunar, B., Lis, R., Zhang, T., Lu, T., Daniel, E., Yokoyama, M., Gomez-Salinero, J. M., Sureshbabu, A., Cleaver, O., Di Lorenzo, A., Choi, M. E., Xiang, J., Redmond, D., Rabbany, S. Y., Muthukumar, T. and Rafii, S. (2019) Molecular determinants of nephron vascular specialization in the kidney. Nature Communications 10, 5705. 10.1038/s41467-019-12872-531836710https://www.ncbi.nlm.nih.gov/pubmed/31836710
[5]
Belinsky, S. A. (2004a) Gene-promoter hypermethylation as a biomarker in lung cancer. Nature Reviews Cancer 4, 707-717. 10.1038/nrc143215343277https://www.ncbi.nlm.nih.gov/pubmed/15343277
[6]
Belinsky, S. A. (2005b) Silencing of genes by promoter hypermethylation: key event in rodent and human lung cancer. Carcinogenesis 26, 1481-1487. 10.1093/carcin/bgi02015661809https://www.ncbi.nlm.nih.gov/pubmed/15661809
[7]
Bouma, B. J. and Mulder, B. J. (2017) Changing Landscape of Congenital Heart Disease. Circulation Research 120, 908-922. 10.1161/CIRCRESAHA.116.30930228302739https://www.ncbi.nlm.nih.gov/pubmed/28302739
[8]
Brida, M., Diller, G. P., Nashat, H., Strozzi, M., Milicic, D., Baumgartner, H., and Gatzoulis, M. A. (2019) Pharmacological therapy in adult congenital heart disease: growing need, yet limited evidence. European Heart Journal 40, 1049-1056. 10.1093/eurheartj/ehy48030137263https://www.ncbi.nlm.nih.gov/pubmed/30137263
[9]
Bruun, C., Christensen, G. L., Jacobsen, M. L., Kanstrup, M. B., Jensen, P. R., Fjordvang, H., Mandrup-Poulsen, T., and Billestrup, N. (2014) Inhibition of beta cell growth and function by bone morphogenetic proteins. Diabetologia 57, 2546-2554. 10.1007/s00125-014-3384-8a2ab2200-585e-49c6-a0b1-5e7cea5eec0ahttp://dx.doi.org/10.1007/s00125-014-3384-8
[10]
Daehn, I., Casalena, G., Zhang, T., Shi, S., Fenninger, F., Barasch, N., Yu, L., D'Agati, V., Schlondorff, D., Kriz, W., Haraldsson, B., and Bottinger, E. P. (2014) Endothelial mitochondrial oxidative stress determines podocyte depletion in segmental glomerulosclerosis. The Journal of Clinical Investigation 124, 1608-1621. 10.1172/JCI7119524590287https://www.ncbi.nlm.nih.gov/pubmed/24590287
[11]
David, C. J., Massagué, J. (2018) Contextual determinants of TGFβ action in development, immunity and cancer. Nature Reviews Molecular Cell Biology 19, 419-435. 10.1038/s41580-018-0007-029643418https://www.ncbi.nlm.nih.gov/pubmed/29643418
[12]
De Jong, W. K., Verpooten, G. F., Kramer, H., Louwagie, J., and Groen, H. J. (2009) Promoter methylation primarily occurs in tumor cells of patients with non-small cell lung cancer. Anticancer Research 29, 363-369. 19331174https://www.ncbi.nlm.nih.gov/pubmed/19331174
[13]
Du, X., Patel, A., Anderson, C. S., Dong, J., and Ma, C. (2019) Epidemiology of cardiovascular disease in china and opportunities for improvement: JACC International. Journal of the American College of Cardiology 73, 3135-3147. 10.1016/j.jacc.2019.04.03631221263https://www.ncbi.nlm.nih.gov/pubmed/31221263
[14]
El-Assaad, I., Al-Kindi, S. G., and Aziz, P. F. (2017) Trends of out-of-hospital sudden cardiac death among children and young adults. Journal of Neurosurgery: Pediatrics 140, e20171438.
[15]
Fardoun, M., Dehaini, H., Kamar, A., Bitar, F., Majdalani, M., El-Rassi, I., Nemer, G., and Arabi, M. (2019) A novel somatic variant in HEY2 unveils an alternative splicing isoform linked to ventricular septal defect. Pediatric Cardiology 40, 1084-1091. 10.1007/s00246-019-02099-y30955100https://www.ncbi.nlm.nih.gov/pubmed/30955100
[16]
Fujiwara, T. (2017) GATA transcription factors: basic principles and related human disorders. The Tohoku Journal of Experimental Medicine 242, 83-91. 10.1620/tjem.242.8328566565https://www.ncbi.nlm.nih.gov/pubmed/28566565
[17]
Gao, X., Zheng, P., Yang, L., Luo, H., Zhang, C., Qiu, Y., Huang, G., Sheng, W., Ma, X., and Lu, C. (2019) Association of functional variant in GDF1 promoter with risk of congenital heart disease and its regulation by Nkx2. 5. Clinical Science 133, 1281-1295. 10.1042/CS2018102431171573https://www.ncbi.nlm.nih.gov/pubmed/31171573
[18]
Gongol, B., Marin, T., Peng, I. C., Woo, B., Martin, M., King, S., Sun, W., Johnson, D. A., Chien, S., and Shyy, J. Y. (2013) AMPKα2 exerts its anti-inflammatory effects through PARP-1 and Bcl-6. Proceedings of the National Academy of Sciences of the United States of America 110, 3161-3166. 10.1073/pnas.12220511103daf874b-6c61-440a-9374-7ae5a37a05bahttp://dx.doi.org/10.1073/pnas.1222051110
[19]
Gu, J. Y., Xu, J. H., Yu, H., and Yang, Y. Q. (2012) Novel GATA5 loss-of-function mutations underlie familial atrial fibrillation. Clinics 67, 1393-1399. 10.6061/clinics/2012(12)0823295592https://www.ncbi.nlm.nih.gov/pubmed/23295592
[20]
Guo, J., Wang, Z., Wu, J., Liu, M., Li, M., Sun, Y., Huang, W., Li, Y., Zhang, Y., Tang, W., Li, X., Zhang, C., Hong, F., Li, N., Nie, J., and Yi, F. (2019) Endothelial SIRT6 is vital to prevent hypertension and associated cardiorenal injury through targeting Nkx3. 2-GATA5 signaling. Circulation Research 124, 1448-1461. 10.1161/CIRCRESAHA.118.31403230894089https://www.ncbi.nlm.nih.gov/pubmed/30894089
[21]
Guo, M., Akiyama, Y., House, M. G., Hooker, C. M., Heath, E., Gabrielson, E., Yang, S. C., Han, Y., Baylin, S. B., Herman, J. G., and Brock, M. V. (2004a) Hypermethylation of the GATA genes in lung cancer. Clinical Cancer Research 10, 7917-7924. 10.1158/1078-0432.CCR-04-114015585625https://www.ncbi.nlm.nih.gov/pubmed/15585625
[22]
Guo, M., House, M. G., Akiyama, Y., Qi, Y., Capagna, D., Harmon, J., Baylin, S. B., Brock, M. V., and Herman, J. G. (2006b) Hypermethylation of the GATA gene family in esophageal cancer. International Journal of Cancer 119, 2078-2083. 10.1002/ijc.2209216823849https://www.ncbi.nlm.nih.gov/pubmed/16823849
[23]
Han, D., Tan, H., Sun, C., and Li, G. (2018) Dysfunctional Nav1. 5 channels due to SCN5A mutations. Experimental Biology and Medicine 243, 852-863. 10.1177/153537021877797229806494https://www.ncbi.nlm.nih.gov/pubmed/29806494
[24]
Haworth, K. E., Kotecha, S., Mohun, T. J., and Latinkic, B. V. (2008) GATA4 and GATA5 are essential for heart and liver development in Xenopus embryos. BMC Developmental Biology 8, 74. 10.1186/1471-213X-8-7418662378https://www.ncbi.nlm.nih.gov/pubmed/18662378
[25]
Heicklen-Klein, A., McReynolds, L. J., and Evans, T. (2004) Using the zebrafish model to study GATA transcription factors. Seminars in Cell & Developmental Biology 16, 95-106. 10.1016/j.semcdb.2004.10.00415659344https://www.ncbi.nlm.nih.gov/pubmed/15659344
[26]
Hellebrekers, D. M., Lentjes, M. H., van den Bosch, S. M., Melotte, V., Wouters, K. A., Daenen, K. L., Smits, K. M., Akiyama, Y., Yuasa, Y., and Sanduleanu, S. (2009) GATA4 and GATA5 are potential tumor suppressors and biomarkers in colorectal cancer. Clinical Cancer Research 15, 3990-3997. 10.1158/1078-0432.CCR-09-005519509152https://www.ncbi.nlm.nih.gov/pubmed/19509152
[27]
Hong, O. K., Yoo, S. J., Son, J. W., Kim, M. K., Baek, K. H., Song, K. H., Cha, B. Y., Jo, H., and Kwon, H. S. (2016) High glucose and palmitate increases bone morphogenic protein 4 expression in human endothelial cells. Korean Journal of Physiology and Pharmacology 20, 169-175. 10.4196/kjpp.2016.20.2.16926937213https://www.ncbi.nlm.nih.gov/pubmed/26937213
[28]
Jefferson, J. A. and Shankland, S. J. (2014) The pathogenesis of focal segmental glomerulosclerosis. Advances in Chronic Kidney Disease 21, 408-416. 10.1053/j.ackd.2014.05.0092154f60e-106f-43fd-b19a-a4273bb804d7http://dx.doi.org/10.1053/j.ackd.2014.05.009
[29]
Kalisch-Smith, J. I., Ved, N., and Sparrow, D. B. (2019) Environmental risk factors for congenital heart disease. Cold Spring Harbor Perspectives in Biology (in press).
[30]
Kalyan, A., Carneiro, B. A., Chandra, S., Kaplan, J., Chae, Y. K., Matsangou, M., Hendrix, M. J. C., and Giles, F. (2017) Nodal signaling as a developmental therapeutics target in oncology. Molecular Cancer Therapeutics 16, 787-792. 10.1158/1535-7163.MCT-16-021528468864https://www.ncbi.nlm.nih.gov/pubmed/28468864
[31]
Kassab, K., Hariri, H., Gharibeh, L., Fahed, A. C., Zein, M., El-Rassy, I., Nemer, M., El-Rassi, I., Bitar, F., and Nemer, G. (2016) GATA5 mutation homozygosity linked to a double outlet right ventricle phenotype in a Lebanese patient. Molecular Genetics & Genomic Medicine 4, 160-171. 10.1002/mgg3.19027066509https://www.ncbi.nlm.nih.gov/pubmed/27066509
[32]
Kirsammer, G., Strizzi, L., Margaryan, N. V., Gilgur, A., Hyser, M., Atkinson, J., Kirschmann, D. A., Seftor, E. A., and Hendrix, M. J. (2014) Nodal signaling promotes a tumorigenic phenotype in human breast cancer. Seminars in Cancer Biology 29, 40-50. 10.1016/j.semcancer.2014.07.007faf4501b-8787-4a45-9663-a4eb13d231f6http://dx.doi.org/10.1016/j.semcancer.2014.07.007
[33]
Koga, M., Engberding, N., Dikalova, A. E., Chang, K. H., Seidel-Rogol, B., Long, J. S., Lassègue, B., Jo, H., and Griendling, K. K. (2013) The bone morphogenic protein inhibitor, noggin, reduces glycemia and vascular inflammation in db/db mice. American Journal of Physiology - Heart and Circulatory Physiology 305, 747-755.
