Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 166(1): 88-101, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27293190

RESUMO

Antibodies to DNA and chromatin drive autoimmunity in systemic lupus erythematosus (SLE). Null mutations and hypomorphic variants of the secreted deoxyribonuclease DNASE1L3 are linked to familial and sporadic SLE, respectively. We report that DNASE1L3-deficient mice rapidly develop autoantibodies to DNA and chromatin, followed by an SLE-like disease. Circulating DNASE1L3 is produced by dendritic cells and macrophages, and its levels inversely correlate with anti-DNA antibody response. DNASE1L3 is uniquely capable of digesting chromatin in microparticles released from apoptotic cells. Accordingly, DNASE1L3-deficient mice and human patients have elevated DNA levels in plasma, particularly in circulating microparticles. Murine and human autoantibody clones and serum antibodies from human SLE patients bind to DNASE1L3-sensitive chromatin on the surface of microparticles. Thus, extracellular microparticle-associated chromatin is a potential self-antigen normally digested by circulating DNASE1L3. The loss of this tolerance mechanism can contribute to SLE, and its restoration may represent a therapeutic opportunity in the disease.


Assuntos
Autoanticorpos/imunologia , Micropartículas Derivadas de Células/química , Cromatina/imunologia , DNA/imunologia , Endodesoxirribonucleases/genética , Lúpus Eritematoso Sistêmico/imunologia , Animais , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Endodesoxirribonucleases/deficiência , Endodesoxirribonucleases/metabolismo , Humanos , Células Jurkat , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Hum Mol Genet ; 32(1): 46-54, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-35913761

RESUMO

Glutaminase deficiency has recently been associated with ataxia and developmental delay due to repeat expansions in the 5'UTR of the glutaminase (GLS) gene. Patients with the described GLS repeat expansion may indeed remain undiagnosed due to the rarity of this variant, the challenge of its detection and the recency of its discovery. In this study, we combined advanced bioinformatics screening of ~3000 genomes and ~1500 exomes with optical genome mapping and long-read sequencing for confirmation studies. We identified two GLS families, previously intensely and unsuccessfully analyzed. One family carries an unusual and complex structural change involving a homozygous repeat expansion nested within a quadruplication event in the 5'UTR of GLS. Glutaminase deficiency and its metabolic consequences were validated by in-depth biochemical analysis. The identified GLS patients showed progressive early-onset ataxia, cognitive deficits, pyramidal tract damage and optic atrophy, thus demonstrating susceptibility of several specific neuron populations to glutaminase deficiency. This large-scale screening study demonstrates the ability of bioinformatics analysis-validated by latest state-of-the-art technologies (optical genome mapping and long-read sequencing)-to effectively flag complex repeat expansions using short-read datasets and thus facilitate diagnosis of ultra-rare disorders.


Assuntos
Glutaminase , Humanos , Regiões 5' não Traduzidas , Ataxia/diagnóstico , Ataxia/genética , Glutaminase/genética
3.
Proc Natl Acad Sci U S A ; 119(26): e2204084119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35727972

RESUMO

Discovery of deafness genes and elucidating their functions have substantially contributed to our understanding of hearing physiology and its pathologies. Here we report on DNA variants in MINAR2, encoding membrane integral NOTCH2-associated receptor 2, in four families underlying autosomal recessive nonsyndromic deafness. Neurologic evaluation of affected individuals at ages ranging from 4 to 80 y old does not show additional abnormalities. MINAR2 is a recently annotated gene with limited functional understanding. We detected three MINAR2 variants, c.144G > A (p.Trp48*), c.412_419delCGGTTTTG (p.Arg138Valfs*10), and c.393G > T, in 13 individuals with congenital- or prelingual-onset severe-to-profound sensorineural hearing loss (HL). The c.393G > T variant is shown to disrupt a splice donor site. We show that Minar2 is expressed in the mouse inner ear, with the protein localizing mainly in the hair cells, spiral ganglia, the spiral limbus, and the stria vascularis. Mice with loss of function of the Minar2 protein (Minar2tm1b/tm1b) present with rapidly progressive sensorineural HL associated with a reduction in outer hair cell stereocilia in the shortest row and degeneration of hair cells at a later age. We conclude that MINAR2 is essential for hearing in humans and mice and its disruption leads to sensorineural HL. Progressive HL observed in mice and in some affected individuals and as well as relative preservation of hair cells provides an opportunity to interfere with HL using genetic therapies.


Assuntos
Perda Auditiva Neurossensorial , Receptor Notch2 , Receptores de Superfície Celular , Animais , Perda Auditiva Neurossensorial/genética , Humanos , Mutação com Perda de Função , Camundongos , Receptor Notch2/genética , Receptor Notch2/metabolismo , Receptores de Superfície Celular/genética , Estereocílios/metabolismo
4.
Hum Genomics ; 17(1): 103, 2023 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-37996878

RESUMO

BACKGROUND: We analyzed the genetic causes of sensorineural hearing loss in racial and ethnic minorities of South Florida by reviewing demographic, phenotypic, and genetic data on 136 patients presenting to the Hereditary Hearing Loss Clinic at the University of Miami. In our retrospective chart review, of these patients, half self-identified as Hispanic, and the self-identified racial distribution was 115 (86%) White, 15 (11%) Black, and 6 (4%) Asian. Our analysis helps to reduce the gap in understanding the prevalence, impact, and genetic factors related to hearing loss among diverse populations. RESULTS: The causative gene variant or variants were identified in 54 (40%) patients, with no significant difference in the molecular diagnostic rate between Hispanics and Non-Hispanics. However, the total solve rate based on race was 40%, 47%, and 17% in Whites, Blacks, and Asians, respectively. In Non-Hispanic Whites, 16 different variants were identified in 13 genes, with GJB2 (32%), MYO7A (11%), and SLC26A4 (11%) being the most frequently implicated genes. In White Hispanics, 34 variants were identified in 20 genes, with GJB2 (22%), MYO7A (7%), and STRC-CATSPER2 (7%) being the most common. In the Non-Hispanic Black cohort, the gene distribution was evenly dispersed, with 11 variants occurring in 7 genes, and no variant was identified in 3 Hispanic Black probands. For the Asian cohort, only one gene variant was found out of 6 patients. CONCLUSION: This study demonstrates that the diagnostic rate of genetic studies in hearing loss varies according to race in South Florida, with more heterogeneity in racial and ethnic minorities. Further studies to delineate deafness gene variants in underrepresented populations, such as African Americans/Blacks from Hispanic groups, are much needed to reduce racial and ethnic disparities in genetic diagnoses.


Assuntos
Perda Auditiva Neurossensorial , Humanos , Asiático/genética , Negro ou Afro-Americano/genética , DNA/genética , Florida/epidemiologia , Perda Auditiva Neurossensorial/epidemiologia , Perda Auditiva Neurossensorial/genética , Hispânico ou Latino/genética , Peptídeos e Proteínas de Sinalização Intercelular , Estudos Retrospectivos , Brancos/genética
5.
Am J Med Genet A ; 194(4): e63481, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37984424

RESUMO

Chanarin-Dorfman syndrome is an autosomal recessively inherited disorder characterized by ichthyosis, sensorineural hearing loss, and hepatic dysfunction. We report on a 60-year-old female of Venezuelan descent who presented with congenital ichthyosis, progressive sensorineural hearing loss, and liver cirrhosis. We identify a heterozygous copy number deletion involving exon 1 and another heterozygous deletion involving exon 3 of the ABHD5 gene. Exon 2 is preserved. Both deletions were confirmed with RT-PCR. RNAseq from peripheral blood shows a reduction of ABHD5 expression overall and an absence of exon 3 expression, confirming the deleterious effects of the identified deletions. We present exonic deletions as a potentially common type of ABHD5 variation.


Assuntos
Perda Auditiva Neurossensorial , Eritrodermia Ictiosiforme Congênita , Ictiose , Erros Inatos do Metabolismo Lipídico , Doenças Musculares , Feminino , Humanos , Pessoa de Meia-Idade , Eritrodermia Ictiosiforme Congênita/complicações , Eritrodermia Ictiosiforme Congênita/diagnóstico , Eritrodermia Ictiosiforme Congênita/genética , Erros Inatos do Metabolismo Lipídico/genética , Doenças Musculares/genética , Ictiose/complicações , Ictiose/diagnóstico , Ictiose/genética , Cirrose Hepática , Perda Auditiva Neurossensorial/complicações , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética
6.
Am J Med Genet A ; 194(6): e63556, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38348595

RESUMO

Phenotypic features of a hereditary connective tissue disorder, including craniofacial characteristics, hyperextensible skin, joint laxity, kyphoscoliosis, arachnodactyly, inguinal hernia, and diverticulosis associated with biallelic pathogenic variants in EFEMP1 have been previously described in four patients. Genome sequencing on a proband and her mother with comparable phenotypic features revealed that both patients were heterozygous for a stop-gain variant c.1084C>T (p.Arg362*). Complementary RNA-seq on fibroblasts revealed significantly reduced levels of mutant EFEMP1 transcript. Considering the absence of other molecular explanations, we extrapolated that EFEMP1 could be the cause of the patient's phenotypes. Furthermore, nonsense-mediated decay was demonstrated for the mutant allele as the principal mechanism for decreased levels of EFEMP1 mRNA. We provide strong clinical and genetic evidence for the haploinsufficiency of EFEMP1 due to nonsense-medicated decay to cause severe kyphoscoliosis, generalized hypermobility of joints, high and narrow arched palate, and potentially severe diverticulosis. To the best of our knowledge, this is the first report of an autosomal dominant EFEMP1-associated hereditary connective tissue disorder and therefore expands the phenotypic spectrum of EFEMP1 related disorders.


Assuntos
Doenças do Tecido Conjuntivo , Proteínas da Matriz Extracelular , Haploinsuficiência , Síndrome de Marfan , Fenótipo , Humanos , Haploinsuficiência/genética , Feminino , Síndrome de Marfan/genética , Síndrome de Marfan/patologia , Proteínas da Matriz Extracelular/genética , Doenças do Tecido Conjuntivo/genética , Doenças do Tecido Conjuntivo/patologia , Linhagem , Mutação/genética , Degradação do RNAm Mediada por Códon sem Sentido/genética , Masculino , Adulto , Alelos , Predisposição Genética para Doença , Criança
7.
Am J Med Genet A ; 194(6): e63563, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38352997

RESUMO

Autosomal dominant sensorineural hearing loss (ADSNHL) is a genetically heterogeneous disorder caused by pathogenic variants in various genes, including MYH14. However, the interpretation of pathogenicity for MYH14 variants remains a challenge due to incomplete penetrance and the lack of functional studies and large families. In this study, we performed exome sequencing in six unrelated families with ADSNHL and identified five MYH14 variants, including three novel variants. Two of the novel variants, c.571G > C (p.Asp191His) and c.571G > A (p.Asp191Asn), were classified as likely pathogenic using ACMG and Hearing Loss Expert panel guidelines. In silico modeling demonstrated that these variants, along with p.Gly1794Arg, can alter protein stability and interactions among neighboring molecules. Our findings suggest that MYH14 causative variants may be more contributory and emphasize the importance of considering this gene in patients with nonsyndromic mainly post-lingual severe form of hearing loss. However, further functional studies are needed to confirm the pathogenicity of these variants.


Assuntos
Sequenciamento do Exoma , Perda Auditiva Neurossensorial , Cadeias Pesadas de Miosina , Miosina Tipo II , Linhagem , Humanos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Feminino , Masculino , Cadeias Pesadas de Miosina/genética , Adulto , Mutação/genética , Predisposição Genética para Doença , Criança , Genes Dominantes , Pessoa de Meia-Idade , Adolescente
8.
Brain ; 146(7): 3003-3013, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-36729635

RESUMO

There are few causes of treatable neurodevelopmental diseases described to date. Branched-chain ketoacid dehydrogenase kinase (BCKDK) deficiency causes branched-chain amino acid (BCAA) depletion and is linked to a neurodevelopmental disorder characterized by autism, intellectual disability and microcephaly. We report the largest cohort of patients studied, broadening the phenotypic and genotypic spectrum. Moreover, this is the first study to present newborn screening findings and mid-term clinical outcome. In this cross-sectional study, patients with a diagnosis of BCKDK deficiency were recruited via investigators' practices through a MetabERN initiative. Clinical, biochemical and genetic data were collected. Dried blood spot (DBS) newborn screening (NBS) amino acid profiles were retrieved from collaborating centres and compared to a healthy newborn reference population. Twenty-one patients with BCKDK mutations were included from 13 families. Patients were diagnosed between 8 months and 16 years (mean: 5.8 years, 43% female). At diagnosis, BCAA levels (leucine, valine and isoleucine) were below reference values in plasma and in CSF. All patients had global neurodevelopmental delay; 18/21 had gross motor function (GMF) impairment with GMF III or worse in 5/18, 16/16 intellectual disability, 17/17 language impairment, 12/17 autism spectrum disorder, 9/21 epilepsy, 12/15 clumsiness, 3/21 had sensorineural hearing loss and 4/20 feeding difficulties. No microcephaly was observed at birth, but 17/20 developed microcephaly during follow-up. Regression was reported in six patients. Movement disorder was observed in 3/21 patients: hyperkinetic movements (1), truncal ataxia (1) and dystonia (2). After treatment with a high-protein diet (≥ 2 g/kg/day) and BCAA supplementation (100-250 mg/kg/day), plasma BCAA increased significantly (P < 0.001), motor functions and head circumference stabilized/improved in 13/13 and in 11/15 patients, respectively. Among cases with follow-up data, none of the three patients starting treatment before 2 years of age developed autism at follow-up. The patient with the earliest age of treatment initiation (8 months) showed normal development at 3 years of age. NBS in DBS identified BCAA levels significantly lower than those of the normal population. This work highlights the potential benefits of dietetic treatment, in particular early introduction of BCAA. Therefore, it is of utmost importance to increase awareness about this treatable disease and consider it as a candidate for early detection by NBS programmes.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Microcefalia , Recém-Nascido , Humanos , Feminino , Lactente , Masculino , Deficiência Intelectual/genética , Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/genética , Triagem Neonatal , Estudos Transversais , Fator de Maturação da Glia , Aminoácidos de Cadeia Ramificada/metabolismo , Microcefalia/genética
9.
J Craniofac Surg ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861337

RESUMO

OBJECTIVE: This study aimed to evaluate the utility and efficacy of ChatGPT in addressing questions related to thyroid surgery, taking into account accuracy, readability, and relevance. METHODS: A simulated physician-patient consultation on thyroidectomy surgery was conducted by posing 21 hypothetical questions to ChatGPT. Responses were evaluated using the DISCERN score by 3 independent ear, nose and throat specialists. Readability measures including Flesch Reading Ease), Flesch-Kincaid Grade Level, Gunning Fog Index, Simple Measure of Gobbledygook, Coleman-Liau Index, and Automated Readability Index were also applied. RESULTS: The majority of ChatGPT responses were rated fair or above using the DISCERN system, with an average score of 45.44 ± 11.24. However, the readability scores were consistently higher than the recommended grade 6 level, indicating the information may not be easily comprehensible to the general public. CONCLUSION: While ChatGPT exhibits potential in answering patient queries related to thyroid surgery, its current formulation is not yet optimally tailored for patient comprehension. Further refinements are necessary for its efficient application in the medical domain.

10.
J Hum Genet ; 68(10): 657-669, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37217689

RESUMO

Hearing loss (HL) is a common heterogeneous trait that involves variants in more than 200 genes. In this study, we utilized exome (ES) and genome sequencing (GS) to effectively identify the genetic cause of presumably non-syndromic HL in 322 families from South and West Asia and Latin America. Biallelic GJB2 variants were identified in 58 probands at the time of enrollment these probands were excluded. In addition, upon review of phenotypic findings, 38/322 probands were excluded based on syndromic findings at the time of ascertainment and no further evaluation was performed on those samples. We performed ES as a primary diagnostic tool on one or two affected individuals from 212/226 families. Via ES we detected a total of 78 variants in 30 genes and showed their co-segregation with HL in 71 affected families. Most of the variants were frameshift or missense and affected individuals were either homozygous or compound heterozygous in their respective families. We employed GS as a primary test on a subset of 14 families and a secondary tool on 22 families which were unsolved by ES. Although the cumulative detection rate of causal variants by ES and GS is 40% (89/226), GS alone has led to a molecular diagnosis in 7 of 14 families as the primary tool and 5 of 22 families as the secondary test. GS successfully identified variants present in deep intronic or complex regions not detectable by ES.


Assuntos
Surdez , Perda Auditiva , Humanos , Surdez/genética , Perda Auditiva/genética , Perda Auditiva/diagnóstico , Fenótipo , Homozigoto , Mutação , Linhagem
11.
Am J Med Genet A ; 191(4): 1044-1049, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36628575

RESUMO

Phenotypic features of KBG syndrome include craniofacial anomalies, short stature, cognitive disability and behavioral findings. The syndrome is caused by heterozygous pathogenic single nucleotide variants and indels in ANKRD11, or a heterozygous deletion of 16q24.3 that includes ANKRD11. We performed genome sequencing on a patient with clinical manifestations of KBG syndrome including distinct craniofacial features as well as a history of mild intellectual disability and attention-deficit hyperactivity disorder. This led to the identification of a 43 kb intragenic deletion of ANKRD11 affecting the first noncoding exon while leaving the coding regions intact. Review of the literature shows that this is the smallest 5' deletion affecting only the noncoding exons of ANKRD11. Real-time polymerase chain reaction demonstrated that the copy number variant was not present in either of the proband's parents, suggesting it occurred de novo. RNA expression analysis demonstrated significantly decreased transcript abundance compared to controls. This provides new evidence for haploinsufficiency as a mechanism of disease in KBG syndrome.


Assuntos
Anormalidades Múltiplas , Doenças do Desenvolvimento Ósseo , Deficiência Intelectual , Anormalidades Dentárias , Humanos , Anormalidades Múltiplas/genética , Deficiência Intelectual/genética , Doenças do Desenvolvimento Ósseo/genética , Anormalidades Dentárias/genética , Fácies , Proteínas Repressoras/genética , Deleção Cromossômica , Fatores de Transcrição/genética , Fenótipo
12.
Am J Med Genet A ; 191(7): 1911-1916, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36987712

RESUMO

Recurrent de novo missense variants in H4 histone genes have recently been associated with a novel neurodevelopmental syndrome that is characterized by intellectual disability and developmental delay as well as more variable findings that include short stature, microcephaly, and facial dysmorphisms. A 4-year-old male with autism, developmental delay, microcephaly, and a happy demeanor underwent evaluation through the Undiagnosed Disease Network. He was clinically suspected to have Angelman syndrome; however, molecular testing was negative. Genome sequencing identified the H4 histone gene variant H4C5 NM_003545.4: c.295T>C, p.Tyr99His, which parental testing confirmed to be de novo. The variant met criteria for a likely pathogenic classification and is one of the seven known disease-causing missense variants in H4C5. A comparison of our proband's findings to the initial description of the H4-associated neurodevelopmental syndrome demonstrates that his phenotype closely matches the spectrum of those reported among the 29 affected individuals. As such, this report corroborates the delineation of neurodevelopmental syndrome caused by de novo missense H4 gene variants. Moreover, it suggests that cases of clinically suspected Angelman syndrome without molecular confirmation should undergo exome or genome sequencing, as novel neurodevelopmental syndromes with phenotypes overlapping with Angelman continue to be discovered.


Assuntos
Síndrome de Angelman , Deficiência Intelectual , Microcefalia , Transtornos do Neurodesenvolvimento , Masculino , Humanos , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Microcefalia/genética , Histonas/genética , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Fenótipo , Transtornos do Neurodesenvolvimento/diagnóstico , Transtornos do Neurodesenvolvimento/genética , Mutação de Sentido Incorreto/genética
13.
Eur J Orthop Surg Traumatol ; 33(3): 629-637, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35852612

RESUMO

PURPOSE: Long bone defects due to fractures resulting from high-energy trauma, infections and tumor resections are problems that orthopedic surgeons commonly face. We investigated the effects of a titanium mesh cage on bone healing with an induced membrane technique. METHODS: Three groups, each composed of eight rabbits, were formed. Extraarticular diaphyseal bone defects were created. Femora of the first group were fixed with an empty titanium mesh cage and two K-wires. After formation of the defect, polymethylmethacrylate was inserted and fixed with a K-wire in the second group. At the third week, the cement was removed, a sterilized cancellous graft-filled titanium mesh cage was placed into the defect, and the membrane that was previously formed over the cement was placed on the cage and repaired. In the third group, sterilized cancellous grafts were filled into the titanium mesh cage, and the titanium mesh cage was fitted into the bone defect area. RESULTS: At the end of the third month, all subjects were killed. Radiological data revealed that the healing of the bone in the second and third groups was significantly better than that in the first group. There was no difference between the second and third groups. A histological evaluation of the healing status, such as fibrous tissue, cartilage tissue and mature or immature bone formation, was performed. Histological healing in the second and third groups was also significantly better than that in the first group. CONCLUSION: We concluded that the combination of membrane-induced bone healing and graft-filled titanium mesh cages expedites osteogenesis in extraarticular bone defects.


Assuntos
Fraturas Ósseas , Titânio , Coelhos , Animais , Telas Cirúrgicas , Próteses e Implantes , Fêmur/cirurgia , Fêmur/patologia , Fraturas Ósseas/cirurgia
14.
Am J Med Genet A ; 188(4): 1307-1310, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34995019

RESUMO

Auriculocondylar syndrome (ARCND) is characterized by a distinguished feature of question mark ears and a variation of other minor and major malformations. Monoallelic or biallelic PLCB4 variants have been reported in a subset of affected individuals, referred to as ARCND2. We report on a 3-year-old female with ARCND who presented at birth with question mark ears, micrognathia, and bilateral choanal stenosis that was characterized by difficulty in breathing. She was found to be heterozygous for a novel PLCB4 variant, p.Glu358Gly. Respiratory distress is rare in autosomal dominant ARCND2 and choanal stenosis has not been reported. Our study expands the clinical phenotype of ARCND by adding choanal stenosis as a finding and suggests that PLCB4 play a role in the development of choanal structures.


Assuntos
Atresia das Cóanas , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Atresia das Cóanas/diagnóstico , Atresia das Cóanas/genética , Constrição Patológica/genética , Orelha/anormalidades , Otopatias , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Mutação , Linhagem , Fosfolipase C beta/genética
15.
Proc Natl Acad Sci U S A ; 116(4): 1347-1352, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30610177

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Adaptadora GRB2/metabolismo , Perda Auditiva Neurossensorial/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Surdez/microbiologia , Drosophila/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/fisiologia , Transdução de Sinais/fisiologia
16.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30561639

RESUMO

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.


Assuntos
Cóclea/embriologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Adulto , Animais , Criança , Cóclea/metabolismo , Cóclea/fisiologia , Desenvolvimento Embrionário , Feminino , Células Ciliadas Auditivas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Organogênese , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/fisiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Transdução de Sinais/genética , Sequenciamento Completo do Genoma
17.
Am J Med Genet A ; 182(3): 548-552, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31833199

RESUMO

ZMIZ1, zinc finger MIZ-domain containing 1, has recently been described in association with syndromic intellectual disability in which the primary phenotypic features include intellectual disability/developmental delay, seizures, hearing loss, behavioral issues, failure to thrive, and various congenital malformations. Most reported cases have been found to result from de novo mutations except for one set of three siblings in which parental testing could not be performed. With informed consent from the family, we report on a father and his two sons demonstrating autosomal dominant inheritance of a novel pathogenic ZMIZ1 variant, c.1310delC (p.Pro437ArgfsX84), causing this recently described neurodevelopmental syndrome. While they all show syndromic findings along with short stature and intellectual disability, only one child had sensorineural hearing loss. Moreover, severity of intellectual disability and eyelid ptosis were variable among the affected members. Our report demonstrates that phenotypic features of ZMIZ1-related neurodevelopmental syndrome are variable even within the same family and that parental testing to identify a mildly affected parent is needed.


Assuntos
Perda Auditiva Neurossensorial/genética , Deficiência Intelectual/genética , Malformações do Sistema Nervoso/genética , Transtornos do Neurodesenvolvimento/genética , Fatores de Transcrição/genética , Idoso , Alelos , Criança , Pré-Escolar , Exoma/genética , Feminino , Predisposição Genética para Doença , Perda Auditiva Neurossensorial/complicações , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/patologia , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/patologia , Masculino , Pessoa de Meia-Idade , Mutação/genética , Malformações do Sistema Nervoso/diagnóstico , Malformações do Sistema Nervoso/patologia , Transtornos do Neurodesenvolvimento/complicações , Transtornos do Neurodesenvolvimento/diagnóstico , Transtornos do Neurodesenvolvimento/patologia , Pais , Irmãos
18.
J Inherit Metab Dis ; 43(6): 1199-1204, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32748411

RESUMO

Galactosemia is a rare, treatable hereditary disorder of carbohydrate metabolism. We investigated the etiology of decreased GALT enzyme activity in a cohort of newborns referred by the Florida Newborn Screening Program with no detectable GALT variants in diagnostic molecular tests. Six affected individuals from four families with Guatemalan heritage were included. GALT enzyme activity ranged from 20% to 34% of normal. Clinical findings were unremarkable except for speech delay in two children. Via genome sequencing followed by Sanger confirmation we showed that all affected individuals were homozygous for a deep intronic GALT variant, c.1059+390A>G, which segregated as an autosomal recessive trait in all families. The intronic variant disrupts splicing and leads to a premature termination and is associated with a single haplotype flanking GALT, suggesting a founder effect. In conclusion, we present a deep intronic GALT variant leading to a biochemical variant form of galactosemia. This variant remains undiagnosed until it is specifically targeted in molecular testing.


Assuntos
Galactosemias/diagnóstico , Homozigoto , Mutação , UTP-Hexose-1-Fosfato Uridililtransferase/genética , Pré-Escolar , Saúde da Família , Feminino , Galactosemias/sangue , Galactosemias/genética , Testes Genéticos , Humanos , Lactente , Recém-Nascido , Masculino , Triagem Neonatal , UTP-Hexose-1-Fosfato Uridililtransferase/deficiência
19.
Hum Genet ; 138(10): 1071-1075, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31175426

RESUMO

While the importance of tight junctions in hearing is well established, the role of Claudin- 9 (CLDN9), a tight junction protein, in human hearing and deafness has not been explored. Through whole-genome sequencing, we identified a one base pair deletion (c.86delT) in CLDN9 in a consanguineous family from Turkey with autosomal recessive nonsyndromic hearing loss. Three affected members of the family had sensorineural hearing loss (SNHL) ranging from moderate to profound in severity. The variant is predicted to cause a frameshift and produce a truncated protein (p.Leu29ArgfsTer4) in this single-exon gene. It is absent in public databases as well as in over 1000 Turkish individuals, and co-segregates with SNHL in the family. Our in vitro studies demonstrate that the mutant protein does not localize to cell membrane as demonstrated for the wild-type protein. Mice-lacking Cldn9 have been shown to develop SNHL. We conclude that CLDN9 is essential for proper audition in humans and its disruption leads to SNHL in humans.


Assuntos
Claudinas/genética , Surdez/diagnóstico , Surdez/genética , Genes Recessivos , Estudos de Associação Genética , Predisposição Genética para Doença , Variação Genética , Claudinas/química , Claudinas/metabolismo , Biologia Computacional/métodos , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Humanos , Mutação , Linhagem , Polimorfismo Genético , Transporte Proteico , Turquia , Sequenciamento Completo do Genoma
20.
Genet Med ; 21(10): 2239-2247, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30894701

RESUMO

PURPOSE: Proper interpretation of genomic variants is critical to successful medical decision making based on genetic testing results. A fundamental prerequisite to accurate variant interpretation is the clear understanding of the clinical validity of gene-disease relationships. The Clinical Genome Resource (ClinGen) has developed a semiquantitative framework to assign clinical validity to gene-disease relationships. METHODS: The ClinGen Hearing Loss Gene Curation Expert Panel (HL GCEP) uses this framework to perform evidence-based curations of genes present on testing panels from 17 clinical laboratories in the Genetic Testing Registry. The HL GCEP curated and reviewed 142 genes and 164 gene-disease pairs, including 105 nonsyndromic and 59 syndromic forms of hearing loss. RESULTS: The final outcome included 82 Definitive (50%), 12 Strong (7%), 25 Moderate (15%), 32 Limited (20%), 10 Disputed (6%), and 3 Refuted (2%) classifications. The summary of each curation is date stamped with the HL GCEP approval, is live, and will be kept up-to-date on the ClinGen website ( https://search.clinicalgenome.org/kb/gene-validity ). CONCLUSION: This gene curation approach serves to optimize the clinical sensitivity of genetic testing while reducing the rate of uncertain or ambiguous test results caused by the interrogation of genes with insufficient evidence of a disease link.


Assuntos
Surdez/genética , Testes Genéticos/métodos , Perda Auditiva/genética , Curadoria de Dados/métodos , Bases de Dados Genéticas , Testes Genéticos/normas , Variação Genética , Genoma Humano , Genômica/métodos , Humanos , Mutação , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA