Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hum Mol Genet ; 32(4): 533-542, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36048845

RESUMO

Human spermatogenesis requires an orchestrated expression of numerous genes in various germ cell subtypes. Therefore, the genetic landscape of male infertility is highly complex. Known genetic factors alone account for at least 15% of male infertility. However, ~40% of infertile men remain undiagnosed and are classified as idiopathic infertile men. We performed exome sequencing in 47 idiopathic infertile men (discovery cohort), followed by replication study (40 variants in 33 genes) in 844 infertile men and 709 controls using Sequenom MassARRAY® based genotyping. We report 17 variants in twelve genes that comprise both previously reported (DNAH8, DNAH17, FISP2 and SPEF2) and novel candidate genes (BRDT, CETN1, CATSPERD, GMCL1, SPATA6, TSSK4, TSKS and ZNF318) for male infertility. The latter have a strong biological nexus to human spermatogenesis and their respective mouse knockouts are concordant with human phenotypes. One candidate gene CETN1, identified in this study, was sequenced in another independent cohort of 840 infertile and 689 fertile men. Further, CETN1 variants were functionally characterized using biophysical and cell biology approaches. We demonstrate that CETN1 variant- p.Met72Thr leads to multipolar cells, fragmented nuclei during mitosis leading to cell death and show significantly perturbed ciliary disassembly dynamics. Whereas CETN1-5' UTR variant; rs367716858 leads to loss of a methylation site and increased reporter gene expression in vitro. We report a total of eight novel candidate genes identified by exome sequencing, which may have diagnostic relevance and can contribute to improved diagnostic workup and clinical management of male infertility.


Assuntos
Proteínas de Ligação ao Cálcio , Infertilidade Masculina , Animais , Humanos , Masculino , Camundongos , Divisão Celular , Proteínas do Citoesqueleto/genética , Sequenciamento do Exoma , Fertilidade/genética , Infertilidade Masculina/genética , Espermatogênese/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/genética
2.
Bipolar Disord ; 22(1): 70-78, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31400178

RESUMO

OBJECTIVES: Bipolar disorder (BD) is a neuropsychiatric disorder with a complex pattern of inheritance. Although many genetic studies have been conducted on BD, its genetic correlates remain uncertain. This study was aimed at  identifying the genetic underpinnings of the disorder in an Indian family, which has been under comprehensive clinical evaluation and follow-up for over 12 years. METHODS: We analysed a four-generation family with several of its members diagnosed for BD employing a combination of genetic linkage and exome analysis. RESULTS: We obtained suggestive LOD score for a chromosome 1 and a chromosome 6 marker (D1S410; LOD = 3.01, Ó¨ = 0; and D6S289; LOD = 1.58, Ó¨ = 0). Manual haplotyping of the regions encompassing these two markers helped delimit a critical genomic interval of 32.44 Mb (D1S2700-D1S435; chromosome 1p31.1-13.2) and another of 10.34 Mb (D6S470-D6S422; chromosome 6p22.3-22.2). We examined the exomic sequences corresponding to these two intervals and found rare variants, NM_181712.4: c.2461G>T (p.Asp821Tyr) in KANK4 at 1p31.1-13.2; and NM_006366:c.-93G>A, in the 5' UTR of CAP2 at 6p22.3-22.2. CONCLUSIONS: Our studysuggests involvement of KANK4 or CAP2 or both in BD in this family. Further analysis of these two genes in BD patients and functional evaluation of the allelic variants identified are suggested.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Transtorno Bipolar , Proteínas do Citoesqueleto/genética , Proteínas de Membrana/genética , Adulto , Transtorno Bipolar/diagnóstico , Transtorno Bipolar/genética , Cromossomos Humanos Par 1 , Cromossomos Humanos Par 6 , Saúde da Família , Feminino , Ligação Genética , Humanos , Masculino , Linhagem , Sequenciamento do Exoma/métodos
3.
Sex Dev ; 13(4): 178-186, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32008008

RESUMO

Steroidogenic factor 1 (NR5A1/SF1) is a key transcription factor that is known to regulate the development of adrenal glands and gonads and is also involved in steroidogenesis. Several pathogenic NR5A1 variants have been reported to cause 46,XY disorders of sex development (DSD), with varying clinical phenotypes ranging from hypospadias to complete gonadal dysgenesis. Most often, the primary cause of DSD is due to variants in gene(s) related to gonadal development or the steroidogenic pathway. In the present study, we have analyzed 64 cases of 46,XY DSD for pathogenic NR5A1 variants. We report a total of 3 pathogenic variants of which 2 were novel (p.Gly22Ser and p.Ser143Asn) and 1 was already known (p.Ser32Asn). Functional studies have revealed that the 2 mutations p.Gly22Ser and p.Ser32Asn could significantly affect DNA binding and transactivation abilities. Further, these mutant proteins showed nuclear localization with aggregate formation. The third mutation, p.Ser143Asn, showed unspeckled nuclear localization and normal DNA binding, but the ability of transcriptional activation was significantly reduced. In conclusion, we recommend screening for NR5A1 pathogenic variants in individuals with features of 46,XY DSD for better diagnosis and management.


Assuntos
Transtorno 46,XY do Desenvolvimento Sexual/genética , Transtorno 46,XY do Desenvolvimento Sexual/patologia , Mutação/genética , Fator Esteroidogênico 1/genética , Adolescente , Núcleo Celular/metabolismo , Criança , Simulação por Computador , Feminino , Células HEK293 , Humanos , Masculino , Modelos Moleculares , Linhagem , Fenótipo , Domínios Proteicos , Transporte Proteico , Fator Esteroidogênico 1/química , Transcrição Gênica
4.
J Genet ; 97(2): 391-398, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29932058

RESUMO

Hot water epilepsy (HWE) is a rare form of sensory epilepsy where seizures are precipitated by a stimulus of contact with hot water. While earlier studies have suggested causal role of genes for HWE, specific underpinnings are beginning to be explored only recently. We carried out a whole genome-based linkage analysis in a family where most of its members affected by HWE and found evidence of a previously unknown locus at chromosome 9p24.3-p23. Parametric two-point analysis suggested linkage with the greatest LOD score of 3.42 for the marker D9S286 at 9p24.1 at recombination fraction (θ) = 0, 90% penetrance value and 1% phenocopy rate. The highest multipoint LODscore of 3.42 was obtained for same marker at 9p24. The critical genetic interval of about 10 Mb of DNA was defined by the markers D9S917 and D9S168 corresponding to the centromere-distal and centromere-proximal recombination boundaries, respectively. This observation along with our previous findings of hot water genetic loci at 10q21.3-q22.3 (OMIM: 613339) and 4q24-q28 (OMIM: 613340), indicates unanticipated genetic heterogeneity for the disorder in families from a relatively small geographic region in the southern parts of India.


Assuntos
Cromossomos Humanos Par 9/genética , Epilepsia Parcial Sensorial/genética , Loci Gênicos/genética , Temperatura Alta/efeitos adversos , Água/efeitos adversos , Mapeamento Cromossômico , Epilepsia Parcial Sensorial/etiologia , Saúde da Família , Feminino , Predisposição Genética para Doença/genética , Humanos , Índia , Escore Lod , Masculino , Linhagem
5.
Indian J Med Res ; 145(4): 492-497, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28862181

RESUMO

BACKGROUND & OBJECTIVES: Hearing impairment is a common and heterogeneous sensory disorder in humans. Among about 90 genes, which are known to be associated with hearing impairment, mutations in the GJB2 (gap junction protein beta 2) gene are the most prevalent in individuals with hereditary hearing loss. Contribution of the other deafness-causing genes is relatively poorly understood. Here, we present our findings on two families with transmembrane channel like 1 (TMC1) gene variants of the 47 families with nonsyndromic hearing loss (NSHL) studied. METHODS: Forty seven families including 26 consanguineous families with at least two hearing impaired children and one normal hearing child and 21 non-consanguineous families having at least three hearing impaired children and one normal hearing child were enrolled for this study. Genetic linkage studies were carried out in 41 families that were GJB2 (Connexin 26) negative. Seven polymorphic short tandem repeat markers at the DFNB7/11 locus were studied employing fluorescently labelled markers. RESULTS: A novel homozygous missense mutation c.1283C>A (p.Ala428Asp) was identified co-segregating with hearing loss. This change results in substitution of a highly conserved polar alanine to a charged aspartic acid and is predicted to be deleterious. In addition, a previously reported nonsense mutation, p.R34X in TMC1, was found. INTERPRETATION & CONCLUSIONS: While mutations in TMC1 are not as common a cause of NSHL as those in GJB2, TMC1 should be considered for diagnostic investigations in cases of NSHL in GJB2-negative families.


Assuntos
Conexinas/genética , Surdez/genética , Proteínas de Membrana/genética , Conexina 26 , Surdez/patologia , Feminino , Ligação Genética , Genótipo , Haplótipos/genética , Homozigoto , Humanos , Índia , Masculino , Mutação , Linhagem
6.
Hum Mutat ; 38(7): 816-826, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28370826

RESUMO

Juvenile myoclonic epilepsy (JME) is a common form of epilepsy with a substantial genetic basis to its etiology. While earlier studies have identified EFHC1 as a causative gene for JME, subsequent studies have suggested that ethnicity may play a role in determining expression of the JME phenotype among individuals carrying EFHC1 mutations. Here, we report on our studies on EFHC1 in JME patients from India. We examined the complete structure of the EFHC1 transcript from 480 JME patients and 700 control chromosomes by direct sequencing. Functional correlates of mutations were studied by immunolocalization experiments in cultured mammalian cells and protein homology modeling by in silico methods. Thirteen mutations, of which 11 were previously not known, were identified in 28 JME patients. These mutations accounted for about 6% of the patients examined. Functional studies suggest that these EFHC1 mutations result in microtubule-related abnormalities during cell division. In silico analysis for a subset of mutations suggests that they may affect EFHC1 protein domains, compromising its ability to interact with other proteins. Our observations strengthen the evidence supporting a role for EFHC1 in JME in a population ethnically and geographically distinct from the one in which the gene was initially identified, and broaden the extent of allelic heterogeneity in the gene.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Mutação , Epilepsia Mioclônica Juvenil/genética , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Cromossomos/ultraestrutura , Biologia Computacional , Análise Mutacional de DNA , Feminino , Células HEK293 , Humanos , Índia , Masculino , Domínios Proteicos , Adulto Jovem
7.
Hum Genet ; 136(6): 693-703, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28324217

RESUMO

Hot water epilepsy is sensory epilepsy, wherein seizures are triggered by an unusual stimulus: contact with hot water. Although genetic factors contribute to the etiology of hot water epilepsy, molecular underpinnings of the disorder remain largely unknown. We aimed to identify the molecular genetic basis of the disorder by studying families with two or more of their members affected with hot water epilepsy. Using a combination of genome-wide linkage mapping and whole exome sequencing, a missense variant was identified in SLC1A1 in a three-generation family. Further, we examined SLC1A1in probands of 98 apparently unrelated HWE families with positive histories of seizures provoked by contact with hot water. In doing so, we found three rare variants, p.Asp174Asn, p.Val251Ile and p.Ile304Met in the gene. SLC1A1 is a neuronal glutamate transporter which limits excitotoxicity and its loss-of-function leads to age-dependent neurodegeneration. We examined functional attributes of the variants in cultured mammalian cells. All three non-synonymous variants affected glutamate uptake, exhibited altered glutamate kinetics and anion conductance properties of SLC1A1. These observations provide insights into the molecular basis of hot water epilepsy and show the role of SLC1A1 variants in this intriguing neurobehavioral disorder.


Assuntos
Epilepsia/genética , Transportador 3 de Aminoácido Excitatório/genética , Temperatura Alta , Sequência de Aminoácidos , Animais , Epilepsia/etiologia , Transportador 3 de Aminoácido Excitatório/química , Exoma , Feminino , Ligação Genética , Humanos , Masculino , Técnicas de Patch-Clamp , Linhagem , Homologia de Sequência de Aminoácidos , Água
8.
Indian J Med Res ; 146(4): 489-497, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29434063

RESUMO

Background & objectives: A high incidence of hearing impairment is reported from the village of Dhadkai in the State of Jammu and Kashmir, India. Prevalence of endogamy in this community suggested a common genetic basis for the disorder. A genetic study was undertaken to ascertain the basis for the high incidence of hearing impairment in this region. Methods: In a two-step approach to identify the causative mutation/s, a whole-genome-based linkage analysis of an extended family of 45 members was carried out, which included 23 affected and 22 unaffected members. Mutational analysis for the candidate deafness genes helped reveal causative mutations in the family. In addition, seven deafness-causing genes, Cx26, SLC26A4, CLDN14, TMPRSS3, TMC1, TMIE and USH1C, were analyzed in smaller families with hearing impairment. Results: In the 45-member extended family, the critical chromosomal region mapped to 2p24-p22.The c.2122C>T (p.R708X) mutation in OTOF in 2p24-p22was identified as being the causal change. Linkage to 2p24-p22 locus was not observed in a particular branch of this extended family. Analysis of seven known deafness-causing genes in this branch revealed a mutation, c.254T>A (p.V85D), in CLDN14. Among seven small families unrelated to the 45-member extended family, hearing loss was attributable to p.R708X in OTOF in three families and to p.V85D in CLDN14 in one family; a new mutation c.1668T>A (p.Y556X) SLC26A4 was identified in two families and the causative change could not be identified in one family. Interpretation & conclusions: This study suggested considerable genetic heterogeneity in the causation of hearing loss in Dhadkai. Recessive mutations were observed in at least three genes causing hearing loss: OTOF (p.R708X), SLC26A4 (p.Y556X) and CLDN14 (p.V85D). Mutation p.R708X appeared to be the major cause of hearing impairment in Dhadkai.


Assuntos
Claudinas/genética , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Feminino , Ligação Genética , Predisposição Genética para Doença , Perda Auditiva/epidemiologia , Perda Auditiva/patologia , Humanos , Índia , Masculino , Mutação , Transportadores de Sulfato
9.
Ann Hum Genet ; 80(1): 11-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26620415

RESUMO

Mutations in the gap-junction gene Cx30 (Connexin30, GJB6) are a known cause of hearing loss. Here, we report our findings on a large multigeneration family in which severe to profound sensorineural hearing impairment is associated with a variety of skin-related anomalies. Genome-wide analysis of the family showed that the locus maps to chromosome region 13ptel-q12.1 and that a novel mutation, p.N54K, in Cx30, cosegregates with the phenotype. Unlike wild-type Cx30, p.N54K Cx30 is predominantly localized in the cytoplasm and does not permit transfer of neurobiotin, suggesting improper cellular localization and abolishment of gap-junction activity.


Assuntos
Conexinas/genética , Perda Auditiva Neurossensorial/genética , Dermatopatias Genéticas/genética , Biotina/análogos & derivados , Biotina/metabolismo , Conexina 30 , Análise Mutacional de DNA , Feminino , Ligação Genética , Humanos , Ictiose/genética , Ceratodermia Palmar e Plantar Difusa/genética , Masculino , Linhagem , Fenótipo
10.
PLoS One ; 9(1): e84773, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416283

RESUMO

Mutations in the autosomal genes TMPRSS3, TMC1, USHIC, CDH23 and TMIE are known to cause hereditary hearing loss. To study the contribution of these genes to autosomal recessive, non-syndromic hearing loss (ARNSHL) in India, we examined 374 families with the disorder to identify potential mutations. We found four mutations in TMPRSS3, eight in TMC1, ten in USHIC, eight in CDH23 and three in TMIE. Of the 33 potentially pathogenic variants identified in these genes, 23 were new and the remaining have been previously reported. Collectively, mutations in these five genes contribute to about one-tenth of ARNSHL among the families examined. New mutations detected in this study extend the allelic heterogeneity of the genes and provide several additional variants for structure-function correlation studies. These findings have implications for early DNA-based detection of deafness and genetic counseling of affected families in the Indian subcontinent.


Assuntos
Alelos , Perda Auditiva/genética , Mutação , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas Relacionadas a Caderinas , Caderinas/genética , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Éxons/genética , Feminino , Heterozigoto , Homozigoto , Humanos , Índia , Íntrons/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética
11.
Hum Genet ; 128(2): 123-30, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20467754

RESUMO

We performed a whole genome linkage analysis in a three-generation south Indian family with multiple members affected with juvenile myoclonic epilepsy (JME). The maximum two-point LOD score obtained was 3.32 at recombination fraction (theta) = 0 for D2S2248. The highest multipoint score of 3.59 was observed for the genomic interval between D2S2322 and D2S2228 at the chromosomal region 2q33-q36. Proximal and distal boundaries of the critical genetic interval were defined by D2S116 and D2S2390, respectively. A 24-Mb haplotype was found to co-segregate with JME in the family. While any potentially causative variant in the functional candidate genes, SLC4A3, SLC23A3, SLC11A1 and KCNE4, was not detected, we propose to examine brain-expressed NRP2, MAP2, PAX3, GPR1, TNS1 and DNPEP, and other such positional candidate genes to identify the disease-causing gene for the disorder.


Assuntos
Epilepsia Mioclônica Juvenil/genética , Encéfalo/fisiopatologia , Proteínas de Transporte de Cátions , Genes , Ligação Genética , Haplótipos , Humanos , Escore Lod , Epilepsia Mioclônica Juvenil/fisiopatologia
12.
Hum Genet ; 126(5): 677-83, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19597845

RESUMO

Hot water epilepsy is a reflex or sensory epilepsy in which seizures are triggered by the stimulus of bathing in hot water. Although there is evidence of a genetic basis to its etiology, no gene associated with this disorder has so far been found. In order to identify the genetic locus involved in the pathophysiology of hot water epilepsy, we performed a genome-wide linkage analysis in a four-generation family manifesting the disorder in an autosomal dominant manner. Significant linkage was detected on chromosome 4q24-q28, with the highest two-point LOD score of 3.50 at recombination value (theta) of 0 for the marker D4S402. Centromere-proximal and centromere-distal boundaries of this locus were defined by the markers D4S1572 and D4S2277, respectively. The critical genetic interval spans 22.5 cM and corresponds to about 24 megabases of DNA. The genes NEUROG2, ANK2, UGT8 and CAMK2D, which are known to be expressed in human brain, are strong positional candidates and we propose to examine these and other genes in the locus to identify the causative gene for this intriguing form of epilepsy.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 4 , Epilepsia Reflexa/genética , Banhos/efeitos adversos , Encéfalo/fisiopatologia , Eletroencefalografia , Epilepsia Reflexa/etiologia , Epilepsia Reflexa/fisiopatologia , Feminino , Genes Dominantes , Marcadores Genéticos , Estudo de Associação Genômica Ampla , Temperatura Alta/efeitos adversos , Humanos , Escore Lod , Masculino , Linhagem , Recombinação Genética , Convulsões/genética , Convulsões/fisiopatologia
13.
Hum Genet ; 125(5-6): 541-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19266219

RESUMO

Hot water epilepsy (HWE) is a form of reflex or sensory epilepsy wherein seizures are precipitated by an unusual stimulus, the contact of hot water over the head and body. Genome-wide linkage analysis of a large family with ten affected members, provided evidence of linkage (Z (max) = 3.17 at theta = 0 for D10S412) to chromosome 10q21. Analysis of five additional HWE families, for markers on chromosome 10, further strengthened the evidence of linkage to the same chromosomal region with three out of five families showing concordance for the disease haplotype and providing a two-point LOD score of 4.86 at theta = 0 and 60% penetrance for D10S412. The centromere-proximal and -distal boundaries of the critical genetic interval of about 15 Mb at 10q21.3-q22.3 were defined by D10S581 and D10S201, respectively. Sequence analysis of a group of functional candidate genes, the ion channels KCNMA1, VDAC2 and solute carriers SLC25A16, SLC29A3 revealed no potentially pathogenic mutation. We propose to carry out further analysis of positional candidate genes from this region to identify the gene responsible for this unusual neurobehavioral phenotype.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 10/genética , Epilepsia Reflexa/genética , Genes Dominantes , Análise Mutacional de DNA , Epilepsia Reflexa/epidemiologia , Feminino , Marcadores Genéticos , Haplótipos , Humanos , Índia/epidemiologia , Escore Lod , Masculino , Repetições de Microssatélites , Linhagem
14.
Hum Genet ; 124(6): 669-75, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19030898

RESUMO

Autosomal dominant nonsyndromic hearing loss (ADNSHL) accounts for about one-fifth of hereditary hearing loss in humans. In the present study, we have analyzed a three-generation family with 14 of its members manifesting ADNSHL, using a genome-wide linkage mapping approach. We found a novel locus DFNA59 between the D11S929 and D11S480 markers in the chromosome location 11p14.2-q12.3. The highest two-point lod score of 5.72 at recombination fraction = 0 was obtained for D11S4152, D11S4154, D11S1301, D11S905 and D11S1344. The critical genomic region comprising about 37 megabases of DNA is proposed to carry a gene for ADNSHL in the family. About 50 cochlear-expressed genes mapping to the region are strong candidates which we propose to examine to identify the gene responsible for the hearing impairment.


Assuntos
Cromossomos Humanos Par 11/genética , Perda Auditiva Neurossensorial/genética , Adolescente , Audiometria de Tons Puros , Mapeamento Cromossômico , Feminino , Genes Dominantes , Marcadores Genéticos , Perda Auditiva Bilateral/congênito , Perda Auditiva Bilateral/genética , Perda Auditiva Bilateral/fisiopatologia , Perda Auditiva Neurossensorial/congênito , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Escore Lod , Masculino , Linhagem
15.
Eur J Hum Genet ; 17(4): 502-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18941476

RESUMO

In a study of 530 individuals with non-syndromic, sensorineural hearing loss, we identified 18 mutations at connexin 26 (Cx26), four of which are novel (-23G>T, I33T, 377_383dupTCCGCAT, W172R) and the remaining 14 (ivs1+1G>A, M1V, 35delG, W24X, I35S, V37I, R75W, W77X, 312del14, E120del, Q124X, Y136X, R143W, R184P) being mutations previously described. To gain insight into functional consequences of these mutations, cellular localization of the mutant proteins and their ability to permit lucifer yellow transfer between cells was studied in seven of them (W24X, I33T, I35S, R75W, E120del, W172R and R184P). I35S and R184P showed impaired trafficking of the protein to the plasma membrane. I33T, R75W, E120del and W172R showed predominantly membrane localization but did not form functional gap junction channels. Surprisingly, W24X, a protein-truncating mutation, apparently permits formation of a full-length protein, perhaps due to a stop codon read-through mechanism. These results provide further evidence that Cx26 mutations affect gap junction activity by mis-regulation at multiple levels.


Assuntos
Conexinas/genética , Perda Auditiva Neurossensorial/genética , Mutação , Conexina 26 , Conexinas/análise , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica
16.
Ann Neurol ; 64(2): 158-67, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18756473

RESUMO

OBJECTIVE: To identify the disease locus in a three-generation south Indian family having several of its members affected with idiopathic epilepsy. METHODS: Genome-wide parametric linkage analysis was performed with 382 autosomal markers. Mutational analysis of the positional candidate genes in linked interval was performed by direct sequencing of genomic DNA from the proband in the family. Expression analysis in human adult brain was performed by Western blotting. RESULTS: A novel epilepsy genetic locus on chromosome 3q13.3-q21 was identified by linkage analysis. This locus comprises about 12 megabases of the genomic interval, with its proximal and distal genetic boundaries defined by microsatellite markers, D3S3675 and D3S1551, respectively. In this interval, we found a novel, patient-specific, missense variant, Arg898Gln, at the extracellular calcium sensing receptor (CASR), a gene belonging to the G-protein-coupled receptor family. CASR expression was detected in the temporal lobe, frontal lobe, parietal lobe, cerebellum, and hippocampus. Four additional, potentially pathogenic, missense CASR variants, Glu354Ala, Ile686Val, Ala988Val, and Ala988Gly, were observed in five individuals affected with idiopathic generalized epilepsy. INTERPRETATION: A novel idiopathic epilepsy locus has been mapped on chromosome 3q13.3-q21, as evident by presence of significant genetic linkage. Identification of novel, rare missense CASR variants at evolutionary-conserved residues in epilepsy patients and CASR expression in various subregions of human brain raises an interesting possibility of involvement of CASR in pathophysiology of epileptic disorders.


Assuntos
Cromossomos Humanos Par 3/genética , Epilepsia Generalizada/genética , Ligação Genética/genética , Mutação de Sentido Incorreto/genética , Receptores de Detecção de Cálcio/genética , Adolescente , Epilepsia Generalizada/metabolismo , Epilepsia Generalizada/fisiopatologia , Líquido Extracelular/fisiologia , Feminino , Marcadores Genéticos/genética , Variação Genética/genética , Humanos , Masculino , Linhagem , Locos de Características Quantitativas/genética , Receptores de Detecção de Cálcio/fisiologia , Síndrome
17.
Int J Audiol ; 46(7): 384-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17680470

RESUMO

The aim of the study is to understand the implications of disclosing the results of connexin26 (Cx26) gene testing to the concerned family with hearing impaired individuals. The department of biotechnology is funding a multicentric multidisciplinary team from Jawaharlal Nehru Center for Advanced Scientific Research (Bangalore), AYJNIHH (Mumbai), PGIBMS (Chennai), and MAMC (New Delhi) to profile mutations of deafness genes in India. Under this program, blood samples were taken from various centers and were sent to JNCASR for genetic analysis (screening for Cx26 mutations). This case study is an attempt to bring out issues encountered when disclosing the implications of genetic diagnosis to the concerned family.


Assuntos
Conexinas/genética , Surdez/genética , Revelação , Família , Mutação Puntual/genética , Adulto , Conexina 26 , Humanos , Masculino , Linhagem
18.
Hum Genet ; 121(6): 655-62, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17431681

RESUMO

Juvenile myoclonic epilepsy is a clinically well-defined, age-related common idiopathic generalized epilepsy syndrome with substantial genetic basis to its etiology. We report identification of a novel epilepsy locus at chromosome 5q12-q14 in a family exhibiting autosomal dominant form of juvenile myoclonic epilepsy from south India. The highest two-point LOD score of 3.3344 was obtained for the microsatellite markers D5S641 and D5S459 at 5q14. Centromeric and telomeric chromosomal boundaries of the locus were defined by D5S624 and D5S428, respectively. The 5q12-q14 locus encompasses about 25 megabases of the genomic region and harbours several candidate genes. Further work involving a detailed mutational analysis of the locus, to isolate the gene responsible for the epilepsy disorder in the family, shall help enhance our understanding of molecular basis of epilepsy disorders.


Assuntos
Cromossomos Humanos Par 5/genética , Epilepsia Mioclônica Juvenil/genética , Adulto , Idade de Início , Criança , Mapeamento Cromossômico , Eletroencefalografia , Feminino , Genes Dominantes , Humanos , Índia , Escore Lod , Masculino , Repetições de Microssatélites , Epilepsia Mioclônica Juvenil/fisiopatologia , Linhagem
19.
Epilepsia ; 48(4): 706-12, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17437413

RESUMO

PURPOSE: Although complex idiopathic generalized epilepsies (IGEs) are recognized to have a significant genetic component, as yet there are no known common susceptibility variants. It has recently been suggested that variation in the BRD2 gene confers increased risk of juvenile myoclonic epilepsy (JME), which accounts for around a quarter of all IGE. Here we examine the association between the candidate causal SNP (the promoter variant rs3918149) and JME in five independent cohorts comprising in total 531 JME cases and 1,390 healthy controls. METHODS: The strongest candidate causal variant from the original report (rs3918149) was genotyped across all five cohorts. In an effort to identify novel candidate causal polymorphisms, previously unscreened regions of UTR were resequenced. RESULTS: We observed a significant effect in a small sample recruited in Britain (genotype p = 0.001, allele p = 0.001), a borderline significant effect in a sample recruited in Ireland and no association in larger samples of German, Australian, and Indian populations. There was no association with other common forms of epilepsy or any other clear candidate casual variants in or near the BRD2 region. CONCLUSIONS: The replication of an effect in the British cohort and suggestive evidence from that recruited in Ireland but lack of replication from the larger German, Australian, and Indian cohorts is surprising and difficult to explain. Further replication in carefully matched populations is required. Results presented here do not, however, support a strong effect for susceptibility to JME across populations of European descent.


Assuntos
Variação Genética , Epilepsia Mioclônica Juvenil/genética , Proteínas Serina-Treonina Quinases/genética , Estudos de Casos e Controles , Estudos de Coortes , Heterogeneidade Genética , Predisposição Genética para Doença , Genética Populacional , Genótipo , Humanos , Epilepsia Mioclônica Juvenil/epidemiologia , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Fatores de Risco , Fatores de Transcrição , Reino Unido/epidemiologia , População Branca/genética , População Branca/estatística & dados numéricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...