[34]
Laforest, B. and Nemer, M. (2011) GATA5 interacts with GATA4 and GATA6 in outflow tract development. Developmental Biology 358, 368-378. 10.1016/j.ydbio.2011.07.0373bca77cc-4245-4a5f-9f34-224eca3afdeehttp://dx.doi.org/10.1016/j.ydbio.2011.07.037
[35]
Laforest, B., Andelfinger, G., and Nemer, M. (2011) Loss of Gata5 in mice leads to bicuspid aortic valve. The Journal of Clinical Investigation 121, 2876-2887. 10.1172/JCI4455521633169https://www.ncbi.nlm.nih.gov/pubmed/21633169
[36]
Lage, K., Greenway, S. C., Rosenfeld, J. A., Wakimoto, H., Gorham, J. M., Segrè, A. V., Roberts, A. E., Smoot, L. B., Pu, W. T., Pereira, A. C., Mesquita, S. M., Tommerup, N., Brunak, S., Ballif, B. C., Shaffer, L. G., Donahoe, P. K., Daly, M. J., Seidman, J. G., Seidman, C. E., and Larsen, L. A. (2012) Genetic and environmental risk factors in congenital heart disease functionally converge in protein networks driving heart development. Proceedings of the National Academy of Sciences of the United States of America 109, 14035-14040. 10.1073/pnas.1210730109774c5ba9-0a63-42a3-8659-e730d3046516http://dx.doi.org/10.1073/pnas.1210730109
[37]
Lentjes, M. H., Niessen, H. E., Akiyama, Y., de Bruïne, A. P., Melotte, V., and van Engeland, M. (2016) The emerging role of GATA transcription factors in development and disease. Expert Reviews in Molecular Medicine 18, e3. 10.1017/erm.2016.226953528https://www.ncbi.nlm.nih.gov/pubmed/26953528
[38]
Li, W., Yin, L., Shen, C., Hu, K., Ge, J., and Sun, A. (2018) SCN5A variants: association with cardiac disorders. Frontiers in Physiology 9, 1372. 10.3389/fphys.2018.0137230364184https://www.ncbi.nlm.nih.gov/pubmed/30364184
[39]
Liu, M., Liang, K., Zhen, J., Zhou, M., Wang, X., Wang, Z., Wei, X., Zhang, Y., Sun, Y., Zhou, Z., Su, H., Zhang, C., Li, N., Gao, C., Peng, J., and Yi, F. (2017) Sirt6 deficiency exacerbates podocyte injury and proteinuria through targeting Notch signaling. Nature Communications 8, 413. 10.1038/s41467-017-00498-428871079https://www.ncbi.nlm.nih.gov/pubmed/28871079
[40]
Livide, G., Epistolato, M. C., Amenduni, M., Disciglio, V., Marozza, A., Mencarelli, M. A., Toti, P., Lazzi, S., Hadjistilianou, T., De Francesco, S., D'Ambrosio, A., Renieri, A., and Ariani, F. (2012) Epigenetic and copy number variation analysis in retinoblastoma by MS-MLPA. Pathology and Oncology Research 18, 703-712. 10.1007/s12253-012-9498-822278416https://www.ncbi.nlm.nih.gov/pubmed/22278416
[41]
Lorenzen, J. M., Martino, F., and Thum, T. (2012) Epigenetic modifications in cardiovascular disease. Basic Research in Cardiology 107, 245. 10.1007/s00395-012-0245-922234702https://www.ncbi.nlm.nih.gov/pubmed/22234702
[42]
Lyon, C. M., Klinge, D. M., Liechty, K. C., Gentry, F. D., March, T. H., Kang, T., Gilliland, F. D., Adamova, G., Rusinova, G., and Telnov, V. (2007) Radiation-induced lung adenocarcinoma is associated with increased frequency of genes inactivated by promoter hypermethylation. Radiation Research 168, 409-414. 10.1667/RR0825.117903034https://www.ncbi.nlm.nih.gov/pubmed/17903034
[43]
Malakar, A. K., Choudhury, D., Halder, B., Paul, P., Uddin, A., and Chakraborty, S. (2019) A review on coronary artery disease, its risk factors, and therapeutics. Journal of Cellular Physiology 234, 16812-16823. 10.1002/jcp.2835030790284https://www.ncbi.nlm.nih.gov/pubmed/30790284
[44]
Messaoudi, S., He, Y., Gutsol, A., Wight, A., Hébert, R. L., Vilmundarson, R. O., Makrigiannis, A. P., Chalmers, J., Hamet, P., Tremblay, J., McPherson, R., Stewart, A. F. R., Touyz, R. M., and Nemer, M. (2015) Endothelial Gata5 transcription factor regulates blood pressure. Nature Communications 6, 8835. 10.1038/ncomms983526617239https://www.ncbi.nlm.nih.gov/pubmed/26617239
[45]
Miyagi, H., Nag, K., Sultana, N., Munakata, K., Hirose, S., and Nakamura, N. (2016) Characterization of the zebrafish cx36. 7 gene promoter: Its regulation of cardiac-specific expression and skeletal muscle-specific repression. Gene 577, 265-274. 10.1016/j.gene.2015.12.01326692140https://www.ncbi.nlm.nih.gov/pubmed/26692140
[46]
Mohapatra, B., Casey, B., Li, H., Ho-Dawson, T., Smith, L., Fernbach, S. D., Molinari, L., Niesh, S. R., Jefferies, J. L., Craigen, W. J., Towbin, J. A., Belmont, J. W., and Ware, S. M. (2009) Identification and functional characterization of NODAL rare variants in heterotaxy and isolated cardiovascular malformations. Human Molecular Genetics 18, 861-871. 10.1093/hmg/ddn41119064609https://www.ncbi.nlm.nih.gov/pubmed/19064609
[47]
Moreau, A. and Chahine, M. (2018) A new cardiac channelopathy: from clinical phenotypes to molecular mechanisms associated with Nav1. 5 gating pores. Frontiers in Cardiovascular Medicine 5, 139. 10.3389/fcvm.2018.0013930356750https://www.ncbi.nlm.nih.gov/pubmed/30356750
[48]
Papamichalis, M., Xanthopoulos, A., Papamichalis, P., Skoularigis, J., and Triposkiadis, F. (2019) Adult congenital heart disease with pulmonary arterial hypertension: mechanisms and management. Heart Failure Reviews (in press.)
[49]
Park, J. S., Kim, H. S., Kim, J. D., Seo, J., Chung, K. S., Lee, H. S., Huh, T. L., Jo, I., and Kim, Y. O. (2009) Isolation of a ventricle-specific promoter for the zebrafish ventricular myosin heavy chain (vmhc) gene and its regulation by GATA factors during embryonic heart development. Developmental Dynamics: an Official Publication of the American Association of Anatomists 238, 1574-1581. 10.1002/dvdy.v238:6http://doi.wiley.com/10.1002/dvdy.v238%3A6
[50]
Pillai, V. B., Sundaresan, N. R., and Gupta, M. P. (2014) Regulation of Akt signaling by sirtuins: its implication in cardiac hypertrophy and aging. Circulation Research 114, 368-378. 10.1161/CIRCRESAHA.113.30053693c6163e-ff4b-403b-944c-78808f3adcf6http://dx.doi.org/10.1161/CIRCRESAHA.113.300536
[51]
Rafii, S., Butler, J. M., and Ding, B. S. (2016) Angiocrine functions of organ-specific endothelial cells. Nature 529, 316-325. 10.1038/nature1704026791722https://www.ncbi.nlm.nih.gov/pubmed/26791722
[52]
Regulska, K., Regulski, M., Karolak, B., Michalak, M., Murias, M., and Stanisz, B. (2019) Beyond the boundaries of cardiology: Still untapped anticancer properties of the cardiovascular system-related drugs. Pharmacological Research 147, 104326. 10.1016/j.phrs.2019.10432631340189https://www.ncbi.nlm.nih.gov/pubmed/31340189
[53]
Reiter, J. F., Verkade, H. B., Stainier, D. Y. (2001) mp2b and Oep promote early myocardial differentiation through their regulation of gata5. Developmental Biology 234, 330-338. 10.1006/dbio.2001.025911397003https://www.ncbi.nlm.nih.gov/pubmed/11397003
[54]
Savio-Galimberti, E., Argenziano, M., and Antzelevitch, C. (2018) Cardiac arrhythmias related to sodium channel dysfunction. Handbook of Experimental Pharmacology 246, 331-354. 10.1007/164_2017_4328965168https://www.ncbi.nlm.nih.gov/pubmed/28965168
[55]
Shen, C. and Ge, J. (2018) Epidemic of Cardiovascular Disease in China. Circulation 138, 342-344. 10.1161/CIRCULATIONAHA.118.03348430571361https://www.ncbi.nlm.nih.gov/pubmed/30571361
[56]
Shi, L. M., Tao, J. W., Qiu, X. B., Wang, J., Yuan, F., Xu, L., Liu, H., Li, R. G., Xu, Y. J., Wang, Q., Zheng, H. Z., Li, X., Wang, X. Z., and Zhang, M. (2014) GATA5 loss-of-function mutations associated with congenital bicuspid aortic valve. International Journal of Molecular Medicine 33, 1219-1226. 10.3892/ijmm.2014.17003eb7987c-7ec9-4f74-b76b-5ae399632e95http://dx.doi.org/10.3892/ijmm.2014.1700
[57]
Tanaka, C., Sakuma, R., Nakamura, T., Hamada, H., and Saijoh, Y. (2007) Long-range action of Nodal requires interaction with GDF1. Genes and Development 21, 3272-82. 10.1101/gad.162390718079174https://www.ncbi.nlm.nih.gov/pubmed/18079174
[58]
Tarradas, A., Pinsach-Abuin, M. L., Mackintosh, C., Llorà-Batlle, O., Pérez-Serra, A., Batlle, M., Pérez-Villa, F., Zimmer, T., Garcia-Bassets, I., Brugada, R., Beltran-Alvarez, P., and Pagans, S. (2017) Transcriptional regulation of the sodium channel gene (SCN5A) by GATA4 in human heart. Journal of Molecular and Cellular Cardiology 102, 74-82. 10.1016/j.yjmcc.2016.10.01327894866https://www.ncbi.nlm.nih.gov/pubmed/27894866
[59]
Tominaga, T., Abe, H., Ueda, O., Goto, C., Nakahara, K., Murakami, T., Matsubara, T., Mima, A., Nagai, K., Araoka, T., Kishi, S., Fukushima, N., Jishage, K., and Doi, T. (2011) Activation of bone morphogenetic protein 4 signaling leads to glomerulosclerosis that mimics diabetic nephropathy. The Journal of Biological Chemistry 286, 20109-20116. 10.1074/jbc.M110.17938221471216https://www.ncbi.nlm.nih.gov/pubmed/21471216
[60]
Turbendian, H. K., Gordillo, M., Tsai, S. Y., Lu, J., Kang, G., Liu, T. C., Tang, A., Liu, S., Fishman, G. I., and Evans, T. (2013) GATA factors efficiently direct cardiac fate from embryonic stem cells. Development 140, 1639-1644. 10.1242/dev.093260aedd44a9-6ebe-4961-a48a-ec785ac40a57http://dx.doi.org/10.1242/dev.093260
[61]
Ungerer, M., Knezovich, J., and Ramsay, M. (2013) In utero alcohol exposure, epigenetic changes, and their consequences. Alcohol Research: Current Reviews 35, 37-46
[62]
Vallaster, M., Vallaster, C. D., and Wu, S. M. (2012) Epigenetic mechanisms in cardiac development and disease. Acta Biochimica et Biophysica Sinica 44, 92-102. 10.1093/abbs/gmr0909a49905c-3eb3-4ded-89b2-14f8b0f27999http://dx.doi.org/10.1093/abbs/gmr090
[63]
Verkerk, A. O., Amin, A. S., and Remme, C. A. (2018) Disease modifiers of inherited SCN5A Channelopathy. Frontiers in Cardiovascular Medicine 5, 137. 10.3389/fcvm.2018.0013730327767https://www.ncbi.nlm.nih.gov/pubmed/30327767
[64]
Wang, D., Song, Y., Zhang, J., Pang, W., Wang, X., Zhu, Y., and Li, X. (2017) AMPK-KLF2 signaling pathway mediates the proangiogenic effect of erythropoietin in endothelial colony-forming cells. American Journal of Physiology - Cell Physiology 313, C674-C685. 10.1152/ajpcell.00257.201628978525https://www.ncbi.nlm.nih.gov/pubmed/28978525
[65]
Wang, S., Zhang, M., Liang, B., Xu, J., Xie, Z., Liu, C., Viollet, B., Yan, D., and Zou, M. H. (2010) AMPKalpha2 deletion causes aberrant expression and activation of NAD(P)H oxidase and consequent endothelial dysfunction in vivo: role of 26S proteasomes. Circulation Research 106, 1117-1128. 10.1161/CIRCRESAHA.109.21253020167927https://www.ncbi.nlm.nih.gov/pubmed/20167927
[66]
Wang, X. H., Huang, C. X., Wang, Q., Li, R. G., Xu, Y. J., Liu, X., Fang, W. Y., and Yang, Y. Q. (2013) A novel GATA5 loss-of-function mutation underlies lone atrial fibrillation. International Journal of Molecular Medicine 31, 43-50. 10.3892/ijmm.2012.11892144f8f6-9483-4aac-ad76-0681c4126911http://dx.doi.org/10.3892/ijmm.2012.1189
[67]
Wei, D., Bao, H., Liu, X. Y., Zhou, N., Wang, Q., Li, R. G., Xu, Y. J., and Yang, Y. Q. (2013a) GATA5 loss-of-function mutations underlie tetralogy of fallot. International Journal of Medical Sciences 10, 34-42. 10.7150/ijms.527006911736-7b98-4220-8762-4dc31b7c8838http://dx.doi.org/10.7150/ijms.5270
[68]
Wei, D., Bao, H., Zhou, N., Zheng, G. F., Liu, X. Y., and Yang, Y. Q. (2013b) GATA5 loss-of-function mutation responsible for the congenital ventriculoseptal defect. Pediatric cardiology 34, 504-511. 10.1007/s00246-012-0482-640a179d8-f089-471e-8118-3b18842d7666http://link.springer.com/article/10.1007/s00246-012-0482-6
[69]
Wei, S. and Wang, Q. (2018) Molecular regulation of Nodal signaling during mesendoderm formation. Acta Biochimica et Biophysica Sinica 50, 74-81. 10.1093/abbs/gmx12829206913https://www.ncbi.nlm.nih.gov/pubmed/29206913
[70]
Wen, X. Z., Akiyama, Y., Pan, K. F., Liu, Z. J., Lu, Z. M., Zhou, J., Gu, L. K., Dong, C. X., Zhu, B. D., Ji, J. F., You, W. C., and Deng, D. J. (2010) Methylation of GATA-4 and GATA-5 and development of sporadic gastric carcinomas. World Journal of Gastroenterology 16, 1201-1208. 10.3748/wjg.v16.i10.120120222162https://www.ncbi.nlm.nih.gov/pubmed/20222162
[71]
Yang, Y. Q., Wang, J., Wang, X. H., Wang, Q., Tan, H. W., Zhang, M., Shen, F. F., Jiang, J. Q., Fang, W. Y., and Liu, X. (2012) Mutational spectrum of the GATA5 gene associated with familial atrial fibrillation. International Journal of Cardiology 157, 305-307. 10.1016/j.ijcard.2012.03.13222483626363d19ea-1cb5-4772-bbaf-e7b33e730e31https://www.ncbi.nlm.nih.gov/pubmed/22483626
[72]
Ying, Y., Ueta, T., Jiang, S., Lin, H., Wang, Y., Vavvas, D., Wen, R., Chen, Y. G., and Luo, Z. (2017) Metformin inhibits ALK1-mediated angiogenesis via activation of AMPK. Oncotarget 8, 32794-32806. 10.18632/oncotarget.1582528427181https://www.ncbi.nlm.nih.gov/pubmed/28427181
[73]
Zhang, X. L., Dai, N., Tang, K., Chen, Y. Q., Chen, W., Wang, J., Zhao, C. M., Yuan, F., Qiu, X. B., Qu, X. K., Yang, Y. Q., and Xu, Y. W. (2015) GATA5 loss-of-function mutation in familial dilated cardiomyopathy. International Journal of Molecular Medicine 35, 763-770. 10.3892/ijmm.2014.205025543888https://www.ncbi.nlm.nih.gov/pubmed/25543888
[74]
Zhang, Y., Liu, J., Tian, X. Y., Wong, W. T., Chen, Y., Wang, L., Luo, J., Cheang, W. S., Lau, C. W., Kwan, K. M., Wang, N., Yao, X., and Huang, Y. (2014) Inhibition of bone morphogenic protein 4 restores endothelial function in db/db diabetic mice. Arteriosclerosis, Thrombosis, and Vascular Biology 34, 152-159. 10.1161/ATVBAHA.113.30269624202302https://www.ncbi.nlm.nih.gov/pubmed/24202302
[75]
Zhang, Y., Rath, N., Hannenhalli, S., Wang, Z., Cappola, T., Kimura, S., Atochina-Vasserman, E., Lu, M. M., Beers, M. F., and Morrisey, E. E. (2007) GATA and Nkx factors synergistically regulate tissue-specific gene expression and development in vivo. Development 134, 189-198. 10.1242/dev.0272017164424https://www.ncbi.nlm.nih.gov/pubmed/17164424
[76]
Zibrova, D., Vandermoere, F., Göransson, O., Peggie, M., Mariño, K. V., Knierim, A., Spengler, K., Weigert, C., Viollet, B., Morrice, N. A., Sakamoto, K., and Heller, R. (2017) GFAT1 phosphorylation by AMPK promotes VEGF-induced angiogenesis. Biochemical Journal 474, 983-1001. 10.1042/BCJ2016098028008135https://www.ncbi.nlm.nih.gov/pubmed/28008135
Share
Back to top