Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Neurosci ; 39: 409-35, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27145913

RESUMO

Studies of syndromic hydrocephalus have led to the identification of >100 causative genes. Even though this work has illuminated numerous pathways associated with hydrocephalus, it has also highlighted the fact that the genetics underlying this phenotype are more complex than anticipated originally. Mendelian forms of hydrocephalus account for a small fraction of the genetic burden, with clear evidence of background-dependent effects of alleles on penetrance and expressivity of driver mutations in key developmental and homeostatic pathways. Here, we synthesize the currently implicated genes and inheritance paradigms underlying hydrocephalus, grouping causal loci into functional modules that affect discrete, albeit partially overlapping, cellular processes. These in turn have the potential to both inform pathomechanism and assist in the rational molecular classification of a clinically heterogeneous phenotype. Finally, we discuss conceptual methods that can lead to enhanced gene identification and dissection of disease basis, knowledge that will potentially form a foundation for the design of future therapeutics.


Assuntos
Encéfalo/patologia , Predisposição Genética para Doença , Hidrocefalia/genética , Mutação/genética , Animais , Líquido Cefalorraquidiano/metabolismo , Humanos , Hidrocefalia/patologia , Fenótipo
2.
Am J Hum Genet ; 101(3): 466-477, 2017 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-28886345

RESUMO

RAC1 is a widely studied Rho GTPase, a class of molecules that modulate numerous cellular functions essential for normal development. RAC1 is highly conserved across species and is under strict mutational constraint. We report seven individuals with distinct de novo missense RAC1 mutations and varying degrees of developmental delay, brain malformations, and additional phenotypes. Four individuals, each harboