Take a look at the Recent articles

SNPs, transcriptional factor binding sites and disease

Norman E. Buroker

Department of Pediatrics, University of Washington, Seattle, USA

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

DOI: 10.15761/BGG.1000132

Article
Article Info
Author Info
Figures & Data

Abstract

Single nucleotide polymorphisms (SNPs) located in non-coding regions of the human genome that have been found to be significantly associated with disease or sickness may affect gene regulation by altering binding sites for transcription factors (TF). There have been several reports to identify such punitive changes in transcriptional factor binding sites (TFBS) created by SNP alleles. A few of these reports have been reviewed for the Activating Transcription Factor 3 (ATF3), v-Akt Murine Thymoma Viral Oncogene Homolog 3 (AKT3), Adrenergic Beta Receptor Kinase 1 (G Protein-Coupled Receptor Kinase 2) (ADRBK1), Type 2 Deiodeinase gene (DIO2), Endothetal Per-Arnt-Sim (PAS) domain protein 1 (EPAS1), Lysosomal Acid Lipase A (LIPA), Peroxisome Proliferator-activated Receptors (PPARα/δ/γ), Signal Transducer and Activator of Transcription 4 (STAT4), Thromboxane A2 Receptor (TBXA2R) and Vascular Endothelial Growth Factor (VEGF)-A genes. In each report the SNP alleles created punitive alterations in TFBS that could explain associated disease(s) or changes in a human condition.

Key words

gene regulation, non-coding DNA, SNPs, TFBS, human disease

Introduction

The human genome consists of approximately 2% coding DNA that makes up all of the genes and 98% non-coding DNA which comprises the structural, recombinatorial, origin of replication elements and transcriptional regulatory sequences [1]. Genome-wide association studies (GWAS) indicate that approximately 93% of disease or trait-predisposing single nucleotide polymorphisms (SNPs) believed to be associated with gene regulation fall in these non-coding regions [2-4]. SNPs that have been found to be significantly associated with disease are considered to be risk-associated and cause changes in gene expression [4]. A single nucleotide change in a transcriptional factor motif sequence or a transcriptional factor binding site (TFBS) may affect the process of gene regulation [5-7]. To date, a number of computational approaches and databases have been established to help identify transcriptional factor (TF) motif changes created by SNPs which have been associated with disease or a change in health conditions [8-18]. Of these approaches, there have been several investigations made using the JASPAR CORE [16,17] and ConSite [18] databases to identify alterations in TFBS created by SNPs from a single gene that have been shown to be significantly associated with human disease(s) or illness [19-30]. Using this computational approach has often identified a punitive TFBS created by a causative SNP allele significantly associated with the disease or illness. In an effort to verify which SNP allele is responsible for the significant association, the HaploReg [14] and RegulomeDB [15] databases were screened for the SNP allele substitution and altered TFBS that would most probably affect gene regulation [31]. Some of the research using the JASPAR CORE [16,17] and ConSite [18] databases are reviewed in this manuscript.

Activating Transcription Factor 3 (ATF3)

The activating transcription factor 3 (ATF3) gene is a member of the activating transcription factor/cAMP responsive element binding (CREB) protein family of transcription factors. This gene located on chromosome one has been shown to be up-regulated during sexual differentiation [32]. The gene has three SNPs (rs3125289 (C/T), rs1877474 (C/T) and rs11119982 (C/T) which span a 16 kb region of intron one have been independently found to be significantly associated with the risk of hypospadias [33]. Hypospadias is a congenital birth defect in which the opening of the urethra is on the underside of the penis instead of the normal position in the head. The rs3125289 ATF3-T allele creates the punitive fork head box A2 (FOXA2) and sex determining region Y (SRY) TFBS which are not found with the alternate C allele. Since the SRY TF located on the Y chromosome is a transcriptional regulator that controls a genetic switch in male development, this SNP might be expected to have an impact on male etiology as has been shown to be the case with its risk of hypospadias. The rs1877474 ATF3-T allele creates the punitive fork head box A1 (FOXA1) TFBS that does not occur with the alternate C allele. The TF which is involved with embryonic development and tissue differentiation might also have an impact on hypospadias. The rs11119982 ATF3-T allele creates the punitive AT rich interactive domain 3A (ARID3A), MYC associated factor X (MAX), v-myb myeloblastosis viral oncogene homolog (MYB), upstream transcription factor 1 (USF1) and zinc finger E-box binding homeobox 1 (ZEB1) TFBS which do not occur with the alternate C allele. The TFs for these binding sites are involved the transcriptional machinery including transcriptional regulation and suppression [20] which may in part be involved with sexual differentiation and the hypospadias trait.

v-Akt Murine Thymoma Viral Oncogene Homolog 3 (AKT3)

The phosphatidylinositol 3-kinase (PI3K)/AKT pathway plays a key role in numerous cellular functions including proliferation, adhesion, migration, invasion, metabolism and survival [34]. The v-akt murine thymoma viral oncogene homolog 3 (AKT3) is one of three isoforms of the AKTs which are major downstream targets of growth factor receptor tyrosine kinases that signal through PI3K [35]. There have been seven SNPs from AKT3 intron one that have been significantly associated with chronic mountain sickness (CMS) [36], aggressive prostate cancer

(PCA) [37] and renal cell carcinoma risk (RCC) [38]. The AKT3 SNP (rs4590656) (C/T) has been found to be significantly associated with Hb and Hct in Tibetan Chinese with CMS [36]. The AKT3-C allele creates two punitive TFBS not found with the alternate T-allele for the aryl hydrocarbon receptor nuclear translocator: aryl hydrocarbon receptor (ARNT: AHR) and hypoxia-inducible factor 1: aryl hydrocarbon receptor nuclear translocator (HIF1α: ARNT) TFs which are involved xenobiotic metabolism, cellular and systemic responses to hypoxia.

The AKT3 SNPs (rs10157763 (C/T), rs10927067 (G/A) and rs2125230 (G/A) are significantly associated with the risk of PCA [37]. The rs10157763 AKT3-C allele creates a TFBS not present with the alternate T-allele for the v-myc myelocytomatosis viral related oncogene (MYCN) TF which it is a member of the MYC family and is associated with a variety of tumors including neuroblastoma. The rs10157763 AKT3-T allele creates a TFBS not present with the alternate C-allele for the CCCTC-binding factor (CTCF) TF which is a transcriptional regulator protein with 11 highly conserved zinc finger (ZF) domains. This gene is a member of the BORIS + CTCF gene family and encodes a transcriptional regulator. This nuclear protein is able to use different combinations of the ZF domains to bind different DNA target sequences and proteins. The TF is implicated in diverse genomic regulatory functions, including transcriptional activation/repression, insulation, imprinting [39]. The rs10927067 AKT3-G allele creates a TFBS not found with the alternate A-allele for the v-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB) TF which is important in the regulation of lineage-specific hematopoiesis. The rs10927067 AKT3-A allele creates a TFBS not found with the alternate G-allele for the hepatocyte nuclear factor 1 homeobox B (HNF1B) TF which has been shown to function in nephron development. This TF is a member of the homeodomain-containing superfamily of transcription factors and regulates development of the embryonic pancreas. The rs2125230 AKT3-A allele creates a TFBS not found with the alternate G-allele for the homeobox A5 (HOXA5) TFs which upregulates the tumor suppressor p53 and plays an important role in tumorigenesis.      

The AKT3 SNPs (rs4132509 (C/A), rs12031994 (G/A) and rs2345994 (G/A)) are significantly associated with the risk of RCC [38]. The rs4132509 AKT3-C allele creates a TFBS not available with the alternate A-allele for the nuclear factor erythroid 2-related factor 1: MAF BZIP Transcription Factor G (NFE2L1: MAFG) TF, where NFE2L1 coordinates the up-regulation of cytoprotective genes via the antioxidant response element while MAFG is involved in cell differentiation of erythrocytes. The rs12031994 AKT3-G allele creates a TFBS not available with the alternate A-allele for the TF Forkhead box D1 (FOXD1) which plays a role in tumor formation and should have an effect on RCC. The rs12031994 AKT3-A allele creates a TFBS not available with the alternate G-allele for the T-cell acute lymphocytic leukemia 1: GATA binding protein 1 (TAL1: GALA1) TF which is involved with the genesis of hemopoietic malignancies and plays a role in hemopoietic differentiation and a positive regulator of erythroid differentiation. The rs2345994 AKT3-G allele creates two TFBS not found with the alternate A-allele for the SRY and SRY (sex determining region Y)-box 10 (SOX10) TFs which are part of a family of TFs involved in the regulation of embryonic development and in the determination of the cell fate. The rs2345994 AKT3-A allele creates the punitive Myocyte enhancer factor 2 (MEF2A) TFBS not found with the alternate G-allele who’s TF it activates many muscle-specific, growth factor-induced and stress-induced genes.

Adrenergic Beta Receptor Kinase 1 (G Protein-Coupled Receptor Kinase 2) (ADRBK1)

 The ADRBK1 gene, which transcribes the G protein-coupled receptor kinase 2 (GRK2), is an important regulator of adrenergic signaling and plays a central role in heart failure pathology [40-42]. The GRK2 gene is an important regulator of beta-adrenergic signaling and plays a central role in heart failure (HF) pathology [43-45]. Consequently, the inhibition of GRK2 has become an emerging treatment option of HF since the inhibition appears to be a powerful therapeutic approach and appears to provide complementation to β-blockade [44]. ADRBK1 SNPs resulting in TFBS changes have been examined with respect to cardiovascular disease in the black population [21]. The rs1894111 ADRBK1 SNP from intron one has been found to be significantly associated with blood pressure (BP) levels in a white population [43]. The rs1894111 ADRBK1-C allele in blacks creates a TFBS not found with the alternate T-allele for the aryl hydrocarbon receptor nuclear translocator: aryl hydrocarbon receptor (ARNT:AHR) TF which participates in xenobiotic metabolism. The rs1894111 ADRBK1-T allele creates three TFBS not found with the alternate C-allele for the E74-like factor 5 (ELF5), v-ets avian erythroblastosis virus E26 oncogene homolog (ERG) and zinc finger protein 354C (ZNF354C) TFs where ELF5 and ERG are members of the erythroblast transformation-specific (ETS) family of transcription factors while Zinc finger protein 354C (ZNF354C) functions as a transcription repressor. The rs7128315 SNP ADRBK1-G allele creates TFBS not found with the alternate A-allele for the sterol regulatory element binding transcription factors 1 & 2 (SREBF1 & 2) TFs which are involved with the cholesterol synthesis pathway and are required for lipid homeostasis and regulate transcription of the LDL receptor gene. The rs948988 SNP ADRBK1-A allele creates a TFBS not found with the alternate G-allele for the estrogen receptor 2 (ESR2) TF which is required for normal function of the cardiovascular system.

Type 2 Deiodeinase gene (DIO2)

The type 2 deiodeinase gene (DIO2) encodes a deiodinase that coverts the thyroid prohormone, thyroxine (T4), to the biologically active triiodothyronine (T3) hormone where T3 is involved in the vital role of regulating energy balance and glucose metabolism [46-49]. DIO2 is found in the thyroid gland, cardiac and skeletal muscle, brown adipose tissue, placenta, pituitary, central nervous system (CNS) and at low levels in kidney and pancreases [50-52]. Several SNPs have been found in the gene which have been studied is association with mental retardation (MR) [53], osteoarthritis [54], early-onset type 2 diabetes mellitus (T2DM) [55] and insulin resistance (IR) [56,57]. DIO2 SNPs resulted in TFBS changes have been examined with respect to these diseases and human conditions [22]. One of the SNPs, rs225017 (T/A) DIO2-T allele creates a punitive TFBS not found with the alternate A-allele for the pancreatic and duodenal homeobox 1 (PDX1) TF which is involved with insulin activation. This SNP has been found to be significantly associated with insulin resistance [56,57] suggesting that the elimination of the PDX1 binding site by the A-allele may result in this human condition. Another DIO2 SNP rs6574549 (T/G) is associated with fasting insulin, insulin action and energy expenditure in Pima Indians [55]. The rs6574549 SNP DIO2-T allele creates a punitive TFBS not found with the alternate G-allele for the hepatocyte nuclear factor 1 homeobox B (HNF1B) TF which is involved with embryonic pancreas development suggesting that the elimination of the HNF1B binding site by the G-allele may be in part responsible for early-onset T2DM [55].

A third DIO2 SNP rs225014 (T/C) has been shown to have a significant association with symptomatic osteoarthritis in Dutch women [54]. The rs225014 SNP DIO2-C allele creates a TFBS not found with the alternate T-allele for the NK3 homeobox 2 (NKX3-2) TF which are involved with the negative regulation of the chondrocyte maturation suggesting that the elimination of the NKX3-2 binding site by the T-allele may in part be responsible for the symptomatic osteoarthritis in Dutch women [54].

Endothetal Per-Arnt-Sim (PAS) domain protein 1 (EPAS1)

The endothetal Per-Arnt-Sim (PAS) domain protein 1 (EPAS1) gene which encodes hypoxia-inducible-factor-2 alpha (HIF2α) is a transcription factor that is involved in the response to hypoxia. Hypoxia is a major geographical condition associated with high-altitude environments [58]. EPAS1 is expressed in organs that are involved in oxygen transport and metabolism, such the lung, placenta and vascular endothelium [59], and is associated with many biological processes and diseases related to metabolism [60], angiogenesis [61,62], inflammation [63,64] and cancer [65-67]. EPAS1 SNPs (rs56721780, rs6756667, rs7589621 and rs1868092) resulted in TFBS changes have been examined with respect to these diseases and human conditions [23]. The rs56721780 (G/C) SNP HIF2γ -G allele creates punitive unique TFBS, which not found with the alternate C-allele, for the V-Rel Avian Reticuloendotheliosis Viral Oncogene Homolog (REL), V-Rel Avian Reticuloendotheliosis Viral Oncogene Homolog A (RELA), Runt-Related Transcription Factor 1 (RUNX1), Activating Enhancer Binding Protein 2α (TFAP2A), Activating Enhancer Binding Protein 2β (TFAP2B, Activating Enhancer Binding Protein 2γ (TFAP2C) TFs. These TFs are involved with inflammation, immunity, differentiation, cell growth, tumorigenesis, apoptosis, hematopoiesis, transcriptional activation and repression, respectively [23] and the elimination of these TFBS by the alternate C-allele should have a tremendous impact on gene regulation. As an example, RUNX1 is a heterodimeric transcription factor that binds to the core element of many enhancers and promoters and can either activate or suppress transcription. The rs6756667 (A/G) SNP HIF2α-A allele creates two unique punitive TFBS for the Nuclear Factor I/A (NFIA) and Nuclear Factor I/C (NFIC) TFs which are capable of activating transcription and replication [23]. The alternate HIF2γ -G allele creates a unique punitive TFBS not found with the A-allele for the MGA, MAX Dimerization Protein (MGA) TF which can act as a repressor or an activator. The rs7589621 (G/A) SNP HIF2γ -G allele creates two unique punitive TFBS not found with the alternate A-allele for the Nuclear Receptor Subfamily 3, Group C, Member 1 & 2 (Glucocorticoid Receptor) (NR3C 1 & 2) TFs. These TFs modulate immune responses through suppression of chemokine and cytokine production and protecting cells from oxidative stress and damage as well as mediates aldosterone actions on salt and water balance within target cells [23]. The rs1868092 (A/G) SNP HIF2α -A allele creates three unique punitive TFBS not found with the alternate G-allele for the Nuclear Receptor Subfamily 2, Group C, Member 2 (NR2C2), YY1 & 2 TFs which are involved with the repression or activation of transcription, and positive and negative control of transcription at the transcription start site, respectively [23].

Lysosomal Acid Lipase A (LIPA)

The lysosomal acid lipase A (LIPA) gene encodes lysosomal acid lipase (LAL) which hydrolyzes cholesteryl esters and triglycerides in the cell lysosome to generate free cholesterol and fatty acids [68]. Alterations in the LAL enzyme activity could produce an accumulation of triglycerides and cholesterol esters in the cell which would result in foam cell formation, complement activation, an inflammation process and atherosclerotic plaque formation [69]. Un-esterified cholesterol is a distinguishing characteristic of atherosclerotic lesions [70]. Cholesteryl ester hydrolysis has been shown to be a critical step in the enzymatic modification of low density lipoprotein (LDL) particles in the intima [71,72] where the particles create a risk for cardiovascular disease (CD). The LIPA SNPs (rs1412444 and rs2246833) which are in linkage disequilibrium (LD) have been found to be significantly associated with coronary artery disease (CAD) [73,74]. These SNPs were found to create alterations in punitive TFBS and these potential changes were examined with respect to disease [75]. The rs1412444 (C/T) SNP LIPA-C allele is located in a unique punitive TFBS not found with the alternate T-allele for the GA binding protein transcription factor (GABPA) TF. This TF is involved in the activation of cytochrome oxidase expression and nuclear control of mitochondrial function. Consequently, individuals carrying T allele maybe at risk for mitochondrial related diseases such skeletal muscles, kidney and the endocrine and respiratory systems. In fact, the T allele has been significantly associated with CAD [73,74,76,77] where the T allele frequency was found to change from 0.491 in a control group compared to 0.561 in the CAD group [74]. The rs2246833 SNP LIPA T allele has also been found to be significantly associated with CAD [73,74] which is not surprising since the two SNPs are strongly linked as indicated by the high LD value (r2 = 0.937).

Peroxisome Proliferator-activated Receptors (PPARα/δ/γ)

The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcriptional factors (TFs) that regulate many genes in cell differentiation and various metabolic processes including lipid and glucose homeostasis. They are nuclear hormone receptors belonging to a steroid receptor superfamily that include estrogen, thyroid hormone, vitamin D3 and glucocorticoid receptors [78-80]. There are three PPAR isotypes, PPARα, PPARβ/δ and PPARγ. PPARα is expressed in the liver, skeletal muscles, heart, intestinal mucosa, and brown adipose tissue while PPARβ/δ is expressed in liver, skeletal and cardiac muscle, adipose tissue and macrophages. PPARγ occurs as three isoforms PPARγ1, PPARγ3, which are expressed in the liver, intestine and spleen, and PPARγ2, which is expressed in brown and white adipose tissue [81]. There has been much published concerning the PPARs significant involvement in the progression of human disease [78,80,82-84]. There have been several PPARα/δ/γ single nucleotide polymorphisms (SNPs) significantly associated with various human diseases or conditions [85-93]. Some of the PPARα/δ/γ SNPs were found to create alterations in punitive TFBS and these potential changes were examined with respect to disease [94]. The rs1800206 PPAR SNP (C/G) has been associated with variation in lipid serum levels in Caucasian and Indian populations [95,96], hypertriglyceridemia risk, dyslipidemia risk and low-density lipoprotein-cholesterol risk in Han Chinese [86,87,92]. The minor G allele generates a unique punitive T-Box 4 (TBX4) TFBS not found with the common G-allele [94]. The TBX4 transcription factor is associated with the disease heritable pulmonary arterial hypertension [97], which may in part be responsible for the variation in lipid serum levels and disease risks found to be significantly associated with the this SNP. The occurrence of the G allele in the Han Chinese population is 0.02 [94] which is a frequency that might be expected among heritable diseases. This SNP has also been associated with changes in triglyceridemia, total cholesterol, low-density lipoprotein-cholesterol (LDL-c), high-density lipoprotein-cholesterol (HDL-c) and apolipoprotein A1 (APOA1) plasma concentrations in populations of African-Americans, Caucasians and Indians [95,96,98,99].

The PPARδ gene is expressed in high levels in liver, kidneys, cardiac and skeletal muscle, adipose tissue, brain, colon and vasculature [100]. One of its SNPs (rs2016520) has been associated with obesity risk and intracerebral hemorrhages while a second SNP (rs9794) has been found to be associated with hypertriglyceridemia and obesity in Han Chinese [85,90,93]. The rs2016520 SNP (A/G) common A-allele creates unique punitive TFBS for the neuronal differentiation 2 (NEUROD2) and oligodendrocyte transcription factor 3 (OLIG3) TFs which do not occur with the minor G-allele [94]. The NEUROD2 TF is a transcriptional regulator implicated in neuronal determination while the OLIG3 TF promotes formation and maturation of oligodendrocytes, especially within the brain. These TFs could in part be associated with obesity risk and intracerebral hemorrhages. The rs9794 SNP (C/G) common C allele creates unique punitive TFBS for glial cells missing homolog 1 and 2 (GCM 1 & 2) TFs which bind to the trophoblast specific element 2 (TSE2) of the aromatase gene enhancer and act as a master regulator of parathyroid development. The deletion of these TFBS caused by the minor G allele for these two TFs could in part be associated with hypertriglyceridemia and obesity risk in Han Chinese [86,93].

The PPARγ gene is expressed at high levels in adipose tissue, lipid storage, glucose metabolism as well as the transcriptional regulation of genes involved in these metabolic processes [101]. One of its SNPs (rs10865710) has been associated with obesity risk, systemic sclerosis and low-density lipoprotein-cholesterol in Han Chinese [85,89,92]. The rs10865710 SNP (C/G) common C-allele creates a unique punitive TFBS for the POU class 2 homeobox 2 (POU2F2) which does not occur with the minor G-allele [94]. This TF is found in immunoglobulin gene promoters and the absence of this BS with the minor G-allele in part may be responsible for the association of this SNP with systemic sclerosis. In addition, the creation of the motor neuron and pancreas homeobox 1 (MNX1) and regulatory factor X3 (RXF3) TFBS by the common C-allele and not the minor G-allele as well as the creation of the NK6 homeobox 1 (NKX6-1) TFBS with the minor G-allele and not the major C-allele may in part be responsible for association of this SNP with obesity risk and low-density lipoprotein-cholesterol since these TFs are involved with the pancreas development and function as well as insulin gene regulation, respectively [94].

The rs12629751 PPARγ SNP (C/T) has been associated with osteoarthritis in southeast Chinese [88]. The common C-allele creates a unique punitive TFBS not found with the minor T-allele for the NK6 Homeobox 1 (NKX3-2) TF which is a repressor that acts as a negative regulator of chondrocyte maturation which may in part be responsible for the association of this SNP with osteoarthritis [94].

The rs1805192 PPARγ SNP (C/G) has been associated with hypertriglyceridemia, dyslipidemia and low-density lipoprotein-cholesterol in Han Chinese [86,87,92]. The common C allele creates a unique punitive TFBS not found with the minor G-allele for the estrogen receptor α (ERS2) TF which is expressed in many tissues including pulmonary epithelial cells. The minor G-allele creates a unique punitive TFBS not found with the common C-allele for the myocyte enhancer factor 2C (MEF2C) TF which is a transcriptional activator that controls cardiac morphogenesis and myogenesis, and is also involved in vascular development [94]. The occurrence of these punitive unique TFBS for the opposite SNP alleles may in part be responsible for hypertriglyceridemia, dyslipidemia, low-density lipoprotein-cholesterol found in Han Chinese.

Signal Transducer and Activator of Transcription 4 (STAT4)

The Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) pathways play a critical role in immune, neuronal, hematopoietic and hepatic systems [102]. JAK-STAT is a principal signal transduction pathway in cytokine and growth factor signaling as well as regulating various cellular processes such as cell proliferation, differentiation migration and survival [103]. JAK-STAT provides the principle intracellular signaling mechanism required for a wide array of cytokines [104,105]. The STAT portion of the signaling cascade has seven mammalian family members which are STAT1, 2, 3, 4, 5a, 5b and 6 [104,105]. These STATs bind thousands of transcriptional factor binding sites (TFBS) in the genome and regulate the transcription of many protein-coding genes, miRNAs and long noncoding RNAs [105]. The STAT4 gene which is important for signaling by interleukins (IL-12 and IL-23) and type 1 interferons [105] has been found to have several simple nucleotide polymorphisms (SNPs) associated with human disease [106-113]. STAT4 transduces IL-12, IL-23 and type 1 interferon-mediated signals into helper T (Th) cells (Th1 and Th17) differentiation, monocyte activation, and interferon-gamma production [113,114]. The STAT4-dependent cytokine regulation is found in the pathogenesis of autoimmune disease [115,116] such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) [117,118].

Several SNPs in the gene have been significantly association with Behcet’s Disease [119], diabetes risk [107], hepatitis B virus-related hepatocellular carcinoma [107,111,120,121], inflammatory bowel disease [122], juvenile idiopathic arthritis [123], primary biliary cirrhosis and Crohn's disease [124], severe renal insufficiency in lupus nephritis [109], systemic lupus erythematosus [106] and ulcerative colitis [125]. Some STAT4 SNPs were found to create alterations in punitive TFBS and these punitive changes were examined with respect to these disease or conditions [25].

The rs7574865 STAT4 SNP (G/T) has been found to be significantly associated with diabetes [112], hepatitis B virus-related hepatocellular carcinoma [107,111,120,121], inflammatory bowel disease [122], juvenile idiopathic arthritis [123], primary biliary cirrhosis and Crohn's disease [124], severe renal insufficiency in lupus nephritis [109], systemic lupus erythematosus [106] and ulcerative colitis [125]. The minor T-allele for the SNP creates a unique punitive TFBS not found with the common G-allele for the nuclear receptor subfamily 1, group H, member 3: retinoid X receptor, alpha (NR1HE:RXRα TF which is a key regulator of macrophage function, controlling transcriptional programs involved in lipid homeostasis and inflammation [25]. Since the NR1HE: RXRα protein duplex is part of the NR1 subfamily of the retinoid nuclear receptor superfamily, the presence of its TFBS created by the T allele could in part be responsible for many of the diseases or conditions mentioned above that are significantly associated with this SNP.

The rs11889341 STAT4 SNP has been found to be significantly associated with diabetes [112], hepatitis B virus (HBV) infection, HBV-related cirrgisus and hepatocellular carcinoma [124], severe renal insufficiency in lupus nephritis [109], and systemic lupus erythematosus [106]. The common C-allele of the rs11889341 STAT4 SNP (C/T) creates a unique punitive TFBS not found with the minor T-allele for the androgen receptor (AR) which is a steroid-hormone activated transcription factor that stimulates transcription of androgen responsive genes expressed in bone marrow, mammary gland, prostate, testicular and muscle tissues [25]. The absence of this TFBS with the minor T-allele should have a major effect relating to the diseases or conditions mentioned above. The T-allele of the SNP creates two unique punitive TFBS not found with the C-allele for the myocyte enhancer factor 2C (MEF2C) TFs which controls cardiac morphogenesis and myogenesis, and is also involved in vascular development and consequently the presence or absence of this TFBS should have an impact on the disease.

The rs8179673 STAT4 SNP has been found to be significantly associated with diabetes [112], hepatitis B virus (HBV) infection, HBV-related cirrgisus and hepatocellular carcinoma [124] and systemic lupus erythematosus [106]. The minor C-allele of the rs8179673 STAT4 SNP (T/C) creates two unique punitive TFBS not found with the common T-allele for the hepatocyte nuclear factor 1 homeobox A (HNF1a) and the hepatocyte nuclear factor 4, gamma (HNF4g) TFs which regulate the tissue specific expression of multiple genes, especially in pancreatic islet cells and in liver [25]. The occurrence of these TFBS with the minor allele and not the major allele could very well explain why the SNP is significantly associated with the above diseases.

The rs7574070 and rs7572482 STAT4 SNPs have been found to be significantly associated with Behcet’s disease [119]. The common A-allele of the rs7574070 STAT4 SNP (A/C) creates three unique punitive TFBS not found with the minor C-allele for the CCAAT/enhancer binding protein (C/EBP), beta (CEBPβ, Regulatory Factors 1 & 5 (RFX1 & 5) TFs [25]. The CEBPα-TF is an important transcriptional activator regulating the expression of genes involved in immune and inflammatory responses while the RFX1 & 5 TFs are important regulatory factors essential for MHC class II gene expression and the loss of these BS with the presence of the minor C allele could have an impact on Behcet’s disease. The minor C-allele of the SNP STAT4-C allele creates a unique punitive TFBS not found with the A-allele for the STAT4 TF which is an important in regulating genes associated with systemic lupus erythematosus and rheumatoid arthritis and could also have an impact on Behcet’s disease.

The common A-allele of the rs7572482 STAT4 SNP (A/G) creates two unique punitive TFBS not found with the minor G-allele for the FBJ Murine Osteosarcoma Viral Oncogene Homolog (FOS) and POU5F1:SOX2 TFs. The FOS TF is a regulator of cell proliferation, differentiation and transformation while the POU Class 5 Homeobox 1: SRY (Sex Determining Region Y)-Box 2 (POU5F1:SOX2) TF play a key role in embryonic development and stem cell pluripotency which both could have an impact on the disease.

Thromboxane A2 Receptor (TBXA2R)

The thromboxane A2 receptor (TBXA2R) gene is a member of the seven-transmembrane G-protein-coupled receptor super family, which interacts with intracellular G proteins, regulates different downstream signaling cascades, and induces many cellular responses including the intracellular calcium influx, cell migration and proliferation, and apoptosis [126]. This gene is abundantly expressed in tissues at the mRNA and protein levels targeted by the TBXA2R ligand thromboxane A2 (TXA2) that include erythroleukaemia cells, vascular and bronchial smooth muscle, uterus and placental tissue, endothelium, epithelium, trophoblasts, thymus, liver and small intestine [127]. The activation of TBXA2R in bronchial smooth muscle cells by its ligand results in intercellular calcium mobilization with subsequent bronchoconstriction, which contributes to bronchial smooth muscle hyperplasia and airway remodeling, which occurs in response to chronic airway inflammation in asthma [128]. There are TBXA2R SNPs that have been associated with asthma in Asians. The rs4523 TBXA2R SNP has been found to be associated with adult asthma in a Japanese population [129] and childhood atopic asthma in a Chinese population [130], while the rs11318632 SNP was found to be associated with atopic asthma in a Korean population [131]. Two haplotypes (H2 & H4) involving the four linked TBXA2R SNPs from intron one where found to influence TBXA2R transcriptional activity and were also associated with asthma-related phenotypes [132]. This suggests that these SNPs may be part of a regulatory network for the TBXA2R gene in Asian populations. TBXA2R SNPs were found to create alterations in punitive TFBS and these punitive changes were examined with respect to asthma in Asians [133].

The minor G-allele of the rs2238631 TBXA2R SNP (A/G) creates two unique punitive TFBS not found with the common A-allele for the Fork Head box C1 (FOXC1) and AP (TFAP2α) TFs which are involved in cell viability and resistance to oxidative stress and activating transcription of some genes while inhibiting the transcription of other genes, respectively [133]. The minor T-allele of the rs2238632 TBXA2R SNP (C/T) creates five unique punitive TFBS not found with the common C-allele for the aryl hydrocarbon receptor nuclear translocator (ARNT), cAMP responsive element binding protein 1 (CREB1), hypoxia-inducible factor 1:Aryl hydrocarbon receptor nuclear translocator (HIF1α:ARNT), MYC associated factor X (MAX) and upstream transcription factor 1 (USF1) TFs which are involved with xenobiotic metabolism, circadian rhythmicity, cellular and systemic responses to hypoxia, transcriptional regulator and a cellular TF, respectively [133]. The minor C-allele of the rs1131882 TBXA2R SNP (T/C) creates a unique punitive TFBS not found with the common T-allele for the insulinoma-associated 1 (INSM1) TF which is a sensitive marker for neuroendocrine differentiation of human lung tumors [133]. The major T-allele of the rs4523 TBXA2R SNP (T/C) creates a unique punitive TFBS not found with the C-allele for the Krueppel-like factor 4 (KLF4) TF which acts as an activator, a repressor and regulates the expression of key TFs during embryonic development. The minor C-allele of the SNP creates a unique punitive TFBS not found with the major T-allele for the nuclear receptor subfamily 3, group C, member 1 (NR3C1) TF which is involved in inflammatory responses, cellular proliferation, and differentiation in target tissues [133].

Vascular Endothelial Growth Factor (VEGF)-A

The vascular endothelial growth factor (VEGF) is a family of key regulators in critical physiological and pathological angiogenesis [134] including tissue growth, wound healing, rheumatoid arthritis, proliferative retinopathies, cardiovascular disease, and cancer [135], and is a growth factor activator for angiogenesis, vasculogenesis, and endothelial cell growth. In most [136-141] but not all [142] studies, It has been shown that the VEGF is an important component of the pathogenesis of high altitude adaptation and sickness. Presently, seven VEGF family members and 14 alternative splicing variants have been identified in humans [143-145]. Of the 14 splicing variants, 12 are VEGFA isoforms [145] with three (VEGFA-121, -165 and -189) being differentially expressed in humans visiting or living in high altitude environments and also in chronic mountain sickness (CMS) patients [137,138]. Among all family members, VEGFA is the most potent and best-known angiogenic protein and exerts its’ biologic effect through interaction with cell-surface receptors, which triggers a cascade of downstream dimerizations and phosphorylations [146]. VEGFA is a signaling protein involved in the regulation of angiogenesis, vasculogenesis and endothelial cell growth. It induces endothelial cell proliferation, promotes cell migration, inhibits apoptosis and induces permeabilization of blood vessels. VEGFA is also a key regulator of hypoxia. There are several VEGFA SNPs associated with high altitude sickness that create alterations in punitive TFBS for the gene [26-28,147-149].

The major C-allele of the rs699947 VEGFA SNP (C/A) creates a unique punitive TFBS not found with the minor A-allele for the hypoxia-inducible factor 1α: aryl hydrocarbon receptor nuclear translocator (HIF1β:ARNT) which plays an essential role in cellular and systemic responses to hypoxia and is detrimental to the individual’s ability to respond to hypoxia. The minor T-allele of the rs79469752 VEGFA SNP (C/T) creates two unique punitive TFBS not found with the common C-allele for the Nuclear Receptor subfamily 2, group C, member 2 (NR2C2) and Runt-related transcription factor 1 (RUNX1) TFs which plays a role in protecting cells from oxidative stress and damage induced by ionizing radiation as well as the involvement in the development of normal hematopoiesis, respectively. The major C-allele of the rs2010963 VEGFA SNP (C/G) creates a unique punitive TFBS not found with the minor G-allele for the GA-binding protein γ chain (GABPαTF which is likely involved in the activation of cytochrome oxidase expression and nuclear control of mitochondrial function. The major C-allele of the rs3025039 VEGFA SNP (C/T) creates two unique punitive TFBS not found with the minor T-allele for the estrogen receptor α (ESR2) and the HIF1β:ARNT TFs where ESR2 is expressed by many tissues including blood monocytes and tissue macrophages, colonic and pulmonary epithelia cells while the function of the HIF1a:ARNT TF is mentioned above.

The rs3025039 VEGFA SNP also affects the has-miR-591 predicted binding target CA[C/T]GGTC in the 3’UTR, where a VEGFA-C allele would eliminate the binding site for the miRNA. These VEGFA SNPs cause alterations in punitive TFBS for the gene and should have an effect on high altitude sickness among humans.

Conclusion

Non-coding SNPs identified by GWAS that have shown to be been significantly associated with human disease or sickness are considered risk-associated SNPs [4]. Those near a gene causing changes in gene expression levels are considered regulatory (r) SNPs [150]. Such SNPs located in TFBS can alter the binding ability of the respective TF. There have been many reports on the possible outcome of such alterations by identifying punitive TFBS based on the two alleles of the SNP associated with a disease or sickness [19-30]. In this review a number of examples from previously reports were discussed with an emphasis on the more important dynamics from the reports.

References

  1. Oetting WS, Béroud C, Brenner SE, Greenblatt M, Karchin R, et al. (2017) Non-Coding Variation: The 2016 Annual Scientific Meeting of the Human Genome Variation Society. Hum Mutat 38: 460-463. [Crossref]
  2. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, et al. (2009) Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 106: 9362-9367. [Crossref]
  3. Kumar V, Westra HJ, Karjalainen J, Zhernakova DV, Esko T, et al. (2013) Human disease-associated genetic variation impacts large intergenic non-coding RNA expression. PLoS Genet 9: e1003201. [Crossref]
  4. Tak YG, Farnham PJ (2015) Making sense of GWAS: using epigenomics and genome engineering to understand the functional relevance of SNPs in non-coding regions of the human genome. Epigenetics Chromatin 8: 57. [Crossref]
  5. Knight JC (2003) Functional implications of genetic variation in non-coding DNA for disease susceptibility and gene regulation. Clin Sci (Lond) 104: 493-501. [Crossref]
  6. Wang X, Tomso DJ, Liu X, Bell DA (2005) Single nucleotide polymorphism in transcriptional regulatory regions and expression of environmentally responsive genes. Toxicol Appl Pharmacol 207(2 Suppl): 84-90. [Crossref]
  7. Chorley BN, Wang X, Campbell MR, Pittman GS, Noureddine MA, et al. (2008) Discovery and verification of functional single nucleotide polymorphisms in regulatory genomic regions: current and developing technologies. Mutat Res 659: 147-157. [Crossref]
  8. Conde L, Vaquerizas JM, Santoyo J, Al-Shahrour F, Ruiz-Llorente S, et al. (2004) PupaSNP Finder: a web tool for finding SNPs with putative effect at transcriptional level. Nucleic Acids Res 32: W242-248. [Crossref]
  9. Chowdhary R, Tan SL, Pavesi G, Jin J, Dong D, et al. (2012) A database of annotated promoters of genes associated with common respiratory and related diseases. Am J Respir Cell Mol Biol 47: 112-119.
  10. Claussnitzer M, Dankel SN, Klocke B, Grallert H, Glunk V, et al. (2014) Leveraging cross-species transcription factor binding site patterns: from diabetes risk loci to disease mechanisms. Cell 156: 343-358. [Crossref]
  11. Jiang J, Jia P, Shen B, Zhao Z (2014) Top associated SNPs in prostate cancer are significantly enriched in cis-expression quantitative trait loci and at transcription factor binding sites. Oncotarget 5: 6168-6177.
  12. Castaldi PJ, Cho MH, Zhou X, Qiu W, Mcgeachie M, et al. (2015) Genetic control of gene expression at novel and established chronic obstructive pulmonary disease loci. Hum Mol Genet 24: 1200-1210. [Crossref]
  13. Benson CC, Zhou Q, Long X, Miano JM (2011) Identifying functional single nucleotide polymorphisms in the human CArGome. Physiol Genomics 43: 1038-1048. [Crossref]
  14. Ward LD, Kellis M (2012) HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res 40: D930-934. [Crossref]
  15. Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, et al. (2012) Annotation of functional variation in personal genomes using RegulomeDB. Genome Res 22: 1790-1797. [Crossref]
  16. Bryne JC, Valen E, Tang MH, Marstrand T, Winther O, et al. (2008) JASPAR, the open access database of transcription factor-binding profiles: new content and tools in the 2008 update. Nucleic Acids Res 36: D102-106. [Crossref]
  17. Sandelin A, Alkema W, Engström P, Wasserman WW, Lenhard B (2004) JASPAR: an open-access database for eukaryotic transcription factor binding profiles. Nucleic Acids Res 32: D91-94. [Crossref]
  18. Sandelin A1, Wasserman WW, Lenhard B (2004) ConSite: web-based prediction of regulatory elements using cross-species comparison. Nucleic Acids Res 32: W249-252. [Crossref]
  19. Buroker NE (2013) AKT3 rSNPs, Transcritional Factor Binding Sites and Human Disease. Open Journal of Blood Diseases 3: 116-129.
  20. Buroker NE (2013) ATF3 rSNPs, transcriptional factor binding sites and human etiology. Open Journal of Genetics 3: 253-261.
  21. Buroker NE (2014) ADRBK1 (GRK2) rSNPs, Transcriptional Factor Binding Sites and Cardiovascular Disease in the Black Population. Journal of Cardiovascular Disease. [Crossref]
  22. Buroker NE (2014) DIO2 rSNPs, transcription factor binding sites and disease. British Journal of Medicine and Medical Research 9: 1-24.
  23. Buroker NE (2016) Computational EPAS1 rSNP analysis, transcriptional factor binding sites and high altitude sickness or adaptation. Journal of Proteomics and Genomics research 1: 31-59.
  24. Buroker NE (2015) LIPA rSNPs (rs1412444 and rs2246833), Transcriptional Factor Binding Sites and Disease. British Biomedical Bulletin 3: 281-294.
  25. Buroker NE (2016) Computational STAT4 rSNP analysis, transcriptional factor binding sites and disease. Bioinformatics and Diabetes 1: 1-36.
  26. Buroker NE (2014) VEGFA rSNPs, transcriptional factor binding sites and human disease. J Physiol Sci 64: 73-76. [Crossref]
  27. Buroker NE (2015) VEGFA SNPs (rs34357231 & rs35569394), Transcriptional Factor Binding Sites and Human Disease. British Journal of Medicine and Medical Research 10: 1-11.
  28. Buroker NE, Ning X-H, Li K, Zhou Z-N, Cen W-J, et al. (2015) SNPs, Linkage Disequilibrium and Transcriptional Factor Binding Sites Associated with Acute Mountain Sickness among Han Chinese at the Qinghai-Tibetan Plateau. International Journal of Genomic Medicine.
  29. Buroker NE, Ning XH, Zhou ZN, Li K, Cen WJ, et al. (2013) VEGFA SNPs and transcriptional factor binding sites associated with high altitude sickness in Han and Tibetan Chinese at the Qinghai-Tibetan Plateau. J Physiol Sci 63: 183-193. [Crossref]
  30. Buroker NE (2015) SNP (rs1570360) in Transcriptional Factor Binding Sites of the VEGFA Promoter is Associated with Hypertensive Nephropathy and Diabetic Retinopathy. Austin Journal of Endocrinology and Diabetes 2: 5.
  31. Buroker NE (2017) Identifying Alterations in Transcriptional Factor Binding Sites. Fetal, Neonatal and Development Medicine 1: 1-4.
  32. Liu B, Agras K, Willingham E, Vilela ML, Baskin LS (2006) Activating transcription factor 3 is estrogen-responsive in utero and upregulated during sexual differentiation. Horm Res 65: 217-222. [Crossref]
  33. Beleza-Meireles A, Töhönen V, Söderhäll C, Schwentner C, Radmayr C, et al. (2008) Activating transcription factor 3: a hormone responsive gene in the etiology of hypospadias. Eur J Endocrinol 158: 729-739. [Crossref]
  34. Bader AG, Kang S, Zhao L, Vogt PK (2005) Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer 5: 921-929. [Crossref]
  35. Testa JR, Bellacosa A (2001) AKT plays a central role in tumorigenesis. Proc Natl Acad Sci U S A 98: 10983-10985. [Crossref]
  36. Buroker NE, Ning XH, Zhou ZN, Li K, Cen WJ, et al. (2012) AKT3, ANGPTL4, eNOS3, and VEGFA associations with high altitude sickness in Han and Tibetan Chinese at the Qinghai-Tibetan Plateau. Int J Hematol 96: 200-213. [Crossref]
  37. Lavender NA, Rogers EN, Yeyeodu S, Rudd J, Hu T, et al. Interaction among apoptosis-associated sequence variants and joint effects on aggressive prostate cancer. BMC Med Genomics 5: 11. [Crossref]
  38. Shu X, Lin J, Wood CG, Tannir NM, Wu X (2013) Energy balance, polymorphisms in the mTOR pathway, and renal cell carcinoma risk. J Natl Cancer Inst 105: 424-432. [Crossref]
  39. Phillips JE, Corces VG (2009) CTCF: master weaver of the genome. Cell 137: 1194-1211. [Crossref]
  40. Pitcher JA, Freedman NJ, Lefkowitz RJ (1998) G protein-coupled receptor kinases. Annu Rev Biochem 67: 653-692. [Crossref]
  41. Ribas C, Penela P, Murga C, Salcedo A, García-Hoz C, et al. (2007) The G protein-coupled receptor kinase (GRK) interactome: role of GRKs in GPCR regulation and signaling. Biochim Biophys Acta 1768: 913-922. [Crossref]
  42. So CH1, Michal A, Komolov KE, Luo J, Benovic JL (2013) G protein-coupled receptor kinase 2 (GRK2) is localized to centrosomes and mediates epidermal growth factor-promoted centrosomal separation. Mol Biol Cell 24: 2795-2806. [Crossref]
  43. Lobmeyer MT, Wang L, Zineh I, Turner ST, Gums JG, et al., Polymorphisms in genes coding for GRK2 and GRK5 and response differences in antihypertensive-treated patients. Pharmacogenet Genomics 21: 42-49. [Crossref]
  44. Cannavo A, Liccardo D, Koch WJ (2013) Targeting cardiac beta-adrenergic signaling via GRK2 inhibition for heart failure therapy. Front Physiol 4: 264. [Crossref]
  45. Lymperopoulos A (2013) Physiology and pharmacology of the cardiovascular adrenergic system. Front Physiol 4: 240. [Crossref]
  46. Danforth E Jr (1983) The role of thyroid hormones and insulin in the regulation of energy metabolism. Am J Clin Nutr 38: 1006-1017. [Crossref]
  47. Krotkiewski M (2002) Thyroid hormones in the pathogenesis and treatment of obesity. Eur J Pharmacol 440: 85-98. [Crossref]
  48. Silva JE (1995) Thyroid hormone control of thermogenesis and energy balance. Thyroid 5: 481-492. [Crossref]
  49. Freake HC, Oppenheimer JH (1995) Thermogenesis and thyroid function. Annu Rev Nutr 15: 263-291. [Crossref]
  50. Bianco AC, Larsen PR (2005) Cellular and structural biology of the deiodinases. Thyroid 15: 777-786. [Crossref]
  51. Bianco, A.C., et al., Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodeiodinases. Endocr Rev 2002. 23: 38-89. [Crossref]
  52. Bianco AC, Salvatore D, Gereben B, Berry MJ, Larsen PR (2000) Characterization of the 5'-flanking and 5'-untranslated regions of the cyclic adenosine 3',5'-monophosphate-responsive human type 2 iodothyronine deiodinase gene. Endocr Rev 141: 229-237. [Crossref]
  53. Guo TW, Zhang FC, Yang MS, Gao XC, Bian L, et al. (2004) Positive association of the DIO2 (deiodinase type 2) gene with mental retardation in the iodine-deficient areas of China. J Med Genet 41: 585-590. [Crossref]
  54. Meulenbelt I, Min JL, Bos S, Riyazi N, Houwing-Duistermaat JJ, et al. (2008) Identification of DIO2 as a new susceptibility locus for symptomatic osteoarthritis. Hum Mol Genet 17: 1867-1875. [Crossref]
  55. Nair S, Muller YL, Ortega E, Kobes S, Bogardus C, et al. (2012) Association analyses of variants in the DIO2 gene with early-onset type 2 diabetes mellitus in Pima Indians. Thyroid 22: 80-87. [Crossref]
  56. Mentuccia D, Proietti-Pannunzi L, Tanner K, Bacci V, Pollin TI, et al. (2002) Association between a novel variant of the human type 2 deiodinase gene Thr92Ala and insulin resistance: evidence of interaction with the Trp64Arg variant of the beta-3-adrenergic receptor. Diabetes 51: 880-883. [Crossref]
  57. Leiria LB, Dora JM, Wajner SM, Estivalet AA, Crispim D, et al. (2014) The rs225017 polymorphism in the 3'UTR of the human DIO2 gene is associated with increased insulin resistance. PLoS One 9: e103960. [Crossref]
  58. Ward MP (2003) Everest 1953, first ascent: a clinical record. High Alt Med Biol 4: 27-37. [Crossref]
  59. Semenza GL (2007) Life with oxygen. Science 318: 62-64. [Crossref]
  60. Majmundar AJ, Wong WJ, Simon MC (2010) Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell 40: 294-309. [Crossref]
  61. Skuli N, Simon MC (2009) HIF-1alpha versus HIF-2alpha in endothelial cells and vascular functions: is there a master in angiogenesis regulation? Cell Cycle 8: 3252-3253.
  62. Skuli N, Liu L, Runge A, Wang T, Yuan L, et al. (2009) Endothelial deletion of hypoxia-inducible factor-2alpha (HIF-2alpha) alters vascular function and tumor angiogenesis. Blood 114: 469-477. [Crossref]
  63. Semenza GL (2011) Oxygen sensing, homeostasis, and disease. N Engl J Med 365: 537-547. [Crossref]
  64. Eltzschig HK, Carmeliet P (2011) Hypoxia and inflammation. N Engl J Med 364: 656-665. [Crossref]
  65. Kaelin WG Jr (2008) The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer. Nat Rev Cancer 8: 865-873. [Crossref]
  66. Han SS, Yeager M, Moore LE, Wei MH, Pfeiffer R, et al. (2012) The chromosome 2p21 region harbors a complex genetic architecture for association with risk for renal cell carcinoma. Hum Mol Genet 21: 1190-1200. [Crossref]
  67. Xue X, Taylor M, Anderson E, Hao C, Qu A, et al., Hypoxia-inducible factor-2alpha activation promotes colorectal cancer progression by dysregulating iron homeostasis. Cancer Res 72: 2285-2293. [Crossref]
  68. Brown MS, Goldstein JL (1986) A receptor-mediated pathway for cholesterol homeostasis. Science 232: 34-47. [Crossref]
  69. Zschenker O, Illies T, Ameis D (2006) Overexpression of lysosomal acid lipase and other proteins in atherosclerosis. J Biochem 140: 23-38. [Crossref]
  70. Kruth HS (1984) Localization of unesterified cholesterol in human atherosclerotic lesions. Demonstration of filipin-positive, oil-red-O-negative particles. Am J Pathol 114: 201-208. [Crossref]
  71. Bhakdi S, Dorweiler B, Kirchmann R, Torzewski J, Weise E, et al. (1995) On the pathogenesis of atherosclerosis: enzymatic transformation of human low density lipoprotein to an atherogenic moiety. J Exp Med 182: 1959-1971. [Crossref]
  72. Bhakdi S, Lackner KJ, Han SR, Torzewski M, Husmann M (2004) Beyond cholesterol: the enigma of atherosclerosis revisited. Thromb Haemost 91: 639-645. [Crossref]
  73. Wild PS, Zeller T, Schillert A, Szymczak S, Sinning CR, et al. (2011) A genome-wide association study identifies LIPA as a susceptibility gene for coronary artery disease. Circ Cardiovasc Genet 4: 403-412. [Crossref]
  74. 2021 Copyright OAT. All rights reserv
  75. Vargas-Alarcón G, Posadas-Romero C, Villarreal-Molina T, Alvarez-León E, Angeles J, et al., Single nucleotide polymorphisms within LIPA (Lysosomal Acid Lipase A) gene are associated with susceptibility to premature coronary artery disease. a replication in the genetic of atherosclerotic disease (GEA) Mexican study. PLoS One 8: e74703. [Crossref]
  76. Buroker NE (2015) LIPA rSNPs (rs1412444 and rs2246833), Transcriptional Factor Binding Sites and Disease. British Biomedical Bulletin 3: 281-294.
  77. Schunkert H, König IR, Kathiresan S, Reilly MP, Assimes TL, et al. (2011) Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet 43: 333-338. [Crossref]
  78. Wang Y, Wang L, Liu X, Zhang Y, Yu L, et al. (2014) Genetic variants associated with myocardial infarction and the risk factors in Chinese population. PLoS One 9: e86332. [Crossref]
  79. Berger J, Moller DE (2002) The mechanisms of action of PPARs. Annu Rev Med 53: 409-435. [Crossref]
  80. Boitier E, Gautier JC, Roberts R (2003) Advances in understanding the regulation of apoptosis and mitosis by peroxisome-proliferator activated receptors in pre-clinical models: relevance for human health and disease. Comp Hepatol 2: 3. [Crossref]
  81. Harmon GS, Lam MT, Glass CK (2011) PPARs and lipid ligands in inflammation and metabolism. Chem Rev 111: 6321-6340. [Crossref]
  82. Reddy JK, Rao MS (2006) Lipid metabolism and liver inflammation. II. Fatty liver disease and fatty acid oxidation. Am J Physiol Gastrointest Liver Physiol 290: G852-858. [Crossref]
  83. Fruchart JC (2009) Peroxisome proliferator-activated receptor-alpha (PPARalpha): at the crossroads of obesity, diabetes and cardiovascular disease. Atherosclerosis 205: 1-8.
  84. Kersten S, Wahli W (2000) Peroxisome proliferator activated receptor agonists. EXS 89: 141-151. [Crossref]
  85. Desvergne B, Wahli W (1999) Peroxisome Proliferator-Activated Receptors: Nuclear control of metabolism. Endocr Rev 20: 649-688.
  86. Luo W, Guo Z, Wu M, Chao H, Hu X, et al. (2013) Association of peroxisome proliferator-activated receptor alpha/delta/gamma with obesity, and gene-gene interaction, in the Chinese Han population. J Epidemiol 23: 187-194.
  87. Gu SJ, Liu MM, Guo ZR, Wu M, Chen Q, et al. (2013) Gene-gene interactions among PPAR polymorphisms for hypertriglyceridemia in Chinese Han population. Gene 515: 272-276. [Crossref]
  88. Gu SJ, Guo ZR, Zhou ZY, Hu XS, Wu M (2014) PPAR alpha and PPAR gamma polymorphisms as risk factors for dyslipidemia in a Chinese Han population. Lipids Health Dis 13: 23.
  89. Zheru D, Peiliang F, Yuli W, Haishan W, Qirong Q, et al. (2014) Association of PPARγ gene polymorphisms with osteoarthritis in a southeast Chinese population. J Genet 93: 719-723. [Crossref]
  90. Marangoni RG, Korman BD, Allanore Y, Dieude P, Armstrong LL, et al. (2015) A candidate gene study reveals association between a variant of the Peroxisome Proliferator-Activated Receptor Gamma (PPAR-gamma) gene and systemic sclerosis. Arthritis Res Ther 17: 128.
  91. Huang Y, Nie S, Zhou S, Li K, Sun J, et al. (2015) PPARD rs2016520 polymorphism and circulating lipid levels connect with brain diseases in Han Chinese and suggest sex-dependent effects. Biomed Pharmacother 70: 7-11. [Crossref]
  92. Lapice E, Monticelli A, Cocozza S, Pinelli M, et al. (2015) The PPARγ2 Pro12Ala variant is protective against progression of nephropathy in people with type 2 diabetes. J Transl Med 13: 85. [Crossref]
  93. Fan W, Shen C, Wu M, Zhou ZY, Guo ZR (2015) Association and interaction of PPARalpha, delta, and gamma gene polymorphisms with low-density lipoprotein-cholesterol in a Chinese Han population. Genet Test Mol Biomarkers 19: 379-386. [Crossref]
  94. Luo W, Chen F, Guo Z, Wu M, Zhou Z, et al. (2016) A population association study of PPAR delta gene rs2016520 and rs9794 polymorphisms and haplotypes with body mass index and waist circumference in a Chinese population. Ann Hum Biol 43: 67-72.
  95. Buroker NE (2017) Identifying Changes in Punitive Transcriptional Factor Binding Sites Created by PPARa /d /? SNPs Associated with Disease. Journal of Biosciences and Medicines 5: 81-100.
  96. Lacquemant C, Lepretre F, Pineda Torra I, Manraj M, Charpentier G, et al. (2000) Mutation screening of the PPARalpha gene in type 2 diabetes associated with coronary heart disease. Diabetes Metab 26: 393-401. [Crossref]
  97. Vohl MC, Lepage P, Gaudet D, Brewer CG, Bétard C, et al. (2000) Molecular scanning of the human PPARa gene: association of the L162v mutation with hyperapobetalipoproteinemia. J Lipid Res 41: 945-952. [Crossref]
  98. Austin ED, Loyd JE, Phillips JA III (1993) Heritable Pulmonary Arterial Hypertension, in GeneReviews(R), R.A. Pagon, et al., Editors.: Seattle (WA).
  99. Flavell DM, Pineda Torra I, Jamshidi Y, Evans D, Diamond JR, et al. (2000) Variation in the PPARalpha gene is associated with altered function in vitro and plasma lipid concentrations in Type II diabetic subjects. Diabetologia 43: 673-680. [Crossref]
  100. Shin MJ, Kanaya AM, Krauss RM (2008) Polymorphisms in the peroxisome proliferator activated receptor alpha gene are associated with levels of apolipoprotein CIII and triglyceride in African-Americans but not Caucasians. Atherosclerosis 198: 313-319. [Crossref]
  101. Robinson E, Grieve DJ (2009) Significance of peroxisome proliferator-activated receptors in the cardiovascular system in health and disease. Pharmacol Ther 122: 246-263. [Crossref]
  102. Semple RK, Chatterjee VK, O'Rahilly S (2006) PPAR gamma and human metabolic disease. J Clin Invest 116: 581-589. [Crossref]
  103. Wang Y, Qu A, Wang H (2015) Signal transducer and activator of transcription 4 in liver diseases. Int J Biol Sci 11: 448-455. [Crossref]
  104. Khanna P, Chua PJ, Bay BH, Baeg GH (2015) The JAK/STAT signaling cascade in gastric carcinoma (Review). Int J Oncol 47: 1617-1626. [Crossref]
  105. Heneghan AF, Pierre JF, Kudsk KA (2013) JAK-STAT and intestinal mucosal immunology. JAKSTAT 2: e25530. [Crossref]
  106. O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, et al. (2015) The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med 66: 311-328. [Crossref]
  107. Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM, et al., (2008) Specificity of the STAT4 genetic association for severe disease manifestations of systemic lupus erythematosus. PLoS genetics 4: e1000084. [Crossref]
  108. Jiang DK, Sun J, Cao G, Liu Y, Lin D, et al. (2013) Genetic variants in STAT4 and HLA-DQ genes confer risk of hepatitis B virus-related hepatocellular carcinoma. Nat Genet 45: 72-75. [Crossref]
  109. Hou S, Yang Z, Du L, Jiang Z, Shu Q, et al. (2012) Identification of a susceptibility locus in STAT4 for Behçet's disease in Han Chinese in a genome-wide association study. Arthritis Rheum 64: 4104-4113. [Crossref]
  110. Bolin K, Sandling JK, Zickert A, Jönsen A, Sjöwall C, et al. (2013) Association of STAT4 polymorphism with severe renal insufficiency in lupus nephritis. PLoS One 8: e84450. [Crossref]
  111. Kim ES, Kim SW, Moon CM, Park JJ, Kim T Il, et al. (2012) Interactions between IL17A, IL23R, and STAT4 polymorphisms confer susceptibility to intestinal Behcet's disease in Korean population. Life Sci 90: 740-746.
  112. Lu Y, Zhu Y, Peng J, Wang X, Wang F, et al. (2015) STAT4 genetic polymorphisms association with spontaneous clearance of hepatitis B virus infection. Immunol Res 62: 146-152. [Crossref]
  113. Yi J, Fang X, Wan Y, Wei J, Huang J (2015) STAT4 polymorphisms and diabetes risk: a meta-analysis with 18931 patients and 23833 controls. Int J Clin Exp Med 8: 3566-3572. [Crossref]
  114. Kumar A, Das S, Agrawal A, Mukhopadhyay I, Ghosh B, et al. (2015) Genetic association of key Th1/Th2 pathway candidate genes, IRF2, IL6, IFNGR2, STAT4 and IL4RA, with atopic asthma in the Indian population. J Hum Genet 60: 443-448. [Crossref]
  115. Mathur AN, Chang HC, Zisoulis DG, Stritesky GL, Yu Q, et al. (2007) Stat3 and Stat4 direct development of IL-17-secreting Th cells. J Immunol 178: 4901-4907. [Crossref]
  116. Chitnis T, Najafian N, Benou C, Salama AD, Grusby MJ, et al. (2001) Effect of targeted disruption of STAT4 and STAT6 on the induction of experimental autoimmune encephalomyelitis. J Clin Invest 108: 739-747. [Crossref]
  117. Mo C, Chearwae W, O'Malley JT, Adams SM, Kanakasabai S, et al. (2008) Stat4 isoforms differentially regulate inflammation and demyelination in experimental allergic encephalomyelitis. J Immunol 181: 5681-5690.
  118. Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, et al. (2007) STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med 357: 977-986. [Crossref]
  119. Mudter J, Weigmann B, Bartsch B, Kiesslich R, Strand D, et al. (2005) Activation pattern of signal transducers and activators of transcription (STAT) factors in inflammatory bowel diseases. Am J Gastroenterol 100: 64-72. [Crossref]
  120. Hou S, Kijlstra A, Yang P (2012) The genetics of Behçet's disease in a Chinese population. Front Med 6: 354-359. [Crossref]
  121. Liao Y, Cai B, Li Y, Chen J, Ying B, et al. (2014) Association of HLA-DP/DQ, STAT4 and IL-28B variants with HBV viral clearance in Tibetans and Uygurs in China. Liver Int 35: 886-896. [Crossref]
  122. Kim LH, Cheong HS, Namgoong S, Kim JO, Kim JH, et al. (2015) Replication of genome wide association studies on hepatocellular carcinoma susceptibility loci of STAT4 and HLA-DQ in a Korean population. Infect Genet Evol 33: 72-76. [Crossref]
  123. Liu QF, Li Y, Zhao Q-H, Wang Z-Y, Hu S, et al. (2015) Association of STAT4 rs7574865 polymorphism with susceptibility to inflammatory bowel disease: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 39: 627-636.
  124. Fan ZD, Wang FF, Huang H, Huang N, Ma HH, et al. (2015) STAT4 rs7574865 G/T and PTPN22 rs2488457 G/C polymorphisms influence the risk of developing juvenile idiopathic arthritis in Han Chinese patients. PLoS One 10: e0117389. [Crossref]
  125. Aiba Y, Yamazaki K, Nishida N, Kawashima M, Hitomi Y, et al. (2015) Disease susceptibility genes shared by primary biliary cirrhosis and Crohn's disease in the Japanese population. J Hum Genet 60: 525-531. [Crossref]
  126. Xu L, Dai WQ, Wang F, He L, Zhou YQ, et al. (2014) Association of STAT4 gene rs7574865G > T polymorphism with ulcerative colitis risk: evidence from 1532 cases and 3786 controls. Arch Med Sci 10: 419-424. [Crossref]
  127. Huang JS, Ramamurthy SK, Lin X, Le Breton GC (2004) Cell signalling through thromboxane A2 receptors. Cell Signal 16: 521-533. [Crossref]
  128. Miggin SM, Kinsella BT (1998) Expression and tissue distribution of the mRNAs encoding the human thromboxane A2 receptor (TP) alpha and beta isoforms. Biochim Biophys Acta 1425: 543-559. [Crossref]
  129. Rolin S, Masereel B, Dogné JM (2006) Prostanoids as pharmacological targets in COPD and asthma. Eur J Pharmacol 533: 89-100. [Crossref]
  130. Unoki M, Furuta S, Onouchi Y, Watanabe O, Doi S, et al. (2000) Association studies of 33 single nucleotide polymorphisms (SNPs) in 29 candidate genes for bronchial asthma: positive association a T924C polymorphism in the thromboxane A2 receptor gene. Hum Genet 106: 440-446.
  131. Leung TF, Tang NL, Lam CW, Li AM, Chan IH, et al. (2002) Thromboxane A2 receptor gene polymorphism is associated with the serum concentration of cat-specific immunoglobulin E as well as the development and severity of asthma in Chinese children. Pediatr Allergy Immunol 13: 10-17.
  132. Shin HD, Park BL, Jung JH, Wang HJ, Park HS, et al. (2003) Association of thromboxane A2 receptor (TBXA2R) with atopy and asthma. J Allergy Clin Immunol 112: 454-457. [Crossref]
  133. Takeuchi K, Mashimo Y, Shimojo N, Arima T, Inoue Y, et al. (2013) Functional variants in the thromboxane A2 receptor gene are associated with lung function in childhood-onset asthma. Clin Exp Allergy 43: 413-424. [Crossref]
  134. Buroker NE (2014) TBXA2R rSNPs, Transcriptional Factor Binding Sites and Asthma in Asians. Open Journal of Pediatrics 4: 148-161.
  135. Nowak DG, Woolard J, Amin EM, Konopatskaya O, Saleem MA, et al. (2008) Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J Cell Sci 121: 3487-3495. [Crossref]
  136. Woolard J, Bevan HS, Harper SJ, Bates DO (2009) Molecular diversity of VEGF-A as a regulator of its biological activity. Microcirculation 16: 572-592. [Crossref]
  137. Maloney J, Wang D, Duncan T, Voelkel N, Ruoss S (2000) Plasma vascular endothelial growth factor in acute mountain sickness. Chest 118: 47-52.
  138. Appenzeller O, Minko T, Pozharov V, Bonfichi M, Malcovati L, et al. (2003) Gene expression in the Andes; relevance to neurology at sea level. J Neurol Sci 207: 37-41. [Crossref]
  139. Appenzeller O, Minko T, Qualls C, Pozharov V, Gamboa J, et al. (2006) Gene expression, autonomic function and chronic hypoxia:lessons from the Andes. Clin Auton Res 16: 217-222. [Crossref]
  140. Patitucci M, Lugrin D, Pages G (2009) Angiogenic/lymphangiogenic factors and adaptation to extreme altitudes during an expedition to Mount Everest. Acta Physiol (Oxf) 196: 259-265. [Crossref]
  141. Walter R, Maggiorini M, Scherrer U, Contesse J, Reinhart WH (2001) Effects of high-altitude exposure on vascular endothelial growth factor levels in man. Eur J Appl Physiol 85: 113-117. [Crossref]
  142. Gao W, Gao Y, Zhang G, Song L, Sun B, et al. (2005) Hypoxia-induced expression of HIF-1alpha and its target genes in umbilical venous endothelial cells of Tibetans and immigrant Han. Comp Biochem Physiol C Toxicol Pharmacol 141: 93-100. [Crossref]
  143. Dorward DA, Thompson AA, Baillie JK, MacDougall M, Hirani N (2007) Change in plasma vascular endothelial growth factor during onset and recovery from acute mountain sickness. Respir Med 101: 587-594. [Crossref]
  144. Dai J, Rabie AB (2007) VEGF: an essential mediator of both angiogenesis and endochondral ossification. J Dent Res 86: 937-950. [Crossref]
  145. Harper SJ1, Bates DO (2008) VEGF-A splicing: the key to anti-angiogenic therapeutics? Nat Rev Cancer 8: 880-887. [Crossref]
  146. Xu J, Dou T, Liu C, Fu M, Huang Y, et al. (2011) The evolution of alternative splicing exons in vascular endothelial growth factor A. Gene 487: 143-150. [Crossref]
  147. Holmes DI, Zachary I (2005) The vascular endothelial growth factor (VEGF) family: angiogenic factors in health and disease. Genome Biol 6: 209. [Crossref]
  148. Buroker NE (2014) VEGFA rSNPs, transcriptional factor binding sites and human disease. J Physiol Sci 64: 73-76. [Crossref]
  149. Buroker NE (2014) VEGFA rSNPs, transcriptional factor binding sites and human disease. J Physiol Sci 64: 73-76.
  150. Buroker NE, Zhou ZN, Li K, Cen W-J, Wu X-F, et al. (2017) SNPs, linkage disequilibrium, and chronic mountain sickness in Tibetan Chinese. Hypoxia 5: 1-8.
  151. Bahreini A, Levine K, Santana-Santos L, Benos PV, Wang P, et al. (2016) Non-coding single nucleotide variants affecting estrogen receptor binding and activity. Genome Med 8: 128. [Crossref]

Article Type

Research Article

Publication history

Received date: May 25, 2017
Accepted date: June 26, 2017
Published date: June 29, 2017

Copyright

© 2017 Buroker NE. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited

Citation

Buroker NE (2017) SNPs, transcriptional factor binding sites and disease. Biomed Genet Genomics 2: DOI: 10.15761/BGG.1000132

Corresponding author

Norman E. Buroker

Department of Pediatrics, University of Washington, Seattle, WA 98195, USA

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

No Figures