Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34050020

ABSTRACT

Genes that are primarily expressed in cochlear glia-like supporting cells (GLSs) have not been clearly associated with progressive deafness. Herein, we present a deafness locus mapped to chromosome 3p25.1 and an auditory neuropathy spectrum disorder (ANSD) gene, TMEM43, mainly expressed in GLSs. We identify p.(Arg372Ter) of TMEM43 by linkage analysis and exome sequencing in two large Asian families segregating ANSD, which is characterized by inability to discriminate speech despite preserved sensitivity to sound. The knock-in mouse with the p.(Arg372Ter) variant recapitulates a progressive hearing loss with histological abnormalities in GLSs. Mechanistically, TMEM43 interacts with the Connexin26 and Connexin30 gap junction channels, disrupting the passive conductance current in GLSs in a dominant-negative fashion when the p.(Arg372Ter) variant is introduced. Based on these mechanistic insights, cochlear implant was performed on three subjects, and speech discrimination was successfully restored. Our study highlights a pathological role of cochlear GLSs by identifying a deafness gene and its causal relationship with ANSD.


Subject(s)
Codon, Nonsense , Connexins/metabolism , Genes, Dominant , Hearing Loss, Central/genetics , Membrane Proteins/genetics , Animals , Cochlear Implantation , Female , Hearing Loss, Central/metabolism , Hearing Loss, Central/physiopathology , Hearing Loss, Central/surgery , Humans , Male , Mice , Mice, Inbred C57BL , Pedigree , Speech Perception
2.
Ear Hear ; 43(1): 1-8, 2022.
Article in English | MEDLINE | ID: mdl-34039936

ABSTRACT

Usher syndrome (USH) encompasses a group of clinically and genetically heterogenous disorders defined by the triad of sensorineural hearing loss (SNHL), vestibular dysfunction, and vision loss. USH is the most common cause of deaf blindness. USH is divided clinically into three subtypes-USH1, USH2, and USH3-based on symptom severity, progression, and age of onset. The underlying genetics of these USH forms are, however, significantly more complex, with over a dozen genes linked to the three primary clinical subtypes and other atypical USH phenotypes. Several of these genes are associated with other deaf-blindness syndromes that share significant clinical overlap with USH, pointing to the limits of a clinically based classification system. The genotype-phenotype relationships among USH forms also may vary significantly based on the location and type of mutation in the gene of interest. Understanding these genotype-phenotype relationships and associated natural disease histories is necessary for the successful development and application of gene-based therapies and precision medicine approaches to USH. Currently, the state of knowledge varies widely depending on the gene of interest. Recent studies utilizing next-generation sequencing technology have expanded the list of known pathogenic mutations in USH genes, identified new genes associated with USH-like phenotypes, and proposed algorithms to predict the phenotypic effects of specific categories of allelic variants. Further work is required to validate USH gene causality, and better define USH genotype-phenotype relationships and disease natural histories-particularly for rare mutations-to lay the groundwork for the future of USH treatment.


Subject(s)
Usher Syndromes , Genetic Association Studies , Humans , Mutation , Phenotype , Usher Syndromes/diagnosis , Usher Syndromes/genetics
3.
Proc Natl Acad Sci U S A ; 116(4): 1347-1352, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30610177

ABSTRACT

We have identified a GRAP variant (c.311A>T; p.Gln104Leu) cosegregating with autosomal recessive nonsyndromic deafness in two unrelated families. GRAP encodes a member of the highly conserved growth factor receptor-bound protein 2 (GRB2)/Sem-5/drk family of proteins, which are involved in Ras signaling; however, the function of the growth factor receptor-bound protein 2 (GRB2)-related adaptor protein (GRAP) in the auditory system is not known. Here, we show that, in mouse, Grap is expressed in the inner ear and the protein localizes to the neuronal fibers innervating cochlear and utricular auditory hair cells. Downstream of receptor kinase (drk), the Drosophila homolog of human GRAP, is expressed in Johnston's organ (JO), the fly hearing organ, and the loss of drk in JO causes scolopidium abnormalities. drk mutant flies present deficits in negative geotaxis behavior, which can be suppressed by human wild-type but not mutant GRAP. Furthermore, drk specifically colocalizes with synapsin at synapses, suggesting a potential role of such adaptor proteins in regulating actin cytoskeleton dynamics in the nervous system. Our findings establish a causative link between GRAP mutation and nonsyndromic deafness and suggest a function of GRAP/drk in hearing.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GRB2 Adaptor Protein/metabolism , Hearing Loss, Sensorineural/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Deafness/microbiology , Drosophila/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Signal Transduction/physiology
4.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30561639

ABSTRACT

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Subject(s)
Cochlea/embryology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Adult , Animals , Child , Cochlea/metabolism , Cochlea/physiology , Embryonic Development , Female , Hair Cells, Auditory/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Organogenesis , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Signal Transduction/genetics , Whole Genome Sequencing
5.
Am J Med Genet A ; 185(10): 3012-3018, 2021 10.
Article in English | MEDLINE | ID: mdl-34152076

ABSTRACT

Identifying hereditary syndromes among patients with renal cell carcinoma (RCC) is essential for surveillance of affected individuals and their at-risk family members and for treatment optimization. We conducted a chart review to determine the percentage of patients with RCC who were seen at the University of Miami Health System (UHealth), and met the American College of Medical Genetics (ACMG) and the National Society of Genetic Counselors (NSGC) genetic referral criteria at the University of Miami. Subsequently, we determined the percentage of those who went on to receive genetic evaluation. Patients selected by International Classification of Diseases (ICD) 9/10 codes corresponding to kidney cancer who were at least 18 years of age at the time of diagnosis were included in the study. We included a total of 1443 patients in the final analysis, and after exclusion of charts with incorrect ICD codes, insufficient clinical data, unknown pathology, and patients who were not seen. We used chi-square analysis, ANOVA, and t-test. Of 1443 charts reviewed, 65.7% were male and 34.3% were female. 47.7% self-identified as White, 39.2% as Hispanic, 9.1% as Black, and 4.0% as "other." The mean age of RCC diagnosis was 60.0 ± 12.4 years old. In total, 47.0% of patients met ACMG/NSGC referral criteria for genetic evaluation. Of those, only 4.2% had documented genetic assessment. This study showed a low adherence to ACMG/NSGC genetic referral guidelines at our institution and a need for increasing patients' and practitioners' awareness about the significance of genetic assessment for RCC patients and their family members.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Genetic Counseling , Genetic Predisposition to Disease , Genetic Testing , Aged , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Guidelines as Topic , Humans , Male , Middle Aged , Societies, Medical , United States/epidemiology
6.
Int J Mol Sci ; 22(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920085

ABSTRACT

Hearing loss is the most common sensory disorder with ~466 million people worldwide affected, representing about 5% of the population. A substantial portion of hearing loss is genetic. Hearing loss can either be non-syndromic, if hearing loss is the only clinical manifestation, or syndromic, if the hearing loss is accompanied by a collage of other clinical manifestations. Usher syndrome is a syndromic form of genetic hearing loss that is accompanied by impaired vision associated with retinitis pigmentosa and, in many cases, vestibular dysfunction. It is the most common cause of deaf-blindness. Currently cochlear implantation or hearing aids are the only treatments for Usher-related hearing loss. However, gene therapy has shown promise in treating Usher-related retinitis pigmentosa. Here we review how the etiologies of Usher-related hearing loss make it a good candidate for gene therapy and discuss how various forms of gene therapy could be applied to Usher-related hearing loss.


Subject(s)
Ear, Inner/pathology , Genetic Therapy , Hearing Loss/therapy , Retinitis Pigmentosa/therapy , Usher Syndromes/therapy , Ear, Inner/growth & development , Hearing Loss/etiology , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Mutation/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Usher Syndromes/etiology , Usher Syndromes/genetics , Usher Syndromes/pathology
7.
Stroke ; 51(9): 2761-2769, 2020 09.
Article in English | MEDLINE | ID: mdl-32811377

ABSTRACT

BACKGROUND AND PURPOSE: Carotid plaque is a heritable trait and a strong predictor of vascular events. Several loci have been identified for carotid plaque, however, studies in minority populations are lacking. Within a multi-ethnic cohort, we have identified individuals with extreme total carotid plaque area (TCPA), that is, higher or lower TCPA than expected based on traditional vascular risk factors (age, sex, smoking, diabetes mellitus, hypertension, etc). We hypothesized that these individuals are enriched with genetic variants accounting for the plaque burden that cannot be explained by traditional vascular risk factors. Herein, we sought to identify the genetic basis for TCPA using the multi-ethnic cohort. METHODS: Three hundred forty participants (170 from each extreme group) from 3 race/ethnic groups (53% Hispanic, 29% non-Hispanic Black, and 18% non-Hispanic White) were genotyped using a genome-wide single-nucleotide polymorphism (SNP) array and imputed using 1000Genome data. SNP-based analyses using logistic regression and gene-based analyses using VEGAS2 were performed within each race/ethnic group and then meta-analyzed. Genes with P<0.001 were included in an overrepresentation enrichment pathway analysis using WebGestalt. Promising findings were tested for association with ischemic stroke using the MEGASTROKE Consortium data set. RESULTS: No SNP or gene reached genome-wide significance. In the pathway analysis, GO:0050913 (sensory perception of bitter taste) gene set was significantly enriched (P=4.5×10-6, false discovery rate=0.04), which was confirmed in MEGASTROKE (P=0.01). Within the GO:0050913 gene set, 3 genes were associated with extreme TCPA in our study (P<0.001): TAS2R20, TAS2R50, and ITPR3. In TAS2R50, rs1376251 is the top SNP and has been associated with myocardial infarction by others. In ITPR3, a SNP with high regulatory potential (rs3818527, RegulomeScore=1f), and ITPR3 itself were among the top SNP-based and gene-based results and showed consistent evidence for association in all ethnic groups (P<0.05). CONCLUSIONS: Extreme TCPA analysis identified new candidate genes for carotid plaque in understudied populations.


Subject(s)
Carotid Artery Diseases/genetics , Plaque, Atherosclerotic/genetics , Taste/genetics , Adult , Aged , Black People , Carotid Artery Diseases/epidemiology , Carotid Artery Diseases/physiopathology , Diet , Ethnicity , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Hispanic or Latino , Humans , Male , Metabolic Networks and Pathways , Middle Aged , Phenotype , Plaque, Atherosclerotic/epidemiology , Plaque, Atherosclerotic/physiopathology , Polymorphism, Single Nucleotide/genetics , Risk Factors , United States/epidemiology , White People
8.
Hum Genet ; 139(10): 1261-1272, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32318854

ABSTRACT

Nonsyndromic cleft lip with or without cleft palate (NSCLP) is a common birth defect for which only ~ 20% of the underlying genetic variation has been identified. Variants in noncoding regions have been increasingly suggested to contribute to the missing heritability. In this study, we investigated whether variation in craniofacial enhancers contributes to NSCLP. Candidate enhancers were identified using VISTA Enhancer Browser and previous publications. Prioritization was based on patterning defects in knockout mice, deletion/duplication of craniofacial genes in animal models and results of whole exome/whole genome sequencing studies. This resulted in 20 craniofacial enhancers to be investigated. Custom amplicon-based sequencing probes were designed and used for sequencing 380 NSCLP probands (from multiplex and simplex families of non-Hispanic white (NHW) and Hispanic ethnicities) using Illumina MiSeq. The frequencies of identified variants were compared to ethnically matched European (CEU) and Los Angeles Mexican (MXL) control genomes and used for association analyses. Variants in mm427/MSX1 and hs1582/SPRY1 showed genome-wide significant association with NSCLP (p ≤ 6.4 × 10-11). In silico analysis showed that these enhancer variants may disrupt important transcription factor binding sites. Haplotypes involving these enhancers and also mm435/ABCA4 were significantly associated with NSCLP, especially in NHW (p ≤ 6.3 × 10-7). Importantly, groupwise burden analysis showed several enhancer combinations significantly over-represented in NSCLP individuals, revealing novel NSCLP pathways and supporting a polygenic inheritance model. Our findings support the role of craniofacial enhancer sequence variation in the etiology of NSCLP.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Genetic Variation , Multifactorial Inheritance , ATP-Binding Cassette Transporters/genetics , Animals , Asymptomatic Diseases , Cleft Lip/ethnology , Cleft Lip/pathology , Cleft Palate/ethnology , Cleft Palate/pathology , Embryo, Mammalian , Female , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Hispanic or Latino , Humans , MSX1 Transcription Factor/genetics , Male , Membrane Proteins/genetics , Mice , Pedigree , Phosphoproteins/genetics , United States , White People
9.
Ann Neurol ; 85(3): 316-330, 2019 03.
Article in English | MEDLINE | ID: mdl-30706531

ABSTRACT

OBJECTIVE: Genetic modifiers in rare disease have long been suspected to contribute to the considerable variance in disease expression, including Charcot-Marie-Tooth disease type 1A (CMT1A). To address this question, the Inherited Neuropathy Consortium collected a large standardized sample of such rare CMT1A patients over a period of 8 years. CMT1A is caused in most patients by a uniformly sized 1.5 Mb duplication event involving the gene PMP22. METHODS: We genotyped DNA samples from 971 CMT1A patients on Illumina BeadChips. Genome-wide analysis was performed in a subset of 330 of these patients, who expressed the extremes of a hallmark symptom: mild and severe foot dorsiflexion strength impairment. SIPA1L2 (signal-induced proliferation-associated 1 like 2), the top identified candidate modifier gene, was expressed in the peripheral nerve, and our functional studies identified and confirmed interacting proteins using coimmunoprecipitation analysis, mass spectrometry, and immunocytochemistry. Chromatin immunoprecipitation and in vitro siRNA experiments were used to analyze gene regulation. RESULTS: We identified significant association of 4 single nucleotide polymorphisms (rs10910527, rs7536385, rs4649265, rs1547740) in SIPA1L2 with foot dorsiflexion strength (p < 1 × 10-7 ). Coimmunoprecipitation and mass spectroscopy studies identified ß-actin and MYH9 as SIPA1L2 binding partners. Furthermore, we show that SIPA1L2 is part of a myelination-associated coexpressed network regulated by the master transcription factor SOX10. Importantly, in vitro knockdown of SIPA1L2 in Schwannoma cells led to a significant reduction of PMP22 expression, hinting at a potential strategy for drug development. INTERPRETATION: SIPA1L2 is a potential genetic modifier of CMT1A phenotypic expressions and offers a new pathway to therapeutic interventions. ANN NEUROL 2019;85:316-330.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Foot/physiopathology , GTPase-Activating Proteins/genetics , Genes, Modifier/genetics , Muscle Weakness/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Charcot-Marie-Tooth Disease/physiopathology , Child , Child, Preschool , Female , Gene Expression Regulation , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , In Vitro Techniques , Male , Middle Aged , Muscle Weakness/physiopathology , Myelin Proteins/genetics , Neurilemmoma/genetics , Phenotype , Polymorphism, Single Nucleotide , Rats , Severity of Illness Index , Young Adult
10.
Am J Hum Genet ; 98(4): 744-54, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27018472

ABSTRACT

Cleft palate (CP) is a common birth defect occurring in 1 in 2,500 live births. Approximately half of infants with CP have a syndromic form, exhibiting other physical and cognitive disabilities. The other half have nonsyndromic CP, and to date, few genes associated with risk for nonsyndromic CP have been characterized. To identify such risk factors, we performed a genome-wide association study of this disorder. We discovered a genome-wide significant association with a missense variant in GRHL3 (p.Thr454Met [c.1361C>T]; rs41268753; p = 4.08 × 10(-9)) and replicated the result in an independent sample of case and control subjects. In both the discovery and replication samples, rs41268753 conferred increased risk for CP (OR = 8.3, 95% CI 4.1-16.8; OR = 2.16, 95% CI 1.43-3.27, respectively). In luciferase transactivation assays, p.Thr454Met had about one-third of the activity of wild-type GRHL3, and in zebrafish embryos, perturbed periderm development. We conclude that this mutation is an etiologic variant for nonsyndromic CP and is one of few functional variants identified to date for nonsyndromic orofacial clefting. This finding advances our understanding of the genetic basis of craniofacial development and might ultimately lead to improvements in recurrence risk prediction, treatment, and prognosis.


Subject(s)
Cleft Palate/genetics , DNA-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Animals , Case-Control Studies , Cleft Palate/diagnosis , Disease Models, Animal , Ethnicity/genetics , Genetic Loci , Genome-Wide Association Study , Genotyping Techniques , Humans , Mutation, Missense , Risk Factors , Zebrafish/embryology , Zebrafish/genetics
11.
Genet Med ; 21(12): 2744-2754, 2019 12.
Article in English | MEDLINE | ID: mdl-31273342

ABSTRACT

PURPOSE: To determine the genetic etiology of deafness in a family (HN-SD01) with autosomal dominant nonsyndromic hearing loss (NSHL). METHODS: Stepwise genetic analysis was performed on family HN-SD01, including hotspot variant screening, exome sequencing, virtual hearing loss gene panel, and genome-wide linkage analysis. Targeted region sequencing was used to screen ABCC1 in additional cases. Cochlear expression of Abcc1 was evaluated by messenger RNA (mRNA) and protein levels. Computational prediction, immunofluorescence, real-time quantitative polymerase chain reaction, and flow cytometry were conducted to uncover functional consequences of candidate variants. RESULTS: Stepwise genetic analysis identified a heterozygous missense variant, ABCC1:c.1769A>G (p.Asn590Ser), cosegregating with phenotype in HN-SD01. Screening of ABCC1 in an additional 217 cases identified candidate pathogenic variants c.692G>A (p.Gly231Asp) in a sporadic case and c.887A>T (p.Glu296Val) in a familial proband. Abcc1 expressed in stria vascularis and auditory nerve of mouse cochlea. Immunofluorescence showed p.Asn590Ser distributed in cytomembrane and cytoplasm, while wild type was shown only in cytomembrane. Besides, it generated unstable mRNA and decreased efflux capacity of ABCC1. CONCLUSION: Stepwise genetic analysis is efficient to analyze the genetic etiology of NSHL. Variants in ABCC1 are linked with NSHL and suggest an important role of extruding pumps in maintaining cochlea function.


Subject(s)
Deafness/genetics , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Adolescent , Adult , Aged , Animals , China , Cochlea/metabolism , Deafness/etiology , Deafness/metabolism , Exome , Family , Female , Genetic Linkage , Genetic Testing , Genotype , Hearing Loss/genetics , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Mutation, Missense , Pedigree , Phenotype , Sequence Analysis, DNA/methods , Exome Sequencing
12.
Int J Audiol ; 58(12): 834-850, 2019 12.
Article in English | MEDLINE | ID: mdl-31264897

ABSTRACT

Objective: Universal newborn hearing screening (UNHS) uses otoacoustic emissions testing (OAE) and auditory brainstem response testing (ABR) to screen all newborn infants for hearing loss (HL), but may not identify infants with mild HL at birth or delayed onset HL. The purpose of this review is to examine the role of genetic screening to diagnose children with pre-lingual HL that is not detected at birth by determining the rate of children who pass UNHS but have a positive genetic screening. This includes a summary of the current UNHS and its limitations and a review of genetic mutations and screening technologies used to detect patients with an increased risk of undiagnosed pre-lingual HL.Design: Literature review of studies that compare UNHS with concurrent genetic screening.Study sample: Infants and children with HLResults: Sixteen studies were included encompassing 137,895 infants. Pathogenic mutations were detected in 8.66% of patients. In total, 545 patients passed the UNHS but had a positive genetic screening. The average percentage of patients who passed UNHS but had a positive genetic screening was 1.4%.Conclusions: This review demonstrates the positive impact of concurrent genetic screening with UNHS to identify patients with pre-lingual HL.


Subject(s)
Genetic Testing , Hearing Loss/diagnosis , Hearing Tests , Neonatal Screening , Humans , Infant, Newborn
13.
Hum Mol Genet ; 25(13): 2862-2872, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27033726

ABSTRACT

Orofacial clefts (OFCs), which include non-syndromic cleft lip with or without cleft palate (CL/P), are among the most common birth defects in humans, affecting approximately 1 in 700 newborns. CL/P is phenotypically heterogeneous and has a complex etiology caused by genetic and environmental factors. Previous genome-wide association studies (GWASs) have identified at least 15 risk loci for CL/P. As these loci do not account for all of the genetic variance of CL/P, we hypothesized the existence of additional risk loci. We conducted a multiethnic GWAS in 6480 participants (823 unrelated cases, 1700 unrelated controls and 1319 case-parent trios) with European, Asian, African and Central and South American ancestry. Our GWAS revealed novel associations on 2p24 near FAM49A, a gene of unknown function (P = 4.22 × 10-8), and 19q13 near RHPN2, a gene involved in organizing the actin cytoskeleton (P = 4.17 × 10-8). Other regions reaching genome-wide significance were 1p36 (PAX7), 1p22 (ARHGAP29), 1q32 (IRF6), 8q24 and 17p13 (NTN1), all reported in previous GWASs. Stratification by ancestry group revealed a novel association with a region on 17q23 (P = 2.92 × 10-8) among individuals with European ancestry. This region included several promising candidates including TANC2, an oncogene required for development, and DCAF7, a scaffolding protein required for craniofacial development. In the Central and South American ancestry group, significant associations with loci previously identified in Asian or European ancestry groups reflected their admixed ancestry. In summary, we have identified novel CL/P risk loci and suggest new genes involved in craniofacial development, confirming the highly heterogeneous etiology of OFCs.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Asian People/genetics , Black People/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 2/genetics , Ethnicity , Female , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide/genetics , Risk Factors , White People/genetics
14.
Hum Genet ; 137(6-7): 437-446, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29860631

ABSTRACT

Identification of genes with variants causing non-syndromic hearing loss (NSHL) is challenging due to genetic heterogeneity. The difficulty is compounded by technical limitations that in the past prevented comprehensive gene identification. Recent advances in technology, using targeted capture and next-generation sequencing (NGS), is changing the face of gene identification and making it possible to rapidly and cost-effectively sequence the whole human exome. Here, we characterize a five-generation Chinese family with progressive, postlingual autosomal dominant nonsyndromic hearing loss (ADNSHL). By combining population-specific mutation arrays, targeted deafness genes panel, whole exome sequencing (WES), we identified PDE1C (Phosphodiesterase 1C) c.958G>T (p.A320S) as the disease-associated variant. Structural modeling insights into p.A320S strongly suggest that the sequence alteration will likely affect the substrate-binding pocket of PDE1C. By whole-mount immunofluorescence on postnatal day 3 mouse cochlea, we show its expression in outer (OHC) and inner (IHC) hair cells cytosol co-localizing with Lamp-1 in lysosomes. Furthermore, we provide evidence that the variant alters the PDE1C hydrolytic activity for both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Collectively, our findings indicate that the c.958G>T variant in PDE1C may disrupt the cross talk between cGMP-signaling and cAMP pathways in Ca2+ homeostasis.


Subject(s)
Cochlea/growth & development , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Deafness/genetics , Lysosomal Membrane Proteins/genetics , Animals , Asian People/genetics , Cochlea/metabolism , Cochlea/physiopathology , Cyclic AMP/genetics , Deafness/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/genetics , Genes, Dominant , Genotype , Homeostasis/genetics , Humans , Lysosomes/genetics , Male , Mice , Mutation , Pedigree , Exome Sequencing
15.
Hum Genet ; 137(6-7): 479-486, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29982980

ABSTRACT

While recent studies have revealed a substantial portion of the genes underlying human hearing loss, the extensive genetic landscape has not been completely explored. Here, we report a loss-of-function variant (c.72delA) in MPZL2 in three unrelated multiplex families from Turkey and Iran with autosomal recessive nonsyndromic hearing loss. The variant co-segregates with moderate sensorineural hearing loss in all three families. We show a shared haplotype flanking the variant in our families implicating a single founder. While rare in other populations, the allele frequency of the variant is ~ 0.004 in Ashkenazi Jews, suggesting that it may be an important cause of moderate hearing loss in that population. We show that Mpzl2 is expressed in mouse inner ear, and the protein localizes in the auditory inner and outer hair cells, with an asymmetric subcellular localization. We thus present MPZL2 as a novel gene associated with sensorineural hearing loss.


Subject(s)
Cell Adhesion Molecules/genetics , Deafness/genetics , Hair Cells, Auditory, Inner/metabolism , Hearing Loss, Sensorineural/genetics , Animals , Deafness/physiopathology , Ear, Inner/growth & development , Ear, Inner/physiopathology , Female , Gene Frequency , Genes, Recessive , Hair Cells, Auditory, Inner/pathology , Haplotypes/genetics , Hearing Loss, Sensorineural/physiopathology , Humans , Iran/epidemiology , Jews/genetics , Male , Mice , Mutation , Pedigree , Schwann Cells/pathology , Turkey
16.
J Stroke Cerebrovasc Dis ; 27(3): 645-652, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29103864

ABSTRACT

BACKGROUND: Apolipoprotein E (APOE) polymorphism has previously been associated with carotid intima-media thickness (cIMT) in predominantly Caucasian populations. We sought to test the strength of the relationship between APOE-ε4 carrier status and subclinical atherosclerosis in a tri-ethnic population with a large Hispanic representation. METHODS: We assessed the association between APOE polymorphism and cIMT and plaque burden among 1243 stroke-free individuals (mean age 69 years, 65% Hispanic, 18% black, 17% white) using a sequence of multivariable regression models. RESULTS: After adjusting for demographics, vascular risk factors and plasma low-density lipoprotein (LDL) levels, APOE-ε4 carrier status was positively associated with cIMT (mean difference, .013 mm; 95% confidence interval, .003-.023 mm). The APOE-ε4 association with cIMT appeared to be segment-specific with greater differences in IMT between APOE-ε4 carriers and noncarriers in the common carotid artery (CCA, .014 mm) and bifurcation (.017 mm) than in the internal carotid artery (ICA) IMT (.007 mm). This relationship was not modified by race-ethnicity. Presence of diabetes modified the ε4-cIMT relationship in CCA (P = .045) and ICA (P = .046). APOE-ε4 carrier status was not associated with plaque presence or plaque area. CONCLUSIONS: APOE-ε4 carriers had elevated cIMT independent of demographics and vascular risk factors including LDL levels. Diabetes was an effect modifier of the relationship between APOE-ε4 and IMT, such that ε4 carriers with diabetes had greater IMT in the CCA and ICA than those without diabetes. The APOE-IMT relationship was not modified by race-ethnicity.


Subject(s)
Apolipoprotein E4/genetics , Carotid Artery Diseases/genetics , Polymorphism, Genetic , Black or African American/genetics , Aged , Asymptomatic Diseases , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/ethnology , Carotid Intima-Media Thickness , Diabetes Mellitus/ethnology , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Hispanic or Latino/genetics , Humans , Male , Middle Aged , New York City/epidemiology , Phenotype , Plaque, Atherosclerotic , Risk Factors , White People/genetics
17.
Am J Hum Genet ; 94(3): 373-84, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560519

ABSTRACT

Exome sequencing in families affected by rare genetic disorders has the potential to rapidly identify new disease genes (genes in which mutations cause disease), but the identification of a single causal mutation among thousands of variants remains a significant challenge. We developed a scoring algorithm to prioritize potential causal variants within a family according to segregation with the phenotype, population frequency, predicted effect, and gene expression in the tissue(s) of interest. To narrow the search space in families with multiple affected individuals, we also developed two complementary approaches to exome-based mapping of autosomal-dominant disorders. One approach identifies segments of maximum identity by descent among affected individuals; the other nominates regions on the basis of shared rare variants and the absence of homozygous differences between affected individuals. We showcase our methods by using exome sequence data from families affected by autosomal-dominant retinitis pigmentosa (adRP), a rare disorder characterized by night blindness and progressive vision loss. We performed exome capture and sequencing on 91 samples representing 24 families affected by probable adRP but lacking common disease-causing mutations. Eight of 24 families (33%) were revealed to harbor high-scoring, most likely pathogenic (by clinical assessment) mutations affecting known RP genes. Analysis of the remaining 17 families identified candidate variants in a number of interesting genes, some of which have withstood further segregation testing in extended pedigrees. To empower the search for Mendelian-disease genes in family-based sequencing studies, we implemented them in a cross-platform-compatible software package, MendelScan, which is freely available to the research community.


Subject(s)
Chromosome Mapping/methods , Exome , Retinitis Pigmentosa/genetics , Cohort Studies , Computational Biology/methods , Databases, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Genes, Dominant , Genetic Linkage , Genetic Markers , Genetic Variation , Genotype , Heterozygote , Humans , Male , Mutation , Pedigree , Phenotype , Retina/metabolism , Software
18.
Ethn Dis ; 27(1): 11-14, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28115816

ABSTRACT

Sickle cell anemia (SCA) is a common hematological disorder among individuals of African descent in the United States; the disorder results in the production of abnormal hemoglobin. It is caused by homozygosity for a genetic mutation in HBB; rs334. While the presence of a single mutation (sickle cell trait, SCT) has long been considered a benign trait, recent research suggests that SCT is associated with renal dysfunction, including a decrease in estimated glomerular filtration rate (eGFR) and increased risk of chronic kidney disease (CKD) in African Americans. It is currently unknown whether similar associations are observed in Hispanics. Therefore, our study aimed to determine if SCT is associated with mean eGFR and CKD in a sample of 340 Dominican Hispanics from the Northern Manhattan Study. Using regression analyses, we tested rs334 for association with eGFR and CKD, adjusting for age and sex. eGFR was estimated using the Chronic Kidney Disease Epidemiology Collaboration equation and CKD was defined as eGFR < 60 mL/min/1.73 m2. Within our sample, there were 16 individuals with SCT (SCT carriers). We found that SCT carriers had a mean eGFR that was 12.12 mL/min/1.73m2 lower than non-carriers (P=.002). Additionally, SCT carriers had 2.72 times higher odds of CKD compared with non-carriers (P=.09). Taken together, these novel results show that Hispanics with SCT, as found among African Americans with SCT, may also be at increased risk for kidney disease.


Subject(s)
Glomerular Filtration Rate/genetics , Hispanic or Latino/genetics , Renal Insufficiency, Chronic/genetics , Sickle Cell Trait/genetics , Adult , Black or African American/genetics , Aged , Anemia, Sickle Cell/genetics , Cohort Studies , Dominican Republic/ethnology , Female , Genetic Carrier Screening , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , New York City , Odds Ratio , United States
19.
Hum Mutat ; 37(5): 481-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26841241

ABSTRACT

Hereditary hearing loss (HL) is characterized by both allelic and locus genetic heterogeneity. Both recessive and dominant forms of HL may be caused by different mutations in the same deafness gene. In a family with post-lingual progressive non-syndromic deafness, whole-exome sequencing of genomic DNA from five hearing-impaired relatives revealed a single variant, p.Gly488Glu (rs145970949:G>A) in MYO3A, co-segregating with HL as an autosomal dominant trait. This amino acid change, predicted to be pathogenic, alters a highly conserved residue in the motor domain of MYO3A. The mutation severely alters the ATPase activity and motility of the protein in vitro, and the mutant protein fails to accumulate in the filopodia tips in COS7 cells. However, the mutant MYO3A was able to reach the tips of organotypic inner ear culture hair cell stereocilia, raising the possibility of a local effect on positioning of the mechanoelectrical transduction (MET) complex at the stereocilia tips. To address this hypothesis, we investigated the interaction of MYO3A with the cytosolic tail of the integral tip-link protein protocadherin 15 (PCDH15), a core component of MET complex. Interestingly, we uncovered a novel interaction between MYO3A and PCDH15 shedding new light on the function of myosin IIIA at stereocilia tips.


Subject(s)
Cadherins/metabolism , Deafness/genetics , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type III/genetics , Myosin Type III/metabolism , Polymorphism, Single Nucleotide , Amino Acid Substitution , Animals , COS Cells , Cadherin Related Proteins , Cells, Cultured , Child , Child, Preschool , Chlorocebus aethiops , Deafness/metabolism , Female , Genetic Predisposition to Disease , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/metabolism , Humans , Male , Middle Aged , Pedigree
20.
Ann Hum Genet ; 80(6): 327-331, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27870113

ABSTRACT

Identifying the genetic etiology in a person with hearing loss (HL) is challenging due to the extreme genetic heterogeneity in HL and the population-specific variability. In this study, after excluding GJB2 variants, targeted resequencing of 180 deafness-related genes revealed the causative variants in 11 of 19 (58%) Brazilian probands with autosomal recessive HL. Identified pathogenic variants were in MYO15A (10 families) and CLDN14 (one family). Remarkably, the MYO15A p.(Val1400Met) variant was identified in eight families from the city of Monte Santo in the northeast region of Brazil. Haplotype analysis of this variant was consistent with a single founder. No other cases with this variant were detected among 105 simplex cases from other cities of northeastern Brazil, suggesting that this variant is confined to a geographical region. This study suggests that it is feasible to develop population-specific screening for deafness variants once causative variants are identified in different geographical groups.


Subject(s)
Hearing Loss/genetics , Myosins/genetics , Brazil , Case-Control Studies , Claudins/genetics , DNA Mutational Analysis , Founder Effect , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Mutation, Missense
SELECTION OF CITATIONS
SEARCH DETAIL