Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 33(2): 150-169, 2024 Jan 07.
Article in English | MEDLINE | ID: mdl-37815931

ABSTRACT

Developmental studies have shown that the evolutionarily conserved Wnt Planar Cell Polarity (PCP) pathway is essential for the development of a diverse range of tissues and organs including the brain, spinal cord, heart and sensory organs, as well as establishment of the left-right body axis. Germline mutations in the highly conserved PCP gene VANGL2 in humans have only been associated with central nervous system malformations, and functional testing to understand variant impact has not been performed. Here we report three new families with missense variants in VANGL2 associated with heterotaxy and congenital heart disease p.(Arg169His), non-syndromic hearing loss p.(Glu465Ala) and congenital heart disease with brain defects p.(Arg135Trp). To test the in vivo impact of these and previously described variants, we have established clinically-relevant assays using mRNA rescue of the vangl2 mutant zebrafish. We show that all variants disrupt Vangl2 function, although to different extents and depending on the developmental process. We also begin to identify that different VANGL2 missense variants may be haploinsufficient and discuss evidence in support of pathogenicity. Together, this study demonstrates that zebrafish present a suitable pipeline to investigate variants of unknown significance and suggests new avenues for investigation of the different developmental contexts of VANGL2 function that are clinically meaningful.


Subject(s)
Heart Defects, Congenital , Zebrafish , Animals , Humans , Cell Polarity/genetics , Germ Cells/metabolism , Germ-Line Mutation/genetics , Heart Defects, Congenital/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
2.
J Med Genet ; 60(3): 294-300, 2023 03.
Article in English | MEDLINE | ID: mdl-35790350

ABSTRACT

BACKGROUND: Pulmonary hypoplasia, Diaphragmatic anomalies, Anophthalmia/microphthalmia and Cardiac defects delineate the PDAC syndrome. We aim to identify the cause of PDAC syndrome in patients who do not carry pathogenic variants in RARB and STRA6, which have been previously associated with this disorder. METHODS: We sequenced the exome of patients with unexplained PDAC syndrome and performed functional validation of candidate variants. RESULTS: We identified bi-allelic variants in WNT7B in fetuses with PDAC syndrome from two unrelated families. In one family, the fetus was homozygous for the c.292C>T (p.(Arg98*)) variant whereas the fetuses from the other family were compound heterozygous for the variants c.225C>G (p.(Tyr75*)) and c.562G>A (p.(Gly188Ser)). Finally, a molecular autopsy by proxy in a consanguineous couple that lost two babies due to lung hypoplasia revealed that both parents carry the p.(Arg98*) variant. Using a WNT signalling canonical luciferase assay, we demonstrated that the identified variants are deleterious. In addition, we found that wnt7bb mutant zebrafish display a defect of the swimbladder, an air-filled organ that is a structural homolog of the mammalian lung, suggesting that the function of WNT7B has been conserved during evolution for the development of these structures. CONCLUSION: Our findings indicate that defective WNT7B function underlies a form of lung hypoplasia that is associated with the PDAC syndrome, and provide evidence for involvement of the WNT-ß-catenin pathway in human lung, tracheal, ocular, cardiac, and renal development.


Subject(s)
Lung , Zebrafish , Animals , Humans , Lung/pathology , Base Sequence , Wnt Signaling Pathway , Exome , Mammals/metabolism , Wnt Proteins/metabolism
3.
Genet Med ; 25(2): 100323, 2023 02.
Article in English | MEDLINE | ID: mdl-36401616

ABSTRACT

PURPOSE: Pathogenic variants in genes encoding ubiquitin E3 ligases are known to cause neurodevelopmental syndromes. Additional neurodevelopmental disorders associated with the other genes encoding E3 ligases are yet to be identified. METHODS: Chromosomal analysis and exome sequencing were used to identify the genetic causes in 10 patients from 7 unrelated families with syndromic neurodevelopmental, seizure, and movement disorders and neurobehavioral phenotypes. RESULTS: In total, 4 patients were found to have 3 different homozygous loss-of-function (LoF) variants, and 3 patients had 4 compound heterozygous missense variants in the candidate E3 ligase gene, HECTD4, that were rare, absent from controls as homozygous, and predicted to be deleterious in silico. In 3 patients from 2 families with Angelman-like syndrome, paralog-directed candidate gene approach detected 2 LoF variants in the other candidate E3 ligase gene, UBE3C, a paralog of the Angelman syndrome E3 ligase gene, UBE3A. The RNA studies in 4 patients with LoF variants in HECTD4 and UBE3C provided evidence for the LoF effect. CONCLUSION: HECTD4 and UBE3C are novel biallelic rare disease genes, expand the association of the other HECT E3 ligase group with neurodevelopmental syndromes, and could explain some of the missing heritability in patients with a suggestive clinical diagnosis of Angelman syndrome.


Subject(s)
Angelman Syndrome , Neurodevelopmental Disorders , Humans , Angelman Syndrome/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Neurodevelopmental Disorders/genetics , Phenotype
4.
Clin Genet ; 104(4): 497-498, 2023 10.
Article in English | MEDLINE | ID: mdl-37308312

ABSTRACT

A Loss-of-function variant in ZNF808 is associated with non-syndromic neonatal diabetes in a consanguineous family with three affected siblings.


Subject(s)
Diabetes Mellitus , Humans , Infant, Newborn , Consanguinity , Diabetes Mellitus/genetics , Genes, Recessive , Pedigree , Siblings
5.
Hum Mutat ; 42(10): 1221-1228, 2021 10.
Article in English | MEDLINE | ID: mdl-34212438

ABSTRACT

Half of patients with a ciliopathy syndrome remain unsolved after initial analysis of whole exome sequencing (WES) data, highlighting the need for improved variant filtering and annotation. By candidate gene curation of WES data, combined with homozygosity mapping, we detected a homozygous predicted synonymous allele in NPHP3 in two children with hepatorenal fibrocystic disease from a consanguineous family. Analyses on patient-derived RNA shows activation of a cryptic mid-exon splice donor leading to frameshift. Remarkably, the same rare variant was detected in four additional families with hepatorenal disease from UK, US, and Saudi patient cohorts and in addition, another synonymous NPHP3 variant was identified in an unsolved case from the Genomics England 100,000 Genomes data set. We conclude that synonymous NPHP3 variants, not reported before and discarded by pathogenicity pipelines, solved several families with a ciliopathy syndrome. These findings prompt careful reassessment of synonymous variants, especially if they are rare and located in candidate genes.


Subject(s)
Liver Cirrhosis , Polycystic Kidney Diseases , Child , Genetic Diseases, Inborn , Homozygote , Humans , Kinesins , Exome Sequencing
6.
J Hum Genet ; 66(7): 689-695, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33500540

ABSTRACT

Heterozygous pathogenic variants in SLC12A2 are reported in patients with nonsyndromic hearing loss. Recently, homozygous loss-of-function variants have been reported in two patients with syndromic intellectual disability, with or without hearing loss. However, the clinical and molecular spectrum of SLC12A2 disease has yet to be characterized and confirmed. Using whole-exome sequencing, we detected a homozygous splicing variant in four patients from two independent families with severe developmental delay, microcephaly, respiratory abnormalities, and subtle dysmorphic features, with or without congenital hearing loss. We also reviewed the reported cases with pathogenic variants associated with autosomal dominant and recessive forms of the SLC12A2 disease. About 50% of the cases have syndromic and nonsyndromic congenital hearing loss. All patients harboring the recessive forms of the disease presented with severe global developmental delay. Interestingly, all reported variants are located in the c-terminal domain, suggesting a critical role of this domain for the proper function of the encoded co-transporter protein. In conclusion, our study provides an additional confirmation of the autosomal recessive SLC12A2 disease.


Subject(s)
Deafness/genetics , Genetic Predisposition to Disease , Intellectual Disability/genetics , Solute Carrier Family 12, Member 2/genetics , Brain/diagnostic imaging , Brain/pathology , Child , Child, Preschool , Deafness/complications , Deafness/diagnostic imaging , Deafness/pathology , Exome/genetics , Female , Genes, Recessive/genetics , Homozygote , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Male , Mutation/genetics , Pedigree , Phenotype , RNA Splicing/genetics , Solute Carrier Family 12, Member 2/deficiency , Exome Sequencing
7.
J Hum Genet ; 66(2): 151-159, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32759993

ABSTRACT

Biallelic variants in the USP53 gene have recently been reported to segregate with normal gamma glutamyltransferase (GGT) cholestasis. Using whole-exome sequencing (WES), we detected two USP53 homozygous variants (c.951delT; p. Phe317fs and c.1744C>T; p. Arg582*) in five additional cases, including an unpublished cousin of a previously described family with intractable itching and normal GGT cholestasis. Three patients, a child and two adults, presented with recurrent episodes of normal GGT cholestasis, consistent with a diagnosis of benign recurrent intrahepatic cholestasis (BRIC). Cholangiopathic changes, possibly autoimmune in origin, were recognized in some patients. Additional phenotypic details in one patient included an enlarged left kidney, and speech/developmental delay. Notably, two patients exhibited a complete response to rifampicin, and one responded to ursodeoxycholic acid (UDCA). Two adult patients were suspected to have autoimmune liver disease and treated with steroids. This report describes new cases of USP53 disease presenting with normal GGT cholestasis or BRIC in three children and two adults. We also describe the novel finding of a dramatic response to rifampicin. The association of cholangiopathy with normal GGT cholestasis provides a diagnostic challenge and remains poorly understood.


Subject(s)
Cholangitis/drug therapy , Cholestasis/drug therapy , Homozygote , Mutation , Rifampin/pharmacology , Ubiquitin-Specific Proteases/genetics , gamma-Glutamyltransferase/metabolism , Adolescent , Adult , Child , Cholangitis/genetics , Cholangitis/pathology , Cholestasis/genetics , Cholestasis/pathology , Female , Humans , Infant , Male , Nucleic Acid Synthesis Inhibitors/pharmacology , Pedigree , Prognosis , Exome Sequencing
8.
Clin Genet ; 99(5): 694-703, 2021 05.
Article in English | MEDLINE | ID: mdl-33495992

ABSTRACT

Protein disulfide isomerase A6 (PDIA6) is an unfolded protein response (UPR)-regulating protein. PDIA6 regulates the UPR sensing proteins, Inositol requiring enzyme 1, and EIF2AK3. Biallelic inactivation of the two genes in mice and humans resulted in embryonic lethality, diabetes, skeletal defects, and renal insufficiency. We recently showed that PDIA6 inactivation in mice caused embryonic and early lethality, diabetes and immunodeficiency. Here, we present a case with asphyxiating thoracic dystrophy (ATD) syndrome and infantile-onset diabetes. Whole exome sequencing revealed a homozygous frameshift variant in the PDIA6 gene. RNA expression was reduced in a gene dosage-dependent manner, supporting a loss-of-function effect of this variant. Phenotypic correlation with the mouse model recapitulated the growth defect and delay, early lethality, coagulation, diabetes, immunological, and polycystic kidney disease phenotypes. In general, the phenotype of the current patient is consistent with phenotypes associated with the disruption of PDIA6 and the sensors of UPR in mice and humans. This is the first study to associate ATD to the UPR gene, PDIA6. We recommend screening ATD cases with or without insulin-dependent diabetes for variants in PDIA6.


Subject(s)
Ellis-Van Creveld Syndrome/genetics , Infant, Premature, Diseases/genetics , Loss of Function Mutation , Protein Disulfide-Isomerases/genetics , Unfolded Protein Response/genetics , Abnormalities, Multiple/genetics , Alleles , Animals , Consanguinity , Ellis-Van Creveld Syndrome/diagnostic imaging , Gene Knockout Techniques , Gestational Age , Humans , Magnetic Resonance Imaging , Male , Mice , Pedigree
9.
Genet Med ; 22(12): 2071-2080, 2020 12.
Article in English | MEDLINE | ID: mdl-32741967

ABSTRACT

PURPOSE: Asparagine synthetase deficiency (ASNSD) is a rare neurometabolic disease. Patients may not demonstrate low asparagine levels, which highlights the advantage of molecular over biochemical testing in the initial work-up of ASNSD. We aimed to further delineate the ASNSD variant and phenotypic spectrum and determine the value of biochemical testing as a frontline investigation in ASNSD. METHODS: We retrospectively collected the clinical and molecular information on 13 families with ASNSD from the major metabolic clinics in Saudi Arabia. RESULTS: The major phenotypes included congenital microcephaly (100%), facial dysmorphism (100%), global developmental delay (100%), brain abnormalities (100%), spasticity (86%), and infantile-onset seizures (93%). Additional unreported phenotypes included umbilical hernia, osteopenia, eczema, lung hypoplasia, and hearing loss. Overall, seven homozygous variants accounted for ASNSD. The p.Tyr398Cys and p.Asn75Ile variants accounted for 54% of the cases. The clinical sensitivity and specificity of the proposed biochemical analysis of cerebrospinal fluid (CSF) for the detection of patients with ASNSD were 83% and 98%, respectively. CONCLUSION: Our study describes the largest reported ASNSD cohort with clinical, molecular, and biochemical characterization. Taking into consideration the suboptimal sensitivity of biochemical screening, the delineation of the phenotype variant spectrum is of diagnostic utility for accurate diagnosis, prognosis, counseling, and carrier screening.


Subject(s)
Aspartate-Ammonia Ligase , Intellectual Disability , Microcephaly , Aspartate-Ammonia Ligase/genetics , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Retrospective Studies , Saudi Arabia/epidemiology
10.
Clin Genet ; 97(3): 447-456, 2020 03.
Article in English | MEDLINE | ID: mdl-31730227

ABSTRACT

Retinal arterial macroaneurysms with supravalvular pulmonic stenosis (RAMSVPS), also known as Familial Retinal Arterial Macroaneurysms (FRAM) syndrome, is a very rare multisystem disorder. Here, we present a case series comprising ophthalmologic and systemic evaluation of patients homozygous for RAMSVPS syndrome causative IGFBP7 variant. New clinical details on 22 previously published and 8 previously unpublished patients are described. Age at first presentation ranged from 1 to 34 years. The classical feature of macroaneurysms and vascular beading involving the retinal arteries was universal. Follow up extending up to 14 years after initial diagnosis revealed recurrent episodes of bleeding and leakage from macroaneurysms in 55% and 59% of patients, respectively. The majority of patients who underwent echocardiography (18/23) showed evidence of heart involvement, most characteristically pulmonary (valvular or supravalvular) stenosis, often requiring surgical correction (12/18). Four patients died in the course of the study from complications of pulmonary stenosis, cerebral hemorrhage, and cardiac complications. Liver involvement (usually cirrhosis) was observed in eight patients. Cerebral vascular involvement was observed in one patient, and stroke was observed in two. We conclude that RAMSVPS is a recognizable syndrome characterized by a high burden of ocular and systemic morbidity, and risk of premature death. Recommendations are proposed for early detection and management of these complications.


Subject(s)
Genetic Predisposition to Disease , Insulin-Like Growth Factor Binding Proteins/genetics , Pulmonary Valve Stenosis/genetics , Retinal Arterial Macroaneurysm/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Fluorescein Angiography , Fundus Oculi , Homozygote , Humans , Infant , Male , Pulmonary Valve Stenosis/complications , Pulmonary Valve Stenosis/diagnostic imaging , Pulmonary Valve Stenosis/pathology , Retinal Arterial Macroaneurysm/complications , Retinal Arterial Macroaneurysm/diagnostic imaging , Retinal Arterial Macroaneurysm/pathology , Retinal Artery/diagnostic imaging , Retinal Artery/metabolism , Retinal Artery/pathology , Visual Acuity/genetics , Visual Acuity/physiology , Young Adult
11.
Genet Med ; 20(5): 536-544, 2018 04.
Article in English | MEDLINE | ID: mdl-29048421

ABSTRACT

PurposeHearing loss is more prevalent in the Saudi Arabian population than in other populations; however, the full range of genetic etiologies in this population is unknown. We report the genetic findings from 33 Saudi hearing-loss probands of tribal ancestry, with predominantly prelingual severe to profound hearing loss.MethodsTesting was performed over the course of 2012-2016, and involved initial GJB2 sequence and GJB6-D13S1830 deletion screening, with negative cases being reflexed to a next-generation sequencing panel with 70, 71, or 87 hearing-loss genes.ResultsA "positive" result was reached in 63% of probands, with two recurrent OTOF variants (p.Glu57* and p.Arg1792His) accountable for a third of all "positive" cases. The next most common cause was pathogenic variants in MYO7A and SLC26A4, each responsible for three "positive" cases. Interestingly, only one "positive" diagnosis had a DFNB1-related cause, due to a homozygous GJB6-D13S1830 deletion, and no sequence variants in GJB2 were detected.ConclusionOur findings implicate OTOF as a potential major contributor to hearing loss in the Saudi population, while highlighting the low contribution of GJB2, thus offering important considerations for clinical testing strategies for Saudi patients. Further screening of Saudi patients is needed to characterize the genetic spectrum in this population.


Subject(s)
Deafness/epidemiology , Deafness/genetics , Genetic Variation , Membrane Proteins/genetics , Adolescent , Adult , Alleles , Amino Acid Substitution , Child , Child, Preschool , Cohort Studies , Connexin 26 , Connexins/genetics , Deafness/diagnosis , Genetic Testing , Genotype , High-Throughput Nucleotide Sequencing , Humans , Infant , Pedigree , Phenotype , Population Surveillance , Saudi Arabia/epidemiology , Young Adult
14.
Circ Res ; 117(8): 671-83, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26195219

ABSTRACT

RATIONALE: Inflammation impairs macrophage cholesterol clearance from vascular tissues and promotes atherosclerosis. Inflammatory macrophages suppress expression of the transcription cofactor interferon regulatory factor 2-binding protein 2 (IRF2BP2), and genetic variants near IRF2BP2 associate with ischemic heart disease progression in humans. OBJECTIVES: To test whether IRF2BP2 in macrophages affects atherosclerosis in mice and humans. METHODS AND RESULTS: We generated mice that delete IRF2BP2 in macrophages. IRF2BP2-deficient macrophages worsened atherosclerosis in irradiated low-density lipoprotein receptor null-recipient mice and in apolipoprotein E null mice. IRF2BP2-deficient macrophages were inflammatory and had impaired cholesterol efflux because of their inability to activate the cholesterol transporter ABCA1 in response to cholesterol loading. Their expression of the anti-inflammatory transcription factor Krüppel-like factor 2 was markedly reduced. Promoter studies revealed that IRF2BP2 is required for MEF2-dependent activation of Krüppel-like factor 2. Importantly, restoring Krüppel-like factor 2 in IRF2BP2-deficient macrophages attenuated M1 inflammatory and rescued M2 anti-inflammatory gene activation and improved the cholesterol efflux deficit by restoring ABCA1 activation in response to cholesterol loading. In a cohort of 1066 angiographic cases and 1011 controls, homozygous carriers of a deletion polymorphism (rs3045215) in the 3' untranslated region sequence of human IRF2BP2 mRNA had a higher risk of coronary artery disease (recessive model, odds ratio [95% confidence interval]=1.560 [1.179-2.065], P=1.73E-03) and had lower IRF2BP2 (and Krüppel-like factor 2) protein levels in peripheral blood mononuclear cells. The effect of this deletion polymorphism to suppress protein expression was confirmed in luciferase reporter studies. CONCLUSION: Ablation of IRF2BP2 in macrophages worsens atherosclerosis in mice, and a deletion variant that lowers IRF2BP2 expression predisposes to coronary artery disease in humans.


Subject(s)
Atherosclerosis/prevention & control , Carrier Proteins/metabolism , Cholesterol/metabolism , Coronary Artery Disease/prevention & control , Inflammation/prevention & control , Macrophage Activation , Macrophages/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/metabolism , Aged , Aged, 80 and over , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Carrier Proteins/genetics , Case-Control Studies , Cells, Cultured , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , DNA-Binding Proteins , Disease Models, Animal , Female , Genetic Predisposition to Disease , Homozygote , Humans , Inflammation/genetics , Inflammation/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MEF2 Transcription Factors/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nuclear Proteins/genetics , Odds Ratio , Phenotype , Polymorphism, Genetic , Promoter Regions, Genetic , Protective Factors , Radiography , Receptors, LDL/deficiency , Receptors, LDL/genetics , Risk Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection
15.
Circulation ; 132(21): 1969-78, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26487755

ABSTRACT

BACKGROUND: The mechanism whereby the 9p21.3 locus confers risk for coronary artery disease remains incompletely understood. Risk alleles are associated with reduced expression of the cell cycle suppressor genes CDKN2A (p16 and p14) and CDKN2B (p15) and increased vascular smooth muscle cell proliferation. We asked whether risk alleles disrupt transcription factor binding to account for this effect. METHODS AND RESULTS: A bioinformatic screen was used to predict which of 59 single nucleotide polymorphisms at the 9p21.3 locus disrupt (or create) transcription factor binding sites. Electrophoretic mobility shift and luciferase reporter assays examined the binding and functionality of the predicted regulatory sequences. Primary human aortic smooth muscle cells (HAoSMCs) were genotyped for 9p21.3, and HAoSMCs homozygous for the risk allele showed reduced p15 and p16 levels and increased proliferation. rs10811656 and rs4977757 disrupted functional TEF-1 TEC1 AbaA domain (TEAD) transcription factor binding sites. TEAD3 and TEAD4 overexpression induced p16 in HAoSMCs homozygous for the nonrisk allele, but not for the risk allele. Transforming growth factor ß, known to activate p16 and also to interact with TEAD factors, failed to induce p16 or to inhibit proliferation of HAoSMCs homozygous for the risk allele. Knockdown of TEAD3 blocked transforming growth factor ß-induced p16 mRNA and protein expression, and dual knockdown of TEAD3 and TEAD4 markedly reduced p16 expression in heterozygous HAoSMCs. CONCLUSIONS: Here, we identify a novel mechanism whereby sequences at the 9p21.3 risk locus disrupt TEAD factor binding and TEAD3-dependent transforming growth factor ß induction of p16 in HAoSMCs. This mechanism accounts, in part, for the 9p21.3 coronary artery disease risk.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Coronary Disease/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , DNA-Binding Proteins/physiology , Muscle Proteins/physiology , Polymorphism, Single Nucleotide , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Adolescent , Adult , Alleles , Aorta/cytology , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Gene Knockdown Techniques , Genes, Reporter , Genes, p16 , Humans , Male , Middle Aged , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Recombinant Proteins/metabolism , TEA Domain Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Young Adult
18.
Curr Cardiol Rep ; 16(7): 502, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24893939

ABSTRACT

The 9p21.3 locus was the first to yield to genome-wide association studies (GWAS) seeking common genetic variants predisposing to increased risk of coronary artery atherosclerotic disease (CAD). The 59 single nucleotide polymorphisms that show highest association with CAD are clustered in a region 100,000 to 150,000 base pairs 5' to the cyclin-dependent kinase inhibitors CDKN2B (coding for p15(ink4b)) and CDKN2A (coding for p16(ink4a) and p14(ARF)). This region also covers the 3' end of a long noncoding RNA transcribed antisense to CDKN2B (CDKN2BAS, aka ANRIL for antisense noncoding RNA at the ink4 locus) whose expression has been linked to chromatin remodeling at the locus. Despite intensive investigation over the past 7 years, the functional significance of the 9p21.3 locus remains elusive. Other variants at this locus have been associated with glaucoma, glioma, and type 2 diabetes mellitus, diseases that implicate tissue-resident macrophages. Here, we review the evidence that genetic variants at 9p21.3 disrupt tissue-specific enhancers and propose new insights to guide future studies.


Subject(s)
Atherosclerosis/genetics , Chromosomes, Human, Pair 9/genetics , Genome-Wide Association Study , Genomics , Humans , Phenotype , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL