Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Am J Hum Genet ; 102(3): 468-479, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29429572

RESUMO

Variants affecting the function of different subunits of the BAF chromatin-remodelling complex lead to various neurodevelopmental syndromes, including Coffin-Siris syndrome. Furthermore, variants in proteins containing PHD fingers, motifs recognizing specific histone tail modifications, have been associated with several neurological and developmental-delay disorders. Here, we report eight heterozygous de novo variants (one frameshift, two splice site, and five missense) in the gene encoding the BAF complex subunit double plant homeodomain finger 2 (DPF2). Affected individuals share common clinical features described in individuals with Coffin-Siris syndrome, including coarse facial features, global developmental delay, intellectual disability, speech impairment, and hypoplasia of fingernails and toenails. All variants occur within the highly conserved PHD1 and PHD2 motifs. Moreover, missense variants are situated close to zinc binding sites and are predicted to disrupt these sites. Pull-down assays of recombinant proteins and histone peptides revealed that a subset of the identified missense variants abolish or impaire DPF2 binding to unmodified and modified H3 histone tails. These results suggest an impairment of PHD finger structural integrity and cohesion and most likely an aberrant recognition of histone modifications. Furthermore, the overexpression of these variants in HEK293 and COS7 cell lines was associated with the formation of nuclear aggregates and the recruitment of both wild-type DPF2 and BRG1 to these aggregates. Expression analysis of truncating variants found in the affected individuals indicated that the aberrant transcripts escape nonsense-mediated decay. Altogether, we provide compelling evidence that de novo variants in DPF2 cause Coffin-Siris syndrome and propose a dominant-negative mechanism of pathogenicity.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ligação a DNA/genética , Face/anormalidades , Deformidades Congênitas da Mão/genética , Deficiência Intelectual/genética , Micrognatismo/genética , Mutação/genética , Pescoço/anormalidades , Subunidades Proteicas/genética , Adolescente , Sequência de Aminoácidos , Animais , Células COS , Criança , Pré-Escolar , Chlorocebus aethiops , Proteínas de Ligação a DNA/química , Fácies , Feminino , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Fenótipo , Fatores de Transcrição
2.
Am J Hum Genet ; 94(5): 649-61, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24726472

RESUMO

Recently, we identified in two individuals with intellectual disability (ID) different de novo mutations in DEAF1, which encodes a transcription factor with an important role in embryonic development. To ascertain whether these mutations in DEAF1 are causative for the ID phenotype, we performed targeted resequencing of DEAF1 in an additional cohort of over 2,300 individuals with unexplained ID and identified two additional individuals with de novo mutations in this gene. All four individuals had severe ID with severely affected speech development, and three showed severe behavioral problems. DEAF1 is highly expressed in the CNS, especially during early embryonic development. All four mutations were missense mutations affecting the SAND domain of DEAF1. Altered DEAF1 harboring any of the four amino acid changes showed impaired transcriptional regulation of the DEAF1 promoter. Moreover, behavioral studies in mice with a conditional knockout of Deaf1 in the brain showed memory deficits and increased anxiety-like behavior. Our results demonstrate that mutations in DEAF1 cause ID and behavioral problems, most likely as a result of impaired transcriptional regulation by DEAF1.


Assuntos
Deficiência Intelectual/genética , Transtornos Mentais/genética , Proteínas Nucleares/genética , Distúrbios da Fala/genética , Sequência de Aminoácidos , Animais , Criança , Estudos de Coortes , Análise Mutacional de DNA , Proteínas de Ligação a DNA , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína/genética , Fatores de Transcrição
3.
Hum Genet ; 134(6): 553-68, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25724810

RESUMO

Coffin-Siris syndrome (CSS) and Nicolaides-Baraitser syndrome (NCBRS) are rare intellectual disability/congenital malformation syndromes that represent distinct entities but show considerable clinical overlap. They are caused by mutations in genes encoding members of the BRG1- and BRM-associated factor (BAF) complex. However, there are a number of patients with the clinical diagnosis of CSS or NCBRS in whom the causative mutation has not been identified. In this study, we performed trio-based whole-exome sequencing (WES) in ten previously described but unsolved individuals with the tentative diagnosis of CSS or NCBRS and found causative mutations in nine out of ten individuals. Interestingly, our WES analysis disclosed overlapping differential diagnoses including Wiedemann-Steiner, Kabuki, and Adams-Oliver syndromes. In addition, most likely causative de novo mutations were identified in GRIN2A and SHANK3. Moreover, trio-based WES detected SMARCA2 and SMARCA4 deletions, which had not been annotated in a previous Haloplex target enrichment and next-generation sequencing of known CSS/NCBRS genes emphasizing the advantages of WES as a diagnostic tool. In summary, we discuss the phenotypic and diagnostic challenges in clinical genetics, establish important differential diagnoses, and emphasize the cardinal features and the broad clinical spectrum of BAF complex disorders and other disorders caused by mutations in epigenetic landscapers.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Exoma , Face/anormalidades , Deformidades Congênitas do Pé/diagnóstico , Deformidades Congênitas do Pé/genética , Deformidades Congênitas da Mão/diagnóstico , Deformidades Congênitas da Mão/genética , Hipotricose/diagnóstico , Hipotricose/genética , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Micrognatismo/diagnóstico , Micrognatismo/genética , Mutação , Pescoço/anormalidades , Adulto , Idoso de 80 Anos ou mais , Criança , DNA Helicases/genética , Diagnóstico Diferencial , Fácies , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Receptores de N-Metil-D-Aspartato/genética , Fatores de Transcrição/genética
4.
Am J Hum Genet ; 90(3): 565-72, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22405089

RESUMO

Intellectual disability (ID) is a clinically and genetically heterogeneous common condition that remains etiologically unresolved in the majority of cases. Although several hundred diseased genes have been identified in X-linked, autosomal-recessive, or syndromic types of ID, the establishment of an etiological basis remains a difficult task in unspecific, sporadic cases. Just recently, de novo mutations in SYNGAP1, STXBP1, MEF2C, and GRIN2B were reported as relatively common causes of ID in such individuals. On the basis of a patient with severe ID and a 2.5 Mb microdeletion including ARID1B in chromosomal region 6q25, we performed mutational analysis in 887 unselected patients with unexplained ID. In this cohort, we found eight (0.9%) additional de novo nonsense or frameshift mutations predicted to cause haploinsufficiency. Our findings indicate that haploinsufficiency of ARID1B, a member of the SWI/SNF-A chromatin-remodeling complex, is a common cause of ID, and they add to the growing evidence that chromatin-remodeling defects are an important contributor to neurodevelopmental disorders.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , Adolescente , Adulto , Criança , Pré-Escolar , Cromatina/genética , Estudos de Coortes , Análise Mutacional de DNA/métodos , Éxons , Feminino , Haploinsuficiência , Humanos , Deficiência Intelectual , Masculino , Pessoa de Meia-Idade , Mutação , Adulto Jovem
5.
Lancet ; 380(9854): 1674-82, 2012 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-23020937

RESUMO

BACKGROUND: The genetic cause of intellectual disability in most patients is unclear because of the absence of morphological clues, information about the position of such genes, and suitable screening methods. Our aim was to identify de-novo variants in individuals with sporadic non-syndromic intellectual disability. METHODS: In this study, we enrolled children with intellectual disability and their parents from ten centres in Germany and Switzerland. We compared exome sequences between patients and their parents to identify de-novo variants. 20 children and their parents from the KORA Augsburg Diabetes Family Study were investigated as controls. FINDINGS: We enrolled 51 participants from the German Mental Retardation Network. 45 (88%) participants in the case group and 14 (70%) in the control group had de-novo variants. We identified 87 de-novo variants in the case group, with an exomic mutation rate of 1·71 per individual per generation. In the control group we identified 24 de-novo variants, which is 1·2 events per individual per generation. More participants in the case group had loss-of-function variants than in the control group (20/51 vs 2/20; p=0·022), suggesting their contribution to disease development. 16 patients carried de-novo variants in known intellectual disability genes with three recurrently mutated genes (STXBP1, SYNGAP1, and SCN2A). We deemed at least six loss-of-function mutations in six novel genes to be disease causing. We also identified several missense alterations with potential pathogenicity. INTERPRETATION: After exclusion of copy-number variants, de-novo point mutations and small indels are associated with severe, sporadic non-syndromic intellectual disability, accounting for 45-55% of patients with high locus heterogeneity. Autosomal recessive inheritance seems to contribute little in the outbred population investigated. The large number of de-novo variants in known intellectual disability genes is only partially attributable to known non-specific phenotypes. Several patients did not meet the expected syndromic manifestation, suggesting a strong bias in present clinical syndrome descriptions. FUNDING: German Ministry of Education and Research, European Commission 7th Framework Program, and Swiss National Science Foundation.


Assuntos
Exoma/genética , Deficiência Intelectual/genética , Mutação/genética , Estudos de Casos e Controles , Criança , Feminino , Humanos , Masculino
6.
Behav Brain Funct ; 9: 20, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23718928

RESUMO

BACKGROUND: Intellectual disability (ID) is often associated with behavioral problems or disorders. Mutations in the GRIN2B gene (MRD6, MIM613970) have been identified as a common cause of ID (prevalence of 0.5 - 1% in individuals with ID) associated with EEG and behavioral problems. METHODS: We assessed five GRIN2B mutation carriers aged between 3 and 14 years clinically and via standardized questionnaires to delineate a detailed behavioral phenotype. Parents and teachers rated problem behavior of their affected children by completing the Developmental Behavior Checklist (DBC) and the Conners' Rating Scales Revised (CRS-R:L). RESULTS: All individuals had mild to severe ID and needed guidance in daily routine. They showed characteristic behavior problems with prominent hyperactivity, impulsivity, distractibility and a short attention span. Stereotypies, sleeping problems and a friendly but boundless social behavior were commonly reported. CONCLUSION: Our observations provide an initial delineation of the behavioral phenotype of GRIN2B mutation carriers.


Assuntos
Sintomas Comportamentais/genética , Deficiência Intelectual/genética , Deficiência Intelectual/psicologia , Receptores de N-Metil-D-Aspartato/genética , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação , Fenótipo , Escalas de Graduação Psiquiátrica
7.
Neurogenetics ; 12(2): 155-63, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21287218

RESUMO

In order to identify novel genes involved in mental retardation/intellectual disability, we focused on a microdeletion reported in a patient with a mild form of Wolf-Hirschhorn syndrome. This patient presented with attention-deficit hyperactivity disorder, some learning and fine motor deficits as well as facial abnormalities. The deleted region included three genes. Here, we report the first characterization of one of these genes, C4ORF48. C4ORF48 encodes a short (139 aa) evolutionarily conserved protein with a predicted signal peptide and two potential dibasic convertase cleavage sites. In mice, we demonstrated expression of the corresponding protein exclusively in brain tissue using an anti-mouse C4Orf48 polyclonal antibody. Detailed RNA in situ hybridization experiments revealed expression of C4Orf48 in different zones during cortical and cerebellar development, as well as in almost all cortical and subcortical regions of the adult mouse brain. Based on the present data, we propose that C4Orf48 probably encodes a novel neuropeptide, which, if hemizygously deleted, may be involved in the observed intellectual and fine motor disabilities and thus in the overall neurological aspects of Wolf-Hirschhorn syndrome.


Assuntos
Cerebelo/embriologia , Neocórtex/embriologia , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/genética , Síndrome de Wolf-Hirschhorn/genética , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células COS , Cerebelo/metabolismo , Chlorocebus aethiops , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Proteínas/genética , Proteínas/fisiologia , Homologia de Sequência de Aminoácidos
8.
J Med Genet ; 47(2): 91-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19843505

RESUMO

BACKGROUND: The authors observed a patient with a cryptic subtelomeric de novo balanced translocation 46,XY.ish t(11;20)(p15.4;q13.2) presenting with severe mental retardation, muscular hypotonia, seizures, bilateral sensorineural hearing loss, submucous cleft palate, persistent ductus Botalli, unilateral cystic kidney dysplasia and frequent infections. METHODS AND RESULTS: Fluorescence in situ hybridisation mapping and sequencing of the translocation breakpoints showed that no known genes are disrupted at 20q13.2 and that ST5 (suppression of tumorigenicity 5; MIM 140750) is disrupted on 11p15.4. By quantitative PCR from different human tissues, the authors found ST5 to be relatively evenly expressed in fetal tissues. ST5 expression was more pronounced in adult brain, kidney and muscle than in the corresponding fetal tissues, whereas expression in other tissues was generally lower than in the fetal tissue. Using RNA in situ hybridisation in mouse, the authors found that St5 is expressed in the frontal cortex during embryonic development. In adult mouse brain, expression of St5 was especially high in the hippocampal area and cerebellum. CONCLUSION: Hence, the authors suppose that ST5 plays an important role in central nervous system development probably due to disturbance of DENN-domain-mediated vesicle formation and neurotransmitter trafficking. Thus, these findings implicate ST5 in the aetiology of mental retardation, seizures and multiple congenital anomalies.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ligação a DNA/genética , Deficiência Intelectual/genética , Proteínas Supressoras de Tumor/genética , Animais , Pré-Escolar , Pontos de Quebra do Cromossomo , Mapeamento Cromossômico , Análise Mutacional de DNA , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Dosagem de Genes , Histocitoquímica , Humanos , Hibridização in Situ Fluorescente , Masculino , Camundongos , Especificidade de Órgãos , RNA , Tomografia Óptica , Proteínas Supressoras de Tumor/metabolismo
9.
J Mol Med (Berl) ; 99(12): 1755-1768, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34536092

RESUMO

Pathogenic variants in aminoacyl-tRNA synthetases (ARS1) cause a diverse spectrum of autosomal recessive disorders. Tyrosyl tRNA synthetase (TyrRS) is encoded by YARS1 (cytosolic, OMIM*603,623) and is responsible of coupling tyrosine to its specific tRNA. Next to the enzymatic domain, TyrRS has two additional functional domains (N-Terminal TyrRSMini and C-terminal EMAP-II-like domain) which confer cytokine-like functions. Mutations in YARS1 have been associated with autosomal-dominant Charcot-Marie-Tooth (CMT) neuropathy type C and a heterogenous group of autosomal recessive, multisystem diseases. We identified 12 individuals from 6 families with the recurrent homozygous missense variant c.1099C > T;p.(Arg367Trp) (NM_003680.3) in YARS1. This variant causes a multisystem disorder with developmental delay, microcephaly, failure to thrive, short stature, muscular hypotonia, ataxia, brain anomalies, microcytic anemia, hepatomegaly, and hypothyroidism. In silico analyses show that the p.(Arg367Trp) does not affect the catalytic domain responsible of enzymatic coupling, but destabilizes the cytokine-like C-terminal domain. The phenotype associated with p.(Arg367Trp) is distinct from the other biallelic pathogenic variants that reside in different functional domains of TyrRS which all show some common, but also divergent clinical signs [(e.g., p.(Phe269Ser)-retinal anomalies, p.(Pro213Leu)/p.(Gly525Arg)-mild ID, p.(Pro167Thr)-high fatality)]. The diverse clinical spectrum of ARS1-associated disorders is related to mutations affecting the various non-canonical domains of ARS1, and impaired protein translation is likely not the exclusive disease-causing mechanism of YARS1- and ARS1-associated neurodevelopmental disorders. KEY MESSAGES: The missense variant p.(Arg367Trp) in YARS1 causes a distinct multisystem disorder. p.(Arg367Trp) affects a non-canonical domain with cytokine-like functions. Phenotypic heterogeneity associates with the different affected YARS1 domains. Impaired protein translation is likely not the exclusive mechanism of ARS1-associated disorders.


Assuntos
Transtornos do Neurodesenvolvimento/genética , Tirosina-tRNA Ligase/genética , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação de Sentido Incorreto , Fenótipo , Conformação Proteica , Tirosina-tRNA Ligase/química , Sequenciamento do Exoma
10.
Hum Mol Genet ; 17(2): 201-14, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17925330

RESUMO

Wolf-Hirschhorn syndrome (WHS) is a complex congenital syndrome caused by a monoallelic deletion of the short arm of chromosome 4. Seizures in WHS have been associated with deletion of LETM1 gene. LETM1 encodes for the human homologue of yeast Mdm38p, a mitochondria-shaping protein of unclear function. Here we show that human LETM1 is located in the inner membrane, exposed to the matrix and oligomerized in higher molecular weight complexes of unknown composition. Down-regulation of LETM1 did not disrupt these complexes, but led to DRP1-independent fragmentation of the mitochondrial network. Fragmentation was not associated with changes in the levels of respiratory chain complexes, or with obvious or latent mitochondrial dysfunction, but was recovered by nigericin, which catalyzes the electroneutral exchange of K+ against H+. Down-regulation of LETM1 caused 'necrosis-like' death, without activation of caspases and not inhibited by overexpression of Bcl-2. Primary fibroblasts from a WHS patient displayed reduced LETM1 mRNA and protein, but mitochondrial morphology was surprisingly unaffected, raising the question of whether and how WHS patients counteract the consequences of monoallelic deletion of LETM1. LETM1 highlights the relationship between mitochondrial ion homeostasis, integrity of the mitochondrial network and cell viability.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Síndrome de Wolf-Hirschhorn/genética , Síndrome de Wolf-Hirschhorn/metabolismo , Proteínas de Ligação ao Cálcio/análise , Sobrevivência Celular , Fibroblastos/citologia , Deleção de Genes , Humanos , Proteínas de Membrana/análise , Membranas Mitocondriais/química , Necrose , Forma das Organelas
11.
Neurogenetics ; 10(4): 275-87, 2009 10.
Artigo em Inglês | MEDLINE | ID: mdl-19290556

RESUMO

Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous disorder. All mendelian patterns of inheritance have been described. We identified a homozygous p.A335V mutation in the MED25 gene in an extended Costa Rican family with autosomal recessively inherited Charcot-Marie-Tooth neuropathy linked to the CMT2B2 locus in chromosome 19q13.3. MED25, also known as ARC92 and ACID1, is a subunit of the human activator-recruited cofactor (ARC), a family of large transcriptional coactivator complexes related to the yeast Mediator. MED25 was identified by virtue of functional association with the activator domains of multiple cellular and viral transcriptional activators. Its exact physiological function in transcriptional regulation remains obscure. The CMT2B2-associated missense amino acid substitution p.A335V is located in a proline-rich region with high affinity for SH3 domains of the Abelson type. The mutation causes a decrease in binding specificity leading to the recognition of a broader range of SH3 domain proteins. Furthermore, Med25 is coordinately expressed with Pmp22 gene dosage and expression in transgenic mice and rats. These results suggest a potential role of this protein in the molecular etiology of CMT2B2 and suggest a potential, more general role of MED25 in gene dosage sensitive peripheral neuropathy pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Substituição de Aminoácidos , Proteínas de Ciclo Celular , Doença de Charcot-Marie-Tooth/genética , Complexo Mediador , Proteínas da Mielina , Proteínas Nucleares , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Doença de Charcot-Marie-Tooth/fisiopatologia , Costa Rica , Análise Mutacional de DNA , Modelos Animais de Doenças , Feminino , Dosagem de Genes , Genótipo , Humanos , Masculino , Complexo Mediador/química , Complexo Mediador/genética , Complexo Mediador/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Linhagem , Conformação Proteica , Ratos
12.
Eur J Hum Genet ; 24(4): 556-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26153216

RESUMO

Intellectual disability (ID) affects 2-3% of the population. In the past, many genetic causes of ID remained unidentified due to its vast heterogeneity. Recently, whole exome sequencing (WES) studies have shown that de novo variants underlie a significant portion of sporadic cases of ID. Applying WES to patients with ID or global developmental delay at different centers, we identified three individuals with distinct de novo variants in HIVEP2 (human immunodeficiency virus type I enhancer binding protein), which belongs to a family of zinc-finger-containing transcriptional proteins involved in growth and development. Two of the variants were nonsense changes, and one was a 1 bp deletion resulting in a premature stop codon that was reported previously without clinical detail. In silico prediction programs suggest loss-of-function in the mutated allele leading to haploinsufficiency as a putative mechanism in all three individuals. All three patients presented with moderate-to-severe ID, minimal structural brain anomalies, hypotonia, and mild dysmorphic features. Growth parameters were in the normal range except for borderline microcephaly at birth in one patient. Two of the patients exhibited behavioral anomalies including hyperactivity and aggression. Published functional data suggest a neurodevelopmental role for HIVEP2, and several of the genes regulated by HIVEP2 are implicated in brain development, for example, SSTR-2, c-Myc, and genes of the NF-κB pathway. In addition, HIVEP2-knockout mice exhibit several working memory deficits, increased anxiety, and hyperactivity. On the basis of the genotype-phenotype correlation and existing functional data, we propose HIVEP2 as a causative ID gene.


Assuntos
Códon sem Sentido , Proteínas de Ligação a DNA/genética , Deficiência Intelectual/genética , Fatores de Transcrição/genética , Pré-Escolar , Exoma , Feminino , Humanos , Lactente , Deficiência Intelectual/diagnóstico , Masculino , Adulto Jovem
14.
Anticancer Res ; 25(3A): 1611-4, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16033069

RESUMO

BACKGROUND: Prostate cancer is the leading tumor of the male in Western societies. Genetic alterations of the androgen receptor gene are known in the advanced metastatic disease. In this study, the androgen receptor gene was tested in two human prostate cancer cell lines, the androgen-sensitive PC-EW and the androgen-independent PC-OR. MATERIALS AND METHODS: Genomic DNA was isolated from two cell lines from metastatic prostate adenocarcinoma in heterotransplanted male athymic nude (nu/nu) Balb/c mice. Mutation screening was performed by sequencing of exons 1-8 of the human androgen receptor gene. RESULTS: Despite two polymorphisms found in the transactivation domain of hAR exon 1, no point mutations were detected in the hAR gene of both cell lines. CONCLUSION: Point mutations of hAR are not necessary for metastatic prostate cancer, while alterations in the solyglutamine and polyglycine repeat region in exon 1 of the MR gene are more often found. These repeats are two of many genetic influences that contribute to the overall risk of developing prostate cancer.


Assuntos
Polimorfismo Genético , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Mutação Puntual , Neoplasias da Próstata/patologia
15.
Anticancer Res ; 25(3A): 1607-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16033068

RESUMO

BACKGROUND: The incidence and mortality rate of prostate cancer has been steadily increasing in most countries worldwide. A potential implication of the progesterone receptor has been reported in prostatic carcinogenesis. In this study, an allele of human progesterone receptor gene (PROGINS), which was demonstrated to be associated with an increased risk of sporadic ovarian cancer, was tested in two human prostate cancer cell lines, PC-EW and PC-OR. MATERIALS AND METHODS: Genomic DNA was isolated from the cell lines in athymic nude mice. The polymorphisms in exon 4 and exon 5 and the insertion in intron G (PROGINS) were identified by sequencing and gel electrophoresis. RESULTS: The PROGINS allele and the polymorphisms in exon 4 and exon 5 were found heterozygous in the PC-OR cell line but not in the PC-EW cell line. CONCLUSION: We described the polymorphisms of exon 4, exon 5 and PROGINS in prostate cancer. Mutation screening of the PGR gene may provide information for risk assessment of developing prostate cancer.


Assuntos
Adenocarcinoma/genética , Polimorfismo Genético , Neoplasias da Próstata/genética , Receptores de Progesterona/genética , Adenocarcinoma/patologia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias da Próstata/patologia
16.
Eur J Hum Genet ; 23(5): 602-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25099252

RESUMO

Recent studies revealed the power of whole-exome sequencing to identify mutations in sporadic cases with non-syndromic intellectual disability. We now identified de novo missense variants in NAA10 in two unrelated individuals, a boy and a girl, with severe global developmental delay but without any major dysmorphism by trio whole-exome sequencing. Both de novo variants were predicted to be deleterious, and we excluded other variants in this gene. This X-linked gene encodes N-alpha-acetyltransferase 10, the catalytic subunit of the NatA complex involved in multiple cellular processes. A single hypomorphic missense variant p.(Ser37Pro) was previously associated with Ogden syndrome in eight affected males from two different families. This rare disorder is characterized by a highly recognizable phenotype, global developmental delay and results in death during infancy. In an attempt to explain the discrepant phenotype, we used in vitro N-terminal acetylation assays which suggested that the severity of the phenotype correlates with the remaining catalytic activity. The variant in the Ogden syndrome patients exhibited a lower activity than the one seen in the boy with intellectual disability, while the variant in the girl was the most severe exhibiting only residual activity in the acetylation assays used. We propose that N-terminal acetyltransferase deficiency is clinically heterogeneous with the overall catalytic activity determining the phenotypic severity.


Assuntos
Deficiências do Desenvolvimento/genética , Estudos de Associação Genética , Mutação de Sentido Incorreto , Acetiltransferase N-Terminal A/genética , Acetiltransferase N-Terminal E/genética , Sequência de Aminoácidos , Criança , Pré-Escolar , Análise Mutacional de DNA , Deficiências do Desenvolvimento/diagnóstico , Éxons , Fácies , Feminino , Loci Gênicos , Humanos , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Acetiltransferase N-Terminal A/química , Acetiltransferase N-Terminal E/química , Linhagem , Fenótipo , Conformação Proteica , Alinhamento de Sequência
17.
Eur J Hum Genet ; 23(6): 753-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25138099

RESUMO

Intellectual disability (ID) has an estimated prevalence of 2-3%. Due to its extreme heterogeneity, the genetic basis of ID remains elusive in many cases. Recently, whole exome sequencing (WES) studies revealed that a large proportion of sporadic cases are caused by de novo gene variants. To identify further genes involved in ID, we performed WES in 250 patients with unexplained ID and their unaffected parents and included exomes of 51 previously sequenced child-parents trios in the analysis. Exome analysis revealed de novo intragenic variants in SET domain-containing 5 (SETD5) in two patients. One patient carried a nonsense variant, and the other an 81 bp deletion located across a splice-donor site. Chromosomal microarray diagnostics further identified four de novo non-recurrent microdeletions encompassing SETD5. CRISPR/Cas9 mutation modelling of the two intragenic variants demonstrated nonsense-mediated decay of the resulting transcripts, pointing to a loss-of-function (LoF) and haploinsufficiency as the common disease-causing mechanism of intragenic SETD5 sequence variants and SETD5-containing microdeletions. In silico domain prediction of SETD5, a predicted SET domain-containing histone methyltransferase (HMT), substantiated the presence of a SET domain and identified a novel putative PHD domain, strengthening a functional link to well-known histone-modifying ID genes. All six patients presented with ID and certain facial dysmorphisms, suggesting that SETD5 sequence variants contribute substantially to the microdeletion 3p25.3 phenotype. The present report of two SETD5 LoF variants in 301 patients demonstrates a prevalence of 0.7% and thus SETD5 variants as a relatively frequent cause of ID.


Assuntos
Códon sem Sentido , Deficiência Intelectual/genética , Metiltransferases/genética , Fenótipo , Adolescente , Sequência de Aminoácidos , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 3/genética , Exoma , Feminino , Humanos , Masculino , Metiltransferases/química , Dados de Sequência Molecular , Linhagem , Polimorfismo de Nucleotídeo Único , Estrutura Terciária de Proteína , Síndrome , Adulto Jovem
19.
Gene ; 312: 165-71, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12909352

RESUMO

A human myosin heavy chain gene was identified in chromosome 19q13 by computational sequence analysis, RT-PCR and DNA sequencing of the cDNA. The complete cDNA has a length of 6786 bp and comprises 41 exons (40 coding) included in 108 kb of genomic sequence. Alternative splicing variants were also identified. The gene is expressed in a multitude of tissues, but mainly in small intestine, colon and skeletal muscle. The putative protein (228 kDa) carries the common myosin domains and presents high homology with the non-muscle myosin heavy chains (MYH9 and MYH10) as well as the smooth muscle myosin heavy chain MYH11. Nevertheless, phylogenetic analysis indicated that these homologous proteins are more related among themselves than to MYH14, suggesting that possibly this myosin heavy chain should be classified in a new myosin-subfamily.


Assuntos
Cromossomos Humanos Par 19/genética , Cadeias Pesadas de Miosina/genética , Northern Blotting , DNA Complementar/química , DNA Complementar/genética , Feminino , Expressão Gênica , Humanos , Masculino , Dados de Sequência Molecular , Miosina Tipo II , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA
20.
Eur J Hum Genet ; 22(2): 289-92, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23674175

RESUMO

Intellectual disability (ID) is frequent in the general population, with 1 in 50 individuals directly affected worldwide. The multiple etiologies include X-linked ID (XLID). Among syndromic XLID, few syndromes present severe ID associated with postnatal microcephaly and midline stereotypic hand movements. We report on three male patients with ID, midline stereotypic hand movements, hypotonia, hyperkinesia, strabismus, as well as seizures (2/3), and non-inherited and postnatal onset microcephaly (2/3). Using array CGH and exome sequencing we characterised two truncating mutations in IQSEC2, namely two de novo intragenic duplication mapped to the Xp11.22 region and a nonsense mutation in exon 7. We propose that truncating mutations in IQSEC2 are responsible for syndromic severe ID in male patients and should be screened in patients without mutations in MECP2, FOXG1, CDKL5 and MEF2C.


Assuntos
Anormalidades Múltiplas/diagnóstico , Fatores de Troca do Nucleotídeo Guanina/genética , Deficiência Intelectual/diagnóstico , Anormalidades Múltiplas/genética , Adulto , Pré-Escolar , Códon sem Sentido , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Deficiência Intelectual/classificação , Deficiência Intelectual/genética , Masculino , Fenótipo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa