Next Article in Journal
Novel Inhibitory Role of Fenofibric Acid by Targeting Cryptic Site on the RBD of SARS-CoV-2
Previous Article in Journal
Mechanistic Analysis of CCP1 in Generating ΔC2 α-Tubulin in Mammalian Cells and Photoreceptor Neurons
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Variants of ISL1 Gene Promoter Identified from Congenital Tetralogy of Fallot Patients Alter Cellular Function Forming Disease Basis

1
The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin 300457, China
2
School of Pharmacy, Drug Research & Development Center, Wannan Medical College, Wuhu 241002, China
*
Author to whom correspondence should be addressed.
Submission received: 21 November 2022 / Revised: 29 December 2022 / Accepted: 6 January 2023 / Published: 13 February 2023
(This article belongs to the Section Molecular Medicine)

Abstract

:
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease in newborns. ISL1 is a master transcription factor in second heart field development, whereas the roles of ISL1 gene promoter variants in TOF patients have not been genetically investigated. Total DNA extraction from 601 human subjects, including 308 TOF patients and 293 healthy controls, and Sanger sequencing were performed. Four variants (including one novel heterozygous variant) within the ISL1 gene promoter were only found in TOF patients. Functional analysis of DNA sequence variants was performed by using the dual-luciferase reporter assay and demonstrated that three of the four variants significantly decreased the transcriptional activity of ISL1 gene promoter in HL-1 cells (p < 0.05). Further, the online JASPAR database and electrophoretic mobility shift assay showed that the three variants affected the binding of transcription factors and altered ISL1 expression levels. In conclusion, the current study for the first time demonstrated that the variants identified from the ISL1 gene promoter region are likely involved in the development of TOF by affecting the transcriptional activity and altering the ISL1 expression level. Therefore, these findings may provide new insights into the molecular etiology and potential therapeutic strategy of TOF.

Graphical Abstract

1. Introduction

Tetralogy of Fallot (TOF) is the most common form of cyanotic congenital heart disease in newborns, with a prevalence of 4 per 10,000 live births, accounting for up to 7–10% of congenital heart diseases (CHDs) [1,2]. TOF is characterized as a combination of abnormalities (ventricular septal defect, obstruction of the right ventricular outflow tract, override of the ventricular septum by the aortic root, and right ventricular hypertrophy) that together lead to tissue hypoxia. Although surgical correction during infancy repaired most cases of TOF and increased the survival of TOF patients [3], there is significant morbidity in adulthood, particularly with pulmonary regurgitation and arrhythmias [4].
The genetic factor has been demonstrated to be the major etiology of CHDs [5]. Accumulating evidence has indicated that the cause of TOF is complex and genetic factors are major contributors to the incidence of TOF [6]. The presence of newer genomic technologies, especially genome-wide sequencing, has identified novel TOF-specific copy number variants and revealed the genetic basis of TOF [7]. Studies have identified susceptible genes related to the etiology of TOF, such as NOTCH1 [6], FLT4 [6], ISL1, TBX1 [8], PIPN11 [9], and NRP1 [10]. However, the roles of these associated genes and pathways in the formation of TOF remain largely unknown.
The transcription factor ISL1 (OMIM: 600366), which is located on chromosome 5q11.1, has been considered an important regulator of heart development. As a crucial member of the LIM homeobox family of transcriptions, ISL1 is a marker for cardiac progenitor cells, primarily in marking undifferentiated, proliferating progenitors in the second heart field, and is essential for the proliferation, migration, and survival of cardiac progenitor cells [11,12,13]. In mice, homozygous null for ISL1 appears to lead to serious heart development defects, including absence of right ventricle, much of the right atrium, and outflow tract [11]. Furthermore, it has been found that several mutations of ISL1 were associated with diverse types of CHDs in humans [14,15,16,17]. It is well known that the promoter, located at 5′upstream of gene, plays a key role in gene transcriptional regulation. Accumulating evidence has reported that the variants of gene promoter region may affect the expression level of genes, leading to the development of disease [14,18,19]. However, there are no studies focusing on the variant of ISL1 gene promoter region in TOF patients. Therefore, we hypothesized the genetic variants in ISL1 gene promoter may contribute to the development of TOF by altering the ISL1 gene expression level and affecting the binding ability of transcription factor. To confirm this hypothesis, we designed this study and identified variants within the ISL1 gene promoter in patients with TOF in comparison with healthy controls and then functionally analyzed the effect of the variants on the ISL1 gene expression.

2. Materials and Methods

2.1. Study Population

In this study, a total of 308 Chinese Han patients (175 males and 133 females; age range: 3 months to 19 years) diagnosed with TOF from the Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University (Tianjin, China) and 293 Chinese Han healthy control subjects (165 males and 128 females; age range: 1 month to 14 years) with similar sex and age were recruited. We made an endeavor to recruit sporadic TOF patients without family history of CHD. All the normal control subjects were confirmed by echocardiography without CHDs or any other diseases. The investigation was in compliance with the outlined guidelines in the Declaration of Helsinki. The informed written consent was obtained from all subjects or their legal guardians prior to the study. Research protocol for this study was approved by the Ethics Committee of TEDA International Cardiovascular Hospital (Clinical Research Ethics Review Number: 2021-0715-4). The flow chart is shown in Figure 1.

2.2. ISL1 Promoter Sequence Analysis

Peripheral blood samples were collected according to the standard clinical procedure. Genomic DNA was extracted from peripheral blood for each TOF patient and control subject with TIANamp Blood DNA Kit (TIANGEN, China). The polymerase chain reaction (PCR) primers were designed based upon the reference sequence of the human ISL1 gene in GenBank database (NCBI: NG_023040.1) as previously reported [14]. The promoter region of the human ISL1 gene (1398 bp, from −1286 bp to +111 bp to the transcription start site) was amplified by PCR and directly bidirectionally sequenced. The primers for Sanger sequencing are listed in Table 1. Subsequently, the DNA sequences of above PCR products were compared with the wild-type ISL1 promoter sequence.

2.3. Plasmid Constructs, Cell Culture, and Transfection

To functionally analyze the variants of ISL1 gene promoter, DNA fragments of ISL1 gene promoter with wild-type or variant-type, containing the terminal restriction sites KpnI and BglII, were generated by PCR and subcloned into the KpnI/BglII sites of the pGL3-basic to constitute expression vectors that were validated by Sanger sequencing. PCR primers are shown in Table 1.
Mouse cardiomyocyte (HL-1) cells were cultured in DMEM (Dullbecco’s modified Eagle’s medium, Gibco, Thermo Fisher, Waltham, MA, USA), to which was added 10% FBS (Fetal Bovine Serum, ExCell Bio, Shanghai, China), 100 μg/mL streptomycin (Beyotime, Shanghai, China), and 100 units/mL penicillin (Beyotime, Shanghai, China) in a 5% CO2 incubator at 37 °C. The above-constructed reporter plasmids with Lipofectamine™ 2000 Transfection Reagent (Invitrogen, Thermo Fisher, USA) were transfected into HL-1 cells. For standardizing the comparison between the variant-group and the wild-group, the expressing renilla luciferase reporter plasmid (pRL-SV40) was co-transfected into all groups as an internal control to normalize transfection efficiency. The empty pGL3-basic plasmid was utilized as a negative control.

2.4. Functional Analysis with Dual-Luciferase Reporter Assay

Transfected cells were harvested and lysed after 36 h. Firely and renilla luciferase activities of cells were measured with Dual Luciferase Reporter Gene Assay Kit (Beyotime, China) in the specified dual-luciferase reporter assay system (Thermo Scientific Fluoroskan FL, Thermo Fisher, Americia) according to the manufacturer’s protocol. The activities of ISL1 gene promoter were assessed by the ratio of firefly luciferase activity to renilla luciferase activity. All experiments were performed in three independent replicates.

2.5. Nuclear Protein Extraction and Electrophoretic Mobility Shift Assay

To investigate the effect of variants within the promoter region of ISL1 gene on the binding ability of transcription factors, electrophoretic mobility shift assay (EMSA) was performed with Chemiluminescent EMSA Kit (Beyotime, Shanghai China). Biotinylated double-stranded oligonucleotides based on the wild-type or variant-type sequence in ISL1 gene promoter were utilized as probes. Biotin-labeled oligonucleotides for the EMSA experiment are shown in Table 1. Nuclear protein extracts from HL-1 cells and HEK-293 cells were prepared using the Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime, Shanghai, China) and protein concentrations were determined with the Enhanced BCA Protein Assay Kit (Beyotime, Shanghai, China). The DNA-protein binding reaction with an equal amount of probe and nuclear extracts was performed at room temperature in 20 min. The mixture of DNA and protein mixed with sample loading buffer was then added to the sample wells of 4% polyacrylamide gel and transferred onto a nylon membrane (Beyotime, Shanghai, China). The oligonucleotides were cross-linked to the membrane using the UV light. Finally, biotinylated oligonucleotides were detected by chemiluminescence using the Chemiluminescent EMSA kit (Beyotime, Shanghai, China).

2.6. Transcription Factor Binding Sites Prediction

The online JASPAR database (http://jaspar.genereg.net, accessed on 7 August 2022) was used to analyze whether ISL1 promoter region variants would alter the putative transcription factor binding site (TFBS). The relative profile score threshold was set at 85% in the JASPAR database.

2.7. Statistical Analysis

For continuous variables, results are expressed as mean ± SD. Non-parametric tests for independent samples were used to analyze the quantitative data and to calculate the statistical significance of the experimental results. The burden test for rare variants was performed by using SKAT package in R. The model parameters and residuals were calculated by ‘SKAT-Null-Model-MomentAdjust’ function for binary triat. ‘SKATBinary’ function was used for the burden test of a group of several rare variants [20]. The level of significance was set at p < 0.05. SPSS 19.0 software (IBM) was used for all statistical analyses.

3. Results

3.1. The Variants Identified in TOF Patients and Healthy Controls

This study recruited 601 participants, including 308 TOF patients and 293 healthy controls (Figure 1). A total of eleven variants in ISL1 gene promoter were identified by Sanger sequencing in these study subjects. The locations and details of these variants are presented in Figure 2A and Table 2. Among those eleven variants found in this study, one novel heterozygous variant [g.5085G > A] and three single-nucleotide variants (SNVs) [g.4581A > G(rs1747215641), g.4630G > A(rs1264694566), g.5221C > T(rs1484364688)] were found only in patients with TOF. The sequencing chromatograms of these four variants are presented in Figure 2B. In addition, the allele frequency of these four variants is less than 0.0001 in the NCBI ALFA database and GnomAD database and two of them [g.5085G > A, g.5221C > T(rs1484364688)] have 0 allele frequency (Table 2). Further, one of these four variants [g.4630G > A(rs1264694566)] was found in two patients. The above three SNVs and the novel variant [g.5085G > A] were further validated for cellular functional experiment.
There were four variants [g.4213C > T(rs36216895), g.4457C > G(rs6899279), g.4613G > A(rs142427249), g.5057A > G(rs3762977)] that were found in both TOF patients and control subjects (Table 2). Three variants [g.4184 T > C, g.4720 A > G (rs1200176972), g.5006 A > C] were only found in the healthy controls. These seven variants were excluded from the further study [21].

3.2. Functional Analysis of the Variants via Dual-Luciferase Reporter Assay

To further investigate the effect of variants on the transcriptional activity of the ISL1 promoter, different reporter plasmids including a null pGL3-basic (negative control), wild-type ISL1 gene promoter (pGL3-WT), and variant-type ISL1 gene promoter (pGL3-4581G, pGL3-4630A, pGL3-5085A, pGL3-5221T) were constructed and transfected into the HL-1 cells, respectively. The dual-luciferase activities were assayed and relative activity of wild-type and variant ISL1 gene promoter was examined.
As illustrated in Figure 3A, dual-luciferase activity indicated that among the four expression vectors carrying genetic variants, three variants (4581G, 4630A, 5085A) significantly decreased the transcriptional activities compared with the wild-type (p < 0.05). In contrast, the rest one variant (5221T) did not significantly alter the transcriptional activity of the ISL1 gene promoter (p > 0.05). The echocardiography of patients with variants is displayed in Figure 3B.

3.3. The Binding for Transcription Factors Affected by the Variants

To experimentally investigate whether the binding ability of transcription factor was affected by the variants identified only in TOF patients, EMSA was performed with wild-type or variant biotinylated oligonucleotides. The above three variants (4581G, 4630A, 5085A) markedly affected the binding of transcription factor in both HL-1 and HEK-293 cells, as shown in Figure 4 (presented with arrows). In addition, similar to the result in dual-luciferase reporter assay, the variant (5221T) did not affect the binding of transcription factor (data not shown).

3.4. Variant-Affected Putative Binding Sites for Transcription Factor

The online JASPAR database was fully utilized to further explore whether the binding sites for transcription factors were affected by variants identified in the ISL1 gene promoter. As summarized in Table 3, the putative binding sites for transcription factor may be disrupted or created by variants identified in patients with TOF. The variant g.4581G may create new binding sites for ZEB1, NKx3-1, and TEAD3, and disrupt the binding sites for FOXL1, RHOXF1, and GATA5. Among these TFBSs, the mutations of GATA5 have been reported in sporadic TOF patients. SOX9, a downstream target gene of ISL1 gene, is regulated by ISL1 gene through ISL1-Fgf10-SOX9 pathway. In combination with the experimental results, the analysis by the JASPAR database, and literature reports, we established a schema (Figure 5) to illustrate the possible effect of the variants in ISL1 gene promoter region and the roles of genes and pathways associated with ISL1 gene in the etiology of TOF.

4. Discussion

The current study for the first time identified that (1) there are four variants within the ISL1 gene promoter that are identified only in the TOF patients with 0 incidence in the control subjects and one of them (g.5085G > A) is novel; (2) three of these four variants (g.4581A > G, g.4630G > A, g.5085G > A) significantly altered the transcriptional activity of the ISL1 gene promoter at cellular functional level; and (3) these three variants (g.4581A > G, g.4630G > A, g.5085G > A) causing functional changes evidently affected the binding of transcription factors verified by EMSA experiments. Therefore, these ISL1 gene promoter variants (g.4581A > G, g.4630G > A, g.5085G > A) likely contribute to the development of TOF as risk factors.
The human ISL1 gene, mapped to chromosome 5q11.1 and comprised with 349 amino acids, was first identified and cloned during the study of the regulation of insulin gene expression [22]. Recently, it has been reported that ISL1 plays an important role in the regulation of the nervous system, the pancreas and cardiac development [23]. ISL1 gene can be specifically expressed in cardiac progenitor cells. Decreased expression of the ISL1 gene has been reported to be associated with many cardiovascular diseases in human, such as CHDs [24], atrial fibrillation [25], and dilated cardiomyopathy [26]. It has been reported that the copy number variations of ISL1 gene is related to the etiology of TOF [27]. The expression of many genes is regulated through the binding of various transcription factors to promoter sequences [19]. To date, variants at the promoter region of human ISL1 gene have not been illustrated in detail. Based on the above reports concerning ISL1 gene, we hypothesized that variants in the ISL1 gene promoter may affect the normal gene activation and lead to the disease. In this study, we analyzed the promoter of ISL1 gene in patients with TOF and the variants were identified. The transcriptional activity analysis and EMSA results performed in the present study have clearly demonstrated that three variants discovered in ISL1 gene promoter region notably changed the transcriptional activity, providing support for our hypothesis. The EMSA is an experiment that may demonstrate the interference (enhancement or suppression) of the binding affinity between transcription factors and promoter by variants. Different variants may affect the binding affinity for various transcription factors (see Table 3 for details) at the different levels, as shown in Figure 4.
It has been reported that variants in the gene promoter region create or disrupt TFBS at the promoter, affecting the transcriptional regulation of genes and putatively contributing to the development of disease. In the present study, three variants discovered in the ISL1 gene promoter with altered cellular function were analyzed with bioinformatics through JASPAR database. We discovered that the variant g.4581A > G may disrupt the potential binding sites for GATA5, a member of the GATA family transcription factor that plays an essential role in the development of embryonic heart. Importantly, the GATA5 loss-of-function mutation is associated with TOF [28]. In addition, the binding sites for SRY (sex determining region Y)-box9(SOX9) may be created by the variant g.4630G > A. The ISL1–Fgf10–SOX9 network is a pivotal pathway in the embryonic lung branching morphogenesis and cell differentiation and SOX9 is regulated by ISL1 [29]. Further investigation to explore the relationship among ISL1 gene promoter variants and these transcription factors is required.
Apart from GATA5 and SOX9, among the 32 transcription factors listed in Table 3, at least 16 factors are closely related to the development of the heart. All those genes may be further studied with regard to the interaction to ISL1 promoter in order to reveal the correlation among them and the pathological role in the development of TOF.
Taken together with the results from the present study, the JASPAR database analysis, and the previous reports, we established a schema (Figure 5) to summarize the possible role of ISL1 gene promoter region variants in the development of TOF. Firstly, variants reduce ISL1 gene promoter activity, contributing to the low expression of ISL1. Consequently, the low expression of ISL1 may be involved in the formation of TOF. In addition, the activity of TOF-related genes, such as GATA4 [30], TBX5 [31], and TBX20 [32], may be decreased by the low expression of ISL1 [33]. Moreover, the low expression of ISL1 may also lead to the reduced-expression of vascular endothelial growth factor related signaling gene FOXO1, which is interacted with ISL1 and involved in the pathogenesis of TOF [34,35,36]. Finally, the low level of ISL1 may reduce the expression of HAND2 and SHH in the HAND2-SHH pathway, which is involved in the development of TOF [37,38,39].
There are some limitations in this study. The exact role of genotype–phenotype relationship needs to be further established. The precise role of the discovered ISL1 promoter variants in the development of TOF requires further in vivo animal experiments. In addition, CHDs are developed with the interaction of related genes and the interaction between ISL1 and other genes shown in Figure 5 needs to be further verified.

5. Conclusions

In conclusion, our study for the first time identified genetic variants in the promoter region of ISL1 gene in patients with TOF. Furthermore, the variants were demonstrated to significantly alter the ISL1 gene expression by the cellular functional experiment. Further EMSA experiment and bioinformatic analysis also demonstrated that these variants likely affect the binding of transcription factors and may contribute to the development of TOF as risk factors. Therefore, this study may provide new insights into the understanding of the promoter region and the genetic etiology involved in the development of CHD.

Author Contributions

Conceptualization, G.-W.H.; methodology, X.-Y.Y., H.-X.C., Z.C., Q.Y., J.H. and G.-W.H.; validation, X.-Y.Y., H.-X.C., Z.C. and G.-W.H.; formal analysis, X.-Y.Y., H.-X.C. and G.-W.H.; investigation, X.-Y.Y., H.-X.C. and G.-W.H.; resources, G.-W.H.; data curation, X.-Y.Y., H.-X.C., Z.C., Q.Y. and G.-W.H.; writing—original draft, X.-Y.Y.; writing—review & editing, G.-W.H.; supervision, G.-W.H.; project administration, G.-W.H. and J.H.; funding acquisition, G.-W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China [82170353 & 81870288]; Tianjin Science and Technology Commission [22ZYQYSY00020]; the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences [2020-PT310-007]; Tianjin Municipal Health Commission [KJ20071]; and TEDA International Cardiovascular Hospital Internal Grant [2021-ZX-002], and Tianjin Key Medical Discipline (Specialty) Construction Project (TJYXZDXK-019A).

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of Tianjin TEDA International Cardiovascular Hospital (clinical research ethics review No. 2021-0715-4).

Informed Consent Statement

Written informed consent has been obtained from the patient(s) to publish this paper.

Data Availability Statement

The individual SNP numbers are given in Table 2. The genetic variants described in this manuscript are available at https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/snp/ (accessed on 29 June 2022). Data supporting the findings of this study are available upon reasonable request from the corresponding authors.

Acknowledgments

We thank the patients and their family members for their collaboration. We sincerely thank Zhao-Nan Yu and Hong-Wei Chen of Hangzhou D.A. Medical Laboratory, Hangzhou, China for their contribution in Gene Burden Test. The assistance of surgical and nursing staff at the Division of Pediatric Cardiac Surgery, Department of Cardiovascular Surgery is gratefully acknowledged.

Conflicts of Interest

No conflict of interest, financial or otherwise, are declared by the authors.

References

  1. Marelli, A.J.; Mackie, A.S.; Ionescu-Ittu, R.; Rahme, E.; Pilote, L. Congenital heart disease in the general population: Changing prevalence and age distribution. Circulation 2007, 115, 163–172. [Google Scholar] [CrossRef]
  2. Ferencz, C.; Rubin, J.D.; McCarter, R.J.; Brenner, J.I.; Neill, C.A.; Perry, L.W.; Hepner, S.I.; Downing, J.W. Congenital heart disease: Prevalence at livebirth. The Baltimore-Washington Infant Study. Am. J. Epidemiol. 1985, 121, 31–36. [Google Scholar] [CrossRef]
  3. Starr, J.P. Tetralogy of fallot: Yesterday and today. World J. Surg. 2010, 34, 658–668. [Google Scholar] [CrossRef] [PubMed]
  4. Villafane, J.; Feinstein, J.A.; Jenkins, K.J.; Vincent, R.N.; Walsh, E.P.; Dubin, A.M.; Geva, T.; Towbin, J.A.; Cohen, M.S.; Fraser, C.; et al. Hot topics in tetralogy of Fallot. J. Am. Coll. Cardiol. 2013, 62, 2155–2166. [Google Scholar] [CrossRef]
  5. Chen, H.X.; Yang, Z.Y.; Hou, H.T.; Wang, J.; Wang, X.L.; Yang, Q.; Liu, L.; He, G.W. Novel mutations of TCTN3/LTBP2 with cellular function changes in congenital heart disease associated with polydactyly. J. Cell. Mol. Med. 2020, 24, 13751–13762. [Google Scholar] [CrossRef]
  6. Page, D.J.; Miossec, M.J.; Williams, S.G.; Monaghan, R.M.; Fotiou, E.; Cordell, H.J.; Sutcliffe, L.; Topf, A.; Bourgey, M.; Bourque, G.; et al. Whole Exome Sequencing Reveals the Major Genetic Contributors to Nonsyndromic Tetralogy of Fallot. Circ. Res. 2019, 124, 553–563. [Google Scholar] [CrossRef]
  7. He, G.W.; Maslen, C.L.; Chen, H.X.; Hou, H.T.; Bai, X.Y.; Wang, X.L.; Liu, X.C.; Lu, W.L.; Chen, X.X.; Chen, W.D.; et al. Identification of Novel Rare Copy Number Variants Associated with Sporadic Tetralogy of Fallot and Clinical Implications. Clin. Genet. 2022, 102, 391–403. [Google Scholar] [CrossRef] [PubMed]
  8. Han, S.; Zhang, Y.Y.; Meng, M.Y.; Hou, Z.L.; Meng, P.; Zhao, Y.Y.; Gao, H.; Tang, J.; Liu, Z.; Yang, L.L.; et al. Generation of human iPSC line from a patient with Tetralogy of Fallot, YAHKMUi001-A, carrying a mutation in TBX1 gene. Stem Cell Res. 2020, 42, 101687. [Google Scholar] [CrossRef]
  9. Goodship, J.A.; Hall, D.; Topf, A.; Mamasoula, C.; Griffin, H.; Rahman, T.J.; Glen, E.; Tan, H.; Palomino Doza, J.; Relton, C.L.; et al. A common variant in the PTPN11 gene contributes to the risk of tetralogy of Fallot. Circ. Cardiovasc. Genet. 2012, 5, 287–292. [Google Scholar] [CrossRef]
  10. Fan, S.H.; Shen, Z.Y.; Xiao, Y.M. Functional polymorphisms of the neuropilin 1 gene are associated with the risk of tetralogy of Fallot in a Chinese Han population. Gene 2018, 653, 72–79. [Google Scholar] [CrossRef] [PubMed]
  11. Cai, C.L.; Liang, X.Q.; Shi, Y.Q.; Chu, P.H.; Pfaff, S.L.; Chen, J.; Evans, S. ISL1 Identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 2003, 5, 877–889. [Google Scholar] [CrossRef] [PubMed]
  12. Laugwitz, K.L.; Moretti, A.; Lam, J.; Gruber, P.; Chen, Y.; Woodard, S.; Lin, L.Z.; Cai, C.L.; Lu, M.M.; Reth, M.; et al. Postnatal ISL1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature 2005, 433, 647–653. [Google Scholar] [CrossRef]
  13. Moretti, A.; Caron, L.; Nakano, A.; Lam, J.T.; Bernshausen, A.; Chen, Y.; Qyang, Y.; Bu, L.; Sasaki, M.; Martin-Puig, S.; et al. Multipotent embryonic ISL1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 2006, 127, 1151–1165. [Google Scholar] [CrossRef]
  14. Zheng, S.Q.; Chen, H.X.; Liu, X.C.; Yang, Q.; He, G.W. Identification of variants of ISL1 gene promoter and cellular functions in isolated ventricular septal defects. Am. J. Physiol. Cell Physiol. 2021, 321, C443–C452. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, Z.; Song, H.M.; Wang, F.; Zhao, C.M.; Huang, R.T.; Xue, S.; Li, R.G.; Qiu, X.B.; Xu, Y.J.; Liu, X.Y.; et al. A new ISL1 loss-of-function mutation predisposes to congenital double outlet right ventricle. Int. Heart J. 2019, 60, 1113–1122. [Google Scholar] [CrossRef] [PubMed]
  16. Ma, L.; Wang, J.; Li, L.; Qiao, Q.; Di, R.M.; Li, X.M.; Xu, Y.J.; Zhang, M.; Li, R.G.; Qiu, X.B.; et al. ISL1 loss-of-function mutation contributes to congenital heart defects. Heart Vessels 2019, 34, 658–668. [Google Scholar] [CrossRef] [PubMed]
  17. Stevens, K.N.; Hakonarson, H.; Kim, C.E.; Doevendans, P.A.; Koeleman, B.P.; Mital, S.; Raue, J.; Glessner, J.T.; Coles, J.G.; Moreno, V.; et al. Common variation in ISL1 confers genetic susceptibility for human congenital heart disease. PLoS ONE 2010, 5, e10855. [Google Scholar] [CrossRef]
  18. Zeng, Z.H.; Chen, H.X.; Liu, X.C.; Yang, Q.; He, G.W. Functional significance of novel variants of the MEF2C gene promoter in congenital ventricular septal defects. Am. J. Med. Genet. A 2022, 188, 2397–2405. [Google Scholar] [CrossRef]
  19. de Vooght, K.M.; van Wijk, R.; van Solinge, W.W. Management of gene promoter mutations in molecular diagnostics. Clin. Chem. 2009, 55, 698–708. [Google Scholar] [CrossRef]
  20. Lee, S.; Fuchsberger, C.; Kim, S.; Scott, L. An efficient resampling method for calibrating single and gene-based rare variant association analysis in case-control studies. Biostatistics 2016, 17, 1–15. [Google Scholar] [CrossRef] [Green Version]
  21. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed]
  22. Karlsson, O.; Thor, S.; Norberg, T.; Ohlsson, H.; Edlund, T. Insulin gene enhancer binding protein Isl-1 is a member of a novel class of proteins containing both a homeo- and a Cys-His domain. Nature 1990, 344, 879–882. [Google Scholar] [CrossRef] [PubMed]
  23. Hao, A.; Novotny-Diermayr, V.; Bian, W.; Lin, B.; Lim, C.P.; Jing, N.; Cao, X. The LIM/homeodomain protein Islet1 recruits Janus tyrosine kinases and signal transducer and activator of transcription 3 and stimulates their activities. Mol. Biol. Cell 2005, 16, 1569–1583. [Google Scholar] [CrossRef]
  24. Osoegawa, K.; Schultz, K.; Yun, K.; Mohammed, N.; Shaw, G.M.; Lammer, E.J. Haploinsufficiency of insulin gene enhancer protein 1 (ISL1) is associated with d-transposition of the great arteries. Mol. Genet. Genom. Med. 2014, 2, 341–351. [Google Scholar] [CrossRef]
  25. Wu, S.H.; Wang, X.H.; Xu, Y.J.; Gu, J.N.; Yang, C.X.; Qiao, Q.; Guo, X.J.; Guo, Y.H.; Qiu, X.B.; Jiang, W.F.; et al. ISL1 loss-of-function variation causes familial atrial fibrillation. Eur. J. Med. Genet. 2020, 63, 104029. [Google Scholar] [CrossRef]
  26. Xu, Y.J.; Wang, Z.S.; Yang, C.X.; Di, R.M.; Qiao, Q.; Li, X.M.; Gu, J.N.; Guo, X.J.; Yang, Y.Q. Identification and Functional Characterization of an ISL1 Mutation Predisposing to Dilated Cardiomyopathy. J. Cardiovasc. Transl. Res. 2019, 12, 257–267. [Google Scholar] [CrossRef] [PubMed]
  27. Bansal, V.; Dorn, C.; Grunert, M.; Klaassen, S.; Hetzer, R.; Berger, F.; Sperling, S.R. Outlier-based identification of copy number variations using targeted resequencing in a small cohort of patients with Tetralogy of Fallot. PLoS ONE 2014, 9, e85375. [Google Scholar] [CrossRef]
  28. Wei, D.; Bao, H.; Liu, X.Y.; Zhou, N.; Wang, Q.; Li, R.G.; Xu, Y.J.; Yang, Y.Q. GATA5 loss-of-function mutations underlie tetralogy of fallot. Int. J. Med. Sci. 2013, 10, 34–42. [Google Scholar] [CrossRef]
  29. Dai, H.T.; Zhu, M.L.; Li, W.Y.; Si, G.H.; Xing, Y.M. Activation of PI3K/p110alpha in the Lung Mesenchyme Affects Branching Morphogenesis and Club Cell Differentiation. Front. Cell Dev. Biol. 2022, 10, 880206. [Google Scholar] [CrossRef]
  30. Yang, Y.Q.; Gharibeh, L.; Li, R.G.; Xin, Y.F.; Wang, J.; Liu, Z.M.; Qiu, X.B.; Xu, Y.J.; Xu, L.; Qu, X.K.; et al. GATA4 loss-of-function mutations underlie familial tetralogy of fallot. Hum. Mutat. 2013, 34, 1662–1671. [Google Scholar] [CrossRef]
  31. Ma, J.; Chen, S.Y.; Hao, L.L.; Sheng, W.; Chen, W.C.; Ma, X.J.; Zhang, B.W.; Ma, D.; Huang, G.Y. Hypermethylation-mediated down-regulation of lncRNA TBX5-AS1:2 in Tetralogy of Fallot inhibits cell proliferation by reducing TBX5 expression. J. Cell Mol. Med. 2020, 24, 6472–6484. [Google Scholar] [CrossRef]
  32. Huang, R.T.; Wang, J.; Xue, S.; Qiu, X.B.; Shi, H.Y.; Li, R.G.; Qu, X.K.; Yang, X.X.; Liu, H.; Li, N.; et al. TBX20 loss-of-function mutation responsible for familial tetralogy of Fallot or sporadic persistent truncus arteriosus. Int. J. Med. Sci. 2017, 14, 323–332. [Google Scholar] [CrossRef]
  33. Gao, R.; Liang, X.Q.; Cheedipudi, S.; Cordero, J.; Jiang, X.; Zhang, Q.Q.; Caputo, L.; Günther, S.; Kuenne, C.; Ren, Y.G.; et al. Pioneering function of ISL1 in the epigenetic control of cardiomyocyte cell fate. Cell Res. 2019, 29, 486–501. [Google Scholar] [CrossRef] [PubMed]
  34. Kappen, C.; Salbaum, J.M. Identification of regulatory elements in the ISL1 gene locus. Int. J. Dev. Biol. 2009, 53, 935–946. [Google Scholar] [CrossRef] [PubMed]
  35. Waclaw, R.R.; Ehrman, L.A.; Merchan-Sala, P.; Kohli, V.; Nardini, D.; Campbell, K. Foxo1 is a downstream effector of ISL1 in direct pathway striatal projection neuron development within the embryonic mouse telencephalon. Mol. Cell. Neurosci. 2017, 80, 44–51. [Google Scholar] [CrossRef]
  36. Reuter, M.S.; Jobling, R.; Chaturvedi, R.R.; Manshaei, R.; Costain, G.; Heung, T.; Curtis, M.; Hosseini, S.M.; Liston, E.; Lowther, C.; et al. Haploinsufficiency of vascular endothelial growth factor related signaling genes is associated with tetralogy of Fallot. Genet. Med. 2019, 21, 1001–1007. [Google Scholar] [CrossRef] [PubMed]
  37. Itou, J.; Kawakami, H.; Quach, T.; Osterwalder, M.; Evans, S.M.; Zeller, R.; Kawakami, Y. Islet1 regulates establishment of the posterior hindlimb field upstream of the Hand2-Shh morphoregulatory gene network in mouse embryos. Development 2012, 139, 1620–1629. [Google Scholar] [CrossRef]
  38. Lu, C.X.; Gong, H.R.; Liu, X.Y.; Wang, J.; Zhao, C.M.; Huang, R.T.; Xue, S.; Yang, Y.Q. A novel HAND2 loss-of-function mutation responsible for tetralogy of Fallot. Int. J. Mol. Med. 2016, 37, 445–451. [Google Scholar] [CrossRef] [PubMed]
  39. Washington Smoak, I.; Byrd, N.A.; Abu-Issa, R.; Goddeeris, M.M.; Anderson, R.; Morris, J.; Yamamura, K.; Klingensmith, J.; Meyers, E.N. Sonic hedgehog is required for cardiac outflow tract and neural crest cell development. Dev. Biol. 2005, 283, 357–372. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The flow chart of this study. A total of 601 subjects were enrolled in this study. Then we performed total DNA extraction, DNA sequence analysis, cellular functional analysis, electrophoretic mobility shift assay, and transcription factor binding sites prediction to test our hypothesis. TOF, Tetralogy of Fallot.
Figure 1. The flow chart of this study. A total of 601 subjects were enrolled in this study. Then we performed total DNA extraction, DNA sequence analysis, cellular functional analysis, electrophoretic mobility shift assay, and transcription factor binding sites prediction to test our hypothesis. TOF, Tetralogy of Fallot.
Biomolecules 13 00358 g001
Figure 2. Identified variants of ISL1 gene. (A) The locations of eleven variants identified within the ISL1 gene promoter region in this study. The genetic variants were named according to the genomic DNA sequence of human ISL1 gene (Genbank accession number NG_023040.1). (B) The sequencing chromatograms of the variants only found in patients with TOF. For all heterozygous variants [g.4581A > G(rs1747215641), g.4630G > A(rs1264694566), g.5085G > A, g.5221C > T(rs1484364688)], the top panel shows the wild−type and the bottom panel shows the variant-type. The variant site is labeled with the red line and arrow.
Figure 2. Identified variants of ISL1 gene. (A) The locations of eleven variants identified within the ISL1 gene promoter region in this study. The genetic variants were named according to the genomic DNA sequence of human ISL1 gene (Genbank accession number NG_023040.1). (B) The sequencing chromatograms of the variants only found in patients with TOF. For all heterozygous variants [g.4581A > G(rs1747215641), g.4630G > A(rs1264694566), g.5085G > A, g.5221C > T(rs1484364688)], the top panel shows the wild−type and the bottom panel shows the variant-type. The variant site is labeled with the red line and arrow.
Biomolecules 13 00358 g002
Figure 3. The result of the dual-luciferase reporter assay and the echocardiography of patients with these three variants. (A) Relative transcriptional activity of wild-type and variant-type ISL1 gene promoter in HL-1 cells. The negative control was empty vector pGL3-basic and the transcriptional activity of the wild-type ISL1 gene promoter was set as 100%. The quantitative data are presented as mean ± SD based on three independent experiments. * p < 0.05; ** p < 0.01. (B) The doppler echocardiography shows TOF in patients with variants of [g.4581A > G(rs1747215641), g.4630G > A(rs1264694566), g.5085G > A], showing the malalignment ventricular septal defect and aortic overriding. The ventricular septal defect is indicated with arrows. AO, aorta; RA, right atrium; RV, right ventricle; VSD, ventricular septal defect.
Figure 3. The result of the dual-luciferase reporter assay and the echocardiography of patients with these three variants. (A) Relative transcriptional activity of wild-type and variant-type ISL1 gene promoter in HL-1 cells. The negative control was empty vector pGL3-basic and the transcriptional activity of the wild-type ISL1 gene promoter was set as 100%. The quantitative data are presented as mean ± SD based on three independent experiments. * p < 0.05; ** p < 0.01. (B) The doppler echocardiography shows TOF in patients with variants of [g.4581A > G(rs1747215641), g.4630G > A(rs1264694566), g.5085G > A], showing the malalignment ventricular septal defect and aortic overriding. The ventricular septal defect is indicated with arrows. AO, aorta; RA, right atrium; RV, right ventricle; VSD, ventricular septal defect.
Biomolecules 13 00358 g003
Figure 4. EMSA of biotin-labeled oligonucleotide-containing variants with nuclear extracts of HL-1 cells and HEK-293 cells. The amount of the labeled probe in VT and WT lane was the same. Free probe is at the bottom. The different binding for transcription factors between WT and VT is marked with arrows. Sequencing chromatograms of the three variants are represented at the right panel. WT, wild type, VT, variant type.
Figure 4. EMSA of biotin-labeled oligonucleotide-containing variants with nuclear extracts of HL-1 cells and HEK-293 cells. The amount of the labeled probe in VT and WT lane was the same. Free probe is at the bottom. The different binding for transcription factors between WT and VT is marked with arrows. Sequencing chromatograms of the three variants are represented at the right panel. WT, wild type, VT, variant type.
Biomolecules 13 00358 g004
Figure 5. Schema describing the role of ISL1 gene promoter region variants in the development of cardiac defect based on the results from the present study, the JASPAR database analysis, and the previous reports. Variants reduced ISL1 gene promoter activity by disrupting and creating the TFBS (total of 32 [see Table 3]; 18 of them are involved in the heart development that are listed in the figure), contributing to the low expression of ISL1. Consequently, the low expression of ISL1 may be directly involved in the formation of TOF and may reduce the expression of downstream target genes and pathway, which are related to the formation of TOF.
Figure 5. Schema describing the role of ISL1 gene promoter region variants in the development of cardiac defect based on the results from the present study, the JASPAR database analysis, and the previous reports. Variants reduced ISL1 gene promoter activity by disrupting and creating the TFBS (total of 32 [see Table 3]; 18 of them are involved in the heart development that are listed in the figure), contributing to the low expression of ISL1. Consequently, the low expression of ISL1 may be directly involved in the formation of TOF and may reduce the expression of downstream target genes and pathway, which are related to the formation of TOF.
Biomolecules 13 00358 g005
Table 1. List of primers and oligonucleotides sequences for EMSA in this study.
Table 1. List of primers and oligonucleotides sequences for EMSA in this study.
Primers NameDNA Sequences 5’-3’LocationPosition
PCR and sequencing primers
  ISL1-F15’-CTGTCTTTGGGAGACCGTAACA-3’4039−1286
  ISL1-R15’-TGCCAATGCTGAAAGAGCCG-3’5436+111
Primers containing restriction sites
  ISL1-KpnI ab5’-(KpnI)-CGGGGTACCCTGTCTTTGGGAGACCGTAACA-3’4039−1286
  ISL1-BglII ab5’-(BglII)-CAAGATCTTGCCAATGCTGAAAGAGCCGG-3’5436+111
The double-stranded biotinylated oligonucleotides for the EMSA
Sequence variantsOligonucleotides sequences
  g.4581 A > G5’-CACCCAACGTTTTTAG(A/G)TATCTGAGGTTGGGG-3’
  g.4630 G > A5’-TAGCAGTCAGGAAACA(G/A)TTCCTCTGGGTTTAATC-3’
  g.5085 G > A5’-AACGGCCTGAGCCCC(G/A)AGCAAGTTGCCTCGGG-3’
  g.5221 C > T5’-TGGGCGCGGCGCT(C/T)GCCTGACGTCCCCG-3’
Note: The PCR primers are designed based on the genomic DNA sequence of the ISL1 gene (NG_023040.1). The transcription start site is at the position of 5325 (+1). Abbreviations: F, forward; R, reverse; PCR, polymerase chain reaction; EMSA, electrophoretic mobility shift assay. a Restriction sites are underlined. b Protective bases are marked in bold. The underline indicates the restriction endonuclease site
Table 2. Variants within the ISL1 gene promoter in TOF patients and controls.
Table 2. Variants within the ISL1 gene promoter in TOF patients and controls.
VariantsTOFControlsBurden Test (p-Value)Position aGenotypesAllele Frequency
Frequency in Control = 0 (Further Validation)GnomADEast Asian
g.4581A > G(rs1747215641)100.04−744AGG = 0.00007G = 0.00 *G = 0.0000 #
g.4630G > A(rs1264694566)20−695GAA = 0.000014A = 0.00 *A = 0.0003 #
g.5085G > A10−240GANone--
g.5221C > T(rs1484364688)10−104CTNoneT = 0.00 *-
Frequency in Control ≠ 0 (No Further Validation)GnomAD
g.4213C > T(rs36216895)14240.29−1112CTNone
g.4457C > G(rs6899279)1424−868CGG = 0.174
g.4613G > A(rs142427249)41−712GAA = 0.00013
g.5057A > G(rs3762977)1424−268AGG = 0.149
Abbreviations: -, not applicable; TOF, Tetralogy of Fallot. a Variants are located upstream (−) to the transcription start site of the ISL1 gene at 5325 of NG_023040.1. * The Allele frequency of East Asian in ALFA database (Version: 20201027095038). # The Allele frequency of East Asian in GnomAD database (Accession: PRJNA398795).
Table 3. Predicted binding sites for transcription factors and promoter activity affected by variants.
Table 3. Predicted binding sites for transcription factors and promoter activity affected by variants.
VariantsBinding Sites for Transcription FactorsPromoter Activity
CreateDisrupt
g.4581A > GZEB1, NKx3-1, TEAD3FOXL1, RHOXF1, GATA5
g.4630G > ASRY, En1, FOXO3, FOXG1, TEAD4, SOX5, SOX9, FOXD1, FOXJ2, FOXN3, SOX15, SOX8, HOXA4, GSX2, ETV2::DRGXELK4, FOXP1, ETV6, PRDM4
g.5085G > AEHF, TcfL5Ahr::Arnt, TFAP2E
g.5221C > T-TFAP2A, NFIX, HES1No change
Abbreviations: -, not applicable.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yin, X.-Y.; Chen, H.-X.; Chen, Z.; Yang, Q.; Han, J.; He, G.-W. Genetic Variants of ISL1 Gene Promoter Identified from Congenital Tetralogy of Fallot Patients Alter Cellular Function Forming Disease Basis. Biomolecules 2023, 13, 358. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13020358

AMA Style

Yin X-Y, Chen H-X, Chen Z, Yang Q, Han J, He G-W. Genetic Variants of ISL1 Gene Promoter Identified from Congenital Tetralogy of Fallot Patients Alter Cellular Function Forming Disease Basis. Biomolecules. 2023; 13(2):358. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13020358

Chicago/Turabian Style

Yin, Xiu-Yun, Huan-Xin Chen, Zhuo Chen, Qin Yang, Jun Han, and Guo-Wei He. 2023. "Genetic Variants of ISL1 Gene Promoter Identified from Congenital Tetralogy of Fallot Patients Alter Cellular Function Forming Disease Basis" Biomolecules 13, no. 2: 358. https://0-doi-org.brum.beds.ac.uk/10.3390/biom13020358

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop