Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Stem Cell Res ; 71: 103181, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37595341

RESUMEN

The DFNA58 locus contains a genomic duplication involving three protein-coding genes (CNRIP1, PLEK, and PPP3R1's exon 1) and other uncharacterized lncRNA genes (LOC101927723, LOC107985892 and LOC102724389). To clarify the role of these genes in hearing and precisely determine their role in hearing loss, four iPSC lines were generated from two carriers and two noncarriers of the duplication.


Asunto(s)
Pérdida Auditiva , Células Madre Pluripotentes Inducidas , Humanos , Leucocitos Mononucleares , Pérdida Auditiva/genética , Audición , Exones
2.
Eur J Hum Genet ; 30(1): 13-21, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33953343

RESUMEN

We recently described a novel missense variant [c.2090T>G:p.(Leu697Trp)] in the MYO3A gene, found in two Brazilian families with late-onset autosomal dominant nonsyndromic hearing loss (ADNSHL). Since then, with the objective of evaluating its contribution to ADNSHL in Brazil, the variant was screened in additional 101 pedigrees with probable ADNSHL without conclusive molecular diagnosis. The variant was found in three additional families, explaining 3/101 (~3%) of cases with ADNSHL in our Brazilian pedigree collection. In order to identify the origin of the variant, 21 individuals from the five families were genotyped with a high-density SNP array (~600 K SNPs- Axiom Human Origins; ThermoFisher). The identity by descent (IBD) approach revealed that many pairs of individuals from the different families have a kinship coefficient equivalent to that of second cousins, and all share a minimum haplotype of ~607 kb which includes the c.2090T>G variant suggesting it probably arose in a common ancestor. We inferred that the mutation occurred in a chromosomal segment of European ancestry and the time since the most common ancestor was estimated in 1100 years (CI = 775-1425). This variant was also reported in a Dutch family, which shares a 87,121 bp haplotype with the Brazilian samples, suggesting that Dutch colonists may have brought it to Northeastern Brazil in the 17th century. Therefore, the present study opens new avenues to investigate this variant not only in Brazilians but also in European families with ADNSHL.


Asunto(s)
Frecuencia de los Genes , Pérdida Auditiva Sensorineural/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Brasil , Efecto Fundador , Genes Dominantes , Haplotipos , Migración Humana , Humanos , Mutación Missense , Linaje , Polimorfismo de Nucleótido Simple
3.
Hum Mol Genet ; 29(9): 1520-1536, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32337552

RESUMEN

Here we define a ~200 Kb genomic duplication in 2p14 as the genetic signature that segregates with postlingual progressive sensorineural autosomal dominant hearing loss (HL) in 20 affected individuals from the DFNA58 family, first reported in 2009. The duplication includes two entire genes, PLEK and CNRIP1, and the first exon of PPP3R1 (protein coding), in addition to four uncharacterized long non-coding (lnc) RNA genes and part of a novel protein-coding gene. Quantitative analysis of mRNA expression in blood samples revealed selective overexpression of CNRIP1 and of two lncRNA genes (LOC107985892 and LOC102724389) in all affected members tested, but not in unaffected ones. Qualitative analysis of mRNA expression identified also fusion transcripts involving parts of PPP3R1, CNRIP1 and an intergenic region between PLEK and CNRIP1, in the blood of all carriers of the duplication, but were heterogeneous in nature. By in situ hybridization and immunofluorescence, we showed that Cnrip1, Plek and Ppp3r1 genes are all expressed in the adult mouse cochlea including the spiral ganglion neurons, suggesting changes in expression levels of these genes in the hearing organ could underlie the DFNA58 form of deafness. Our study highlights the value of studying rare genomic events leading to HL, such as copy number variations. Further studies will be required to determine which of these genes, either coding proteins or non-coding RNAs, is or are responsible for DFNA58 HL.


Asunto(s)
Proteínas Sanguíneas/genética , Calcineurina/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Fosfoproteínas/genética , Adolescente , Adulto , Animales , Calcineurina/sangre , Niño , Duplicación Cromosómica/genética , Cromosomas Humanos Par 2/genética , Variaciones en el Número de Copia de ADN/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Genoma Humano/genética , Pérdida Auditiva Sensorineural/sangre , Pérdida Auditiva Sensorineural/patología , Heterocigoto , Humanos , Masculino , Proteínas de la Membrana/sangre , Ratones , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Fosfoproteínas/sangre , ARN Mensajero/sangre , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/patología , Adulto Joven
4.
Int J Mol Sci ; 19(9)2018 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-30150563

RESUMEN

GJB2 mutations are the leading cause of non-syndromic inherited hearing loss. GJB2 encodes connexin-26 (CX26), which is a connexin (CX) family protein expressed in cochlea, skin, liver, and brain, displaying short cytoplasmic N-termini and C-termini. We searched for CX26 C-terminus binding partners by affinity capture and identified 12 unique proteins associated with cell junctions or cytoskeleton (CGN, DAAM1, FLNB, GAPDH, HOMER2, MAP7, MAPRE2 (EB2), JUP, PTK2B, RAI14, TJP1, and VCL) by using mass spectrometry. We show that, similar to other CX family members, CX26 co-fractionates with TJP1, VCL, and EB2 (EB1 paralogue) as well as the membrane-associated protein ASS1. The adaptor protein CGN (cingulin) co-immuno-precipitates with CX26, ASS1, and TJP1. In addition, CGN co-immunoprecipitation with CX30, CX31, and CX43 indicates that CX association is independent on the CX C-terminus length or sequence. CX26, CGN, FLNB, and DAMM1 were shown to distribute to the organ of Corti and hepatocyte plasma membrane. In the mouse liver, CX26 and TJP1 co-localized at the plasma membrane. In conclusion, CX26 associates with components of other membrane junctions that integrate with the cytoskeleton.


Asunto(s)
Conexina 26/metabolismo , Conexinas/metabolismo , Uniones Intercelulares/metabolismo , Secuencia de Aminoácidos , Animales , Argininosuccinato Sintasa/genética , Argininosuccinato Sintasa/metabolismo , Conexina 26/genética , Conexinas/genética , Citoesqueleto/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Órgano Espiral/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas , Homología de Secuencia de Aminoácido , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
5.
Sci Rep ; 8(1): 8706, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880844

RESUMEN

Whole-exome sequencing of samples from affected members of two unrelated families with late-onset non-syndromic hearing loss revealed a novel mutation (c.2090 T > G; NM_017433) in MYO3A. The mutation was confirmed in 36 affected individuals, showing autosomal dominant inheritance. The mutation alters a single residue (L697W or p.Leu697Trp) in the motor domain of the stereocilia protein MYO3A, leading to a reduction in ATPase activity, motility, and an increase in actin affinity. MYO3A-L697W showed reduced filopodial actin protrusion initiation in COS7 cells, and a predominant tipward accumulation at filopodia and stereocilia when coexpressed with wild-type MYO3A and espin-1, an actin-regulatory MYO3A cargo. The combined higher actin affinity and duty ratio of the mutant myosin cause increased retention time at stereocilia tips, resulting in the displacement of the wild-type MYO3A protein, which may impact cargo transport, stereocilia length, and mechanotransduction. The dominant negative effect of the altered myosin function explains the dominant inheritance of deafness.


Asunto(s)
Genes Dominantes , Enfermedades Genéticas Congénitas/genética , Pérdida Auditiva/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Actinas/genética , Actinas/metabolismo , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Animales , Brasil , Células COS , Movimiento Celular/genética , Niño , Chlorocebus aethiops , Femenino , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Humanos , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Seudópodos/genética , Seudópodos/metabolismo , Seudópodos/patología , Estereocilios/genética , Estereocilios/metabolismo , Estereocilios/patología
6.
PLoS One ; 13(4): e0196360, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29689090

RESUMEN

The analysis of genomic data (~400,000 autosomal SNPs) enabled the reliable estimation of inbreeding levels in a sample of 541 individuals sampled from a highly admixed Brazilian population isolate (an African-derived quilombo in the State of São Paulo). To achieve this, different methods were applied to the joint information of two sets of markers (one complete and another excluding loci in patent linkage disequilibrium). This strategy allowed the detection and exclusion of markers that biased the estimation of the average population inbreeding coefficient (Wright's fixation index FIS), which value was eventually estimated as around 1% using any of the methods we applied. Quilombo demographic inferences were made by analyzing the structure of runs of homozygosity (ROH), which were adapted to cope with a highly admixed population with a complex foundation history. Our results suggest that the amount of ROH <2Mb of admixed populations should be somehow proportional to the genetic contribution from each parental population.


Asunto(s)
Consanguinidad , Genética de Población/métodos , Genética de Población/estadística & datos numéricos , Población Negra/estadística & datos numéricos , Brasil/epidemiología , Femenino , Marcadores Genéticos , Genómica/métodos , Homocigoto , Humanos , Desequilibrio de Ligamiento , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Estadística como Asunto/métodos
7.
Eur J Hum Genet ; 25(12): 1335-1344, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29255178

RESUMEN

Kabuki syndrome is a monogenic disorder caused by loss of function variants in either of two genes encoding histone-modifying enzymes. We performed targeted sequencing in a cohort of 27 probands with a clinical diagnosis of Kabuki syndrome. Of these, 12 had causative variants in the two known Kabuki syndrome genes. In 2, we identified presumptive loss of function de novo variants in KMT2A (missense and splice site variants), a gene that encodes another histone modifying enzyme previously exclusively associated with Wiedermann-Steiner syndrome. Although Kabuki syndrome is a disorder of histone modification, we also find alterations in DNA methylation among individuals with a Kabuki syndrome diagnosis relative to matched normal controls, regardless of whether they carry a variant in KMT2A or KMT2D or not. Furthermore, we observed characteristic global abnormalities of DNA methylation that distinguished patients with a loss of function variant in KMT2D or missense or splice site variants in either KMT2D or KMT2A from normal controls. Our results provide new insights into the relationship of genotype to epigenotype and phenotype and indicate cross-talk between histone and DNA methylation machineries exposed by inborn errors of the epigenetic apparatus.


Asunto(s)
Anomalías Múltiples/genética , Metilación de ADN , Cara/anomalías , Enfermedades Hematológicas/genética , Fenotipo , Enfermedades Vestibulares/genética , Anomalías Múltiples/diagnóstico , Estudios de Casos y Controles , Niño , Femenino , Enfermedades Hematológicas/diagnóstico , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Mutación con Pérdida de Función , Masculino , Proteína de la Leucemia Mieloide-Linfoide/genética , Enfermedades Vestibulares/diagnóstico
8.
J Hum Genet ; 62(12): 1073-1078, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28855715

RESUMEN

We have recently described a family with a condition (Santos syndrome (SS; MIM 613005)) characterized by fibular agenesis/hypoplasia, hypoplastic femora and grossly malformed/deformed clubfeet with severe oligodactyly, ungual hypoplasia/anonychia, sometimes associated with mild brachydactyly and occasional pre-axial polydactyly. Autosomal dominant inheritance with incomplete penetrance was suggested, but autosomal recessive inheritance could not be ruled out, due to the high frequency of consanguineous matings in the region where the family lived. This report deals with linkage studies and exome sequencing, disclosing a novel variant in WNT7A, c.934G>A (p.Gly312Ser), as the cause of this syndrome. This variant was present in homozygous state in five individuals typically affected by the SS syndrome, and in heterozygous state in the son of one affected homozygous individual. The heterozygous boy presented only unilateral complex polysyndactyly and we hypothesize that he either presents a distinct defect or that his phenotype results from a rare, mild clinical manifestation of the variant in heterozygous state. Variants in WNT7A are known to cause at least two other limb defect disorders, the syndromes of Fuhrmann and Al-Awadi/Raas-Rothschild. Despite their variable degree of expressivity and overlap, the three related conditions can be differentiated phenotypically in most instances.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Pie Equinovaro/genética , Peroné/anomalías , Dedos/anomalías , Marcadores Genéticos/genética , Deformidades Congénitas de las Extremidades/genética , Uñas Malformadas/genética , Polidactilia/genética , Proteínas Wnt/genética , Secuencia de Aminoácidos , Consanguinidad , Femenino , Ligamiento Genético , Homocigoto , Humanos , Masculino , Repeticiones de Microsatélite/genética , Mutación , Linaje , Fenotipo , Alineación de Secuencia
9.
Hum Immunol ; 77(3): 307-312, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26582005

RESUMEN

Methods to impute HLA alleles based on dense single nucleotide polymorphism (SNP) data provide a valuable resource to association studies and evolutionary investigation of the MHC region. The availability of appropriate training sets is critical to the accuracy of HLA imputation, and the inclusion of samples with various ancestries is an important pre-requisite in studies of admixed populations. We assess the accuracy of HLA imputation using 1000 Genomes Project data as a training set, applying it to a highly admixed Brazilian population, the Quilombos from the state of São Paulo. To assess accuracy, we compared imputed and experimentally determined genotypes for 146 samples at 4 HLA classical loci. We found imputation accuracies of 82.9%, 81.8%, 94.8% and 86.6% for HLA-A, -B, -C and -DRB1 respectively (two-field resolution). Accuracies were improved when we included a subset of Quilombo individuals in the training set. We conclude that the 1000 Genomes data is a valuable resource for construction of training sets due to the diversity of ancestries and the potential for a large overlap of SNPs with the target population. We also show that tailoring training sets to features of the target population substantially enhances imputation accuracy.


Asunto(s)
Alelos , Biología Computacional/métodos , Genética de Población , Antígenos HLA/genética , Polimorfismo de Nucleótido Simple , Programas Informáticos , Brasil , Bases de Datos Genéticas , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Reproducibilidad de los Resultados , Navegador Web
10.
Int J Audiol ; 54(9): 593-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25926005

RESUMEN

OBJECTIVE: To identify novel genetic causes of syndromic hearing loss in Brazil. DESIGN: To map a candidate chromosomal region through linkage studies in an extensive Brazilian family and identify novel pathogenic variants using sequencing and array-CGH. STUDY SAMPLE: Brazilian pedigree with individuals affected by BO syndrome characterized by deafness and malformations of outer, middle and inner ear, auricular and cervical fistulae, but no renal abnormalities. RESULTS: Whole genome microarray-SNP scanning on samples of 11 affected individuals detected a multipoint Lod score of 2.6 in the EYA1 gene region (chromosome 8). Sequencing of EYA1 in affected patients did not reveal pathogenic mutations. However, oligonucleotide-array-CGH detected a duplication of 71.8Kb involving exons 4 to 10 of EYA1 (heterozygous state). Real-time-PCR confirmed the duplication in fourteen of fifteen affected individuals and absence in 13 unaffected individuals. The exception involved a consanguineous parentage and was assumed to involve a different genetic mechanism. CONCLUSIONS: Our findings implicate this EYA1 partial duplication segregating with BO phenotype in a Brazilian pedigree and is the first description of a large duplication leading to the BOR/BO syndrome.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Duplicación de Gen , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Linaje , Proteínas Tirosina Fosfatasas/genética , Síndrome Branquio Oto Renal/complicaciones , Brasil , Consanguinidad , Oído/anomalías , Exones , Femenino , Perdida Auditiva Conductiva-Sensorineural Mixta/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Escala de Lod , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Hum Genome Var ; 2: 15038, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27081546

RESUMEN

Here we describe a novel missense variant in the KCNQ4 gene and a private duplication at 7q31.1 partially involving two genes (IMMP2L and DOCK4). Both mutations segregated with nonsyndromic hearing loss in a family with three affected individuals. Initially, we identified the duplication in a screening of 132 unrelated cases of hearing loss with a multiplex ligation-dependent probe amplification panel of genes that are candidates to have a role in hearing, including IMMP2L. Mapping of the duplication by array-CGH revealed that the duplication also encompassed the 3'-end of DOCK4. Subsequently, whole-exome sequencing identified the breakpoint of the rearrangement, thereby confirming the existence of a fusion IMMP2L-DOCK4 gene. Transcription products of the fusion gene were identified, indicating that they escaped nonsense-mediated messenger RNA decay. A missense substitution (c.701A>T) in KCNQ4 (a gene at the DFNA2A locus) was also identified by whole-exome sequencing. Because the substitution is predicted to be probably damaging and KCNQ4 has been implicated in hearing loss, this mutation might explain the deafness in the affected individuals, although a hypothetical effect of the product of the fusion gene on hearing cannot be completely ruled out.

12.
Genet Mol Biol ; 37(4): 616-21, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25505834

RESUMEN

We studied a family presenting 10 individuals affected by autosomal dominant deafness in all frequencies and three individuals affected by high frequency hearing loss. Genomic scanning using the 50k Affymetrix microarray technology yielded a Lod Score of 2.1 in chromosome 14 and a Lod Score of 1.9 in chromosome 22. Mapping refinement using microsatellites placed the chromosome 14 candidate region between markers D14S288 and D14S276 (8.85 cM) and the chromosome 22 near marker D22S283. Exome sequencing identified two candidate variants to explain hearing loss in chromosome 14 [PTGDR - c.G894A:p.R298R and PTGER2 - c.T247G:p.C83G], and one in chromosome 22 [MYH9, c.G2114A:p.R705H]. Pedigree segregation analysis allowed exclusion of the PTGDR and PTGER2 variants as the cause of deafness. However, the MYH9 variant segregated with the phenotype in all affected members, except the three individuals with different phenotype. This gene has been previously described as mutated in autosomal dominant hereditary hearing loss and corresponds to DFNA17. The mutation identified in our study is the same described in the prior report. Thus, although linkage studies suggested a candidate gene in chromosome 14, we concluded that the mutation in chromosome 22 better explains the hearing loss phenotype in the Brazilian family.

13.
Int J Mol Epidemiol Genet ; 5(1): 11-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24596593

RESUMEN

The overall aim of this study was to estimate the contribution of genetic factors to the etiology of hearing loss (HL) in two counties in the Brazilian northeastern region. A cross-sectional study, based on the key informant approach (KI) was conducted in Queimadas and Gado Bravo counties (Paraíba, Northeast Brazil). The sample consisted of 182 patients with HL. Genetic screening of the most frequent mutations associated with HL was performed for all samples. DFNB1 mutations were the most frequently found in both counties. The c.35delG mutation was detected in homozygosis in seven non-syndromic probands in Queimadas (7/76, 9.2%) and only a single homozygote with this mutation was found in Gado Bravo (1/44, 2.3%). We also detected the del(GJB6-D13S1854) mutation in non-syndromic probands from Gado Bravo (2/44, 4.5%). The c.189C>A (p.TyrY63*) mutation in the CLRN1 gene was detected in homozygosis in 21/23 Usher syndrome patients from Gado Bravo and it was not found in Queimadas. Cases with probable genetic etiology contributed approximately to half of HL probands in each county (54.6% in Gado Bravo and 45.7% in Queimadas). We confirm the importance of DFNB1 locus to non-syndromic HL but we show that the frequency of mutations in the northeastern region differs somewhat from those reported in southeastern Brazil and other populations. In addition, the extremely high frequency of individuals with Usher syndrome with c.189C>A variation in CLRN1 indicates the need for a specific screening of this mutation.

14.
Eur J Med Genet ; 57(4): 125-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24556497

RESUMEN

In 20% of cases, hereditary non-syndromic hearing loss has an autosomal dominant inheritance (ADNSHL). To date, more than 50 loci for ADNSHL have been mapped to different chromosomal regions. In order to verify whether genomic alterations contribute to the hearing loss etiology and to search for novel deafness candidate loci, we investigated probands from families with ADNSHL by oligonucleotide array-CGH. A deletion in the 5q32 region encompassing only one gene, POU4F3, which corresponds to DFNA15, was detected in one family. POU4F3 protein has an important role in the maturation, differentiation and survival of cochlear hair cells. Defects in these cells may therefore explain sensorineural hearing loss. Mutations in this gene have already been associated with autosomal dominant hearing loss but this is the first description of a germline POUF4F3 deletion associated with hearing impairment.


Asunto(s)
Eliminación de Gen , Genes Dominantes , Pérdida Auditiva Sensorineural/genética , Proteínas de Homeodominio/genética , Factor de Transcripción Brn-3C/genética , Adulto , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Hibridación Genómica Comparativa/métodos , Salud de la Familia , Femenino , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Linaje
15.
Mol Biol Rep ; 41(5): 3113-25, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24477587

RESUMEN

The objective of the present study was to review previous investigations on the association of haplotypes in the G-protein ß3 subunit (GNB3) gene with representative cardiovascular risk factors/phenotypes: hypertension, overweight, and variation in the systolic and diastolic blood pressures (SBP and DBP, respectively) and as well as body mass index (BMI). A comprehensive literature search was undertaken in Pubmed, Web of Science, EMBASE, Biological Abstracts, LILACS and Google Scholar to identify potentially relevant articles published up to April 2011. Six genetic association studies encompassing 16,068 participants were identified. Individual participant data were obtained for all studies. The three most investigated GNB3 polymorphisms (G-350A, C825T and C1429T) were considered. Expectation-maximization and generalized linear models were employed to estimate haplotypic effects from data with uncertain phase while adjusting for covariates. Study-specific results were combined through a random-effects multivariate meta-analysis. After carefully adjustments for relevant confounding factors, our analysis failed to support a role for GNB3 haplotypes in any of the investigated phenotypes. Sensitivity analyses excluding studies violating Hardy-Weinberg expectations, considering gender-specific effects or more extreme phenotypes (e.g. obesity only) as well as a fixed-effects "pooled" analysis also did not disclose a significant influence of GNB3 haplotypes on cardiovascular phenotypes. We conclude that the previous cumulative evidence does not support the proposal that haplotypes formed by common GNB3 polymorphisms might contribute either to the development of hypertension and obesity, or to the variation in the SBP, DBP and BMI.


Asunto(s)
Presión Sanguínea/genética , Estudios de Asociación Genética , Haplotipos , Proteínas de Unión al GTP Heterotriméricas/genética , Hipertensión/genética , Sobrepeso/genética , Regiones no Traducidas 3' , Alelos , Exones , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Oportunidad Relativa , Evaluación del Resultado de la Atención al Paciente , Regiones Promotoras Genéticas
16.
Gene ; 511(2): 280-4, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22995349

RESUMEN

We ascertained a Brazilian family with nine individuals affected by autosomal dominant nonsyndromic sensorineural hearing loss. The bilateral hearing loss affected mainly mid-high frequencies, was apparently stable with an early onset. Microsatellites close to the DFNA8/DFNA12 locus, which harbors the TECTA gene, showed significant multipoint lod scores (3.2) close to marker D11S4107. Sequencing of the exons and exon-intron boundaries of the TECTA gene in one affected subject revealed the deletion c.5383+5delGTGA in the 5' end of intron 16, that includes the last two bases of the donor splice site consensus sequence. This mutation segregates with deafness within the family. To date, 33 different TECTA mutations associated with autossomal dominant hearing loss have been described. Among them is the mutation reported herein, first described by Hildebrand et al. (2011) in a UK family. The audioprofiles from the UK and Brazilian families were similar. In order to investigate the transcripts produced by the mutated allele, we performed cDNA analysis of a lymphoblastoid cell line from an affected heterozygote with the c.5383+5delGTGA and a noncarrier from the same family. The analysis allowed us to identify an aberrant transcript with skipping of exon 16, without affecting the reading frame. One of the dominant TECTA mutations already described, a synonymous substitution in exon 16 (c.5331G

Asunto(s)
Sordera/genética , Proteínas de la Matriz Extracelular/genética , Genes Dominantes , ARN Mensajero/genética , Secuencia de Bases , Brasil , Cartilla de ADN , Femenino , Proteínas Ligadas a GPI/genética , Ligamiento Genético , Humanos , Masculino , Linaje
17.
BMC Res Notes ; 4: 172, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21645382

RESUMEN

BACKGROUND: The generalized odds ratio (GOR) was recently suggested as a genetic model-free measure for association studies. However, its properties were not extensively investigated. We used Monte Carlo simulations to investigate type-I error rates, power and bias in both effect size and between-study variance estimates of meta-analyses using the GOR as a summary effect, and compared these results to those obtained by usual approaches of model specification. We further applied the GOR in a real meta-analysis of three genome-wide association studies in Alzheimer's disease. FINDINGS: For bi-allelic polymorphisms, the GOR performs virtually identical to a standard multiplicative model of analysis (e.g. per-allele odds ratio) for variants acting multiplicatively, but augments slightly the power to detect variants with a dominant mode of action, while reducing the probability to detect recessive variants. Although there were differences among the GOR and usual approaches in terms of bias and type-I error rates, both simulation- and real data-based results provided little indication that these differences will be substantial in practice for meta-analyses involving bi-allelic polymorphisms. However, the use of the GOR may be slightly more powerful for the synthesis of data from tri-allelic variants, particularly when susceptibility alleles are less common in the populations (≤10%). This gain in power may depend on knowledge of the direction of the effects. CONCLUSIONS: For the synthesis of data from bi-allelic variants, the GOR may be regarded as a multiplicative-like model of analysis. The use of the GOR may be slightly more powerful in the tri-allelic case, particularly when susceptibility alleles are less common in the populations.

18.
Obesity (Silver Spring) ; 19(6): 1244-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21233811

RESUMEN

We investigated whether variants in major candidate genes for food intake and body weight regulation contribute to obesity-related traits under a multilocus perspective. We studied 375 Brazilian subjects from partially isolated African-derived populations (quilombos). Seven variants displaying conflicting results in previous reports and supposedly implicated in the susceptibility of obesity-related phenotypes were investigated: ß2-adrenergic receptor (ADRB2) (Arg16Gly), insulin induced gene 2 (INSIG2) (rs7566605), leptin (LEP) (A19G), LEP receptor (LEPR) (Gln223Arg), perilipin (PLIN) (6209T > C), peroxisome proliferator-activated receptor-γ (PPARG) (Pro12Ala), and resistin (RETN) (-420 C > G). Regression models as well as generalized multifactor dimensionality reduction (GMDR) were employed to test the contribution of individual effects and higher-order interactions to BMI and waist-hip ratio (WHR) variation and risk of overweight/obesity. The best multilocus association signal identified in the quilombos was further examined in an independent sample of 334 Brazilian subjects of European ancestry. In quilombos, only the PPARG polymorphism displayed significant individual effects (WHR variation, P = 0.028). No association was observed either with the risk of overweight/obesity (BMI ≥ 25 kg/m2), risk of obesity alone (BMI ≥ 30 kg/m2) or BMI variation. However, GMDR analyses revealed an interaction between the LEPR and ADRB2 polymorphisms (P = 0.009) as well as a third-order effect involving the latter two variants plus INSIG2 (P = 0.034) with overweight/obesity. Assessment of the LEPR-ADRB2 interaction in the second sample indicated a marginally significant association (P = 0.0724), which was further verified to be limited to men (P = 0.0118). Together, our findings suggest evidence for a two-locus interaction between the LEPR Gln223Arg and ADRB2 Arg16Gly variants in the risk of overweight/obesity, and highlight further the importance of multilocus effects in the genetic component of obesity.


Asunto(s)
Obesidad/genética , Obesidad/fisiopatología , PPAR gamma/genética , Polimorfismo de Nucleótido Simple , Receptores Adrenérgicos beta 2/genética , Receptores de Leptina/genética , Adulto , Sustitución de Aminoácidos , Pesos y Medidas Corporales , Brasil , Ingestión de Energía , Femenino , Estudios de Asociación Genética , Humanos , Indígenas Sudamericanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Reducción de Dimensionalidad Multifactorial , Tipificación de Secuencias Multilocus , Obesidad/etnología , Sobrepeso/etnología , Sobrepeso/genética , Sobrepeso/fisiopatología , Análisis de Regresión , Caracteres Sexuales , Población Blanca
19.
Obesity (Silver Spring) ; 19(7): 1523-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21233812

RESUMEN

The objective of the present study was to validate a recently reported synergistic effect between variants located in the leptin receptor (LEPR) gene and in the ß-2 adrenergic receptor (ADRB2) gene on the risk of overweight/obesity. We studied a middle-aged/elderly sample of 4,193 nondiabetic Japanese subjects stratified according gender (1,911 women and 2,282 men). The LEPR Gln223Arg (rs1137101) variant as well as both ADRB2 Arg16Gly (rs1042713) and Gln27Glu (rs1042714) polymorphisms were analyzed. The primary outcome was the risk of overweight/obesity defined as BMI ≥25 kg/m(2), whereas secondary outcomes included the risk of a BMI ≥27 kg/m(2) and BMI as a continuous variable. None of the studied polymorphisms showed statistically significant individual effects, regardless of the group or phenotype studied. Haplotype analysis also did not disclose any associations of ADRB2 polymorphisms with BMI. However, dimensionality reduction-based models confirmed significant interactions among the investigated variants for BMI as a continuous variable as well as for the risk of obesity defined as BMI ≥27 kg/m(2). All disclosed interactions were found in men only. Our results provide external validation for a male specific ADRB2-LEPR interaction effect on the risk of overweight/obesity, but indicate that effect sizes associated with these interactions may be smaller in the population studied.


Asunto(s)
Predisposición Genética a la Enfermedad , Obesidad/genética , Polimorfismo Genético , Receptores Adrenérgicos beta 2/genética , Receptores de Leptina/genética , Anciano , Sustitución de Aminoácidos , Índice de Masa Corporal , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Humanos , Japón , Masculino , Persona de Mediana Edad , Reducción de Dimensionalidad Multifactorial , Sobrepeso/genética , Reproducibilidad de los Resultados , Caracteres Sexuales
20.
J Transl Med ; 8: 119, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21087511

RESUMEN

BACKGROUND: Culturing otospheres from dissociated organ of Corti is an appropriate starting point aiming at the development of cell therapy for hair cell loss. Although guinea pigs have been widely used as an excellent experimental model for studying the biology of the inner ear, the mouse cochlea has been more suitable for yielding otospheres in vitro. The aim of this study was to compare conditions and outcomes of otosphere suspension cultures from dissociated organ of Corti of either mouse or guinea pig at postnatal day three (P3), and to evaluate the guinea pig as a potential cochlea donor for preclinical cell therapy. METHODS: Organs of Corti were surgically isolated from P3 guinea pig or mouse cochlea, dissociated and cultivated under non-adherent conditions. Cultures were maintained in serum-free DMEM:F12 medium, supplemented with epidermal growth factor (EGF) plus either basic fibroblast growth factor (bFGF) or transforming growth factor alpha (TGFα). Immunofluorescence assays were conducted for phenotype characterization. RESULTS: The TGFα group presented a number of spheres significantly higher than the bFGF group. Although mouse cultures yielded more cells per sphere than guinea pig cultures, sox2 and nestin distributed similarly in otosphere cells from both organisms. We present evidence that otospheres retain properties of inner ear progenitor cells such as self-renewal, proliferation, and differentiation into hair cells or supporting cells. CONCLUSIONS: Dissociated guinea pig cochlea produced otospheres in vitro, expressing sox2 and nestin similarly to mouse otospheres. Our data is supporting evidence for the presence of inner ear progenitor cells in the postnatal guinea pig. However, there is limited viability for these cells in neonatal guinea pig cochlea when compared to the differentiation potential observed for the mouse organ of Corti at the same developmental stage.


Asunto(s)
Órgano Espiral/citología , Células Madre/citología , Animales , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Cobayas , Humanos , Ratones , Ratones Endogámicos C57BL , Órgano Espiral/efectos de los fármacos , Fenotipo , Células Madre/efectos de los fármacos , Factor de Crecimiento Transformador alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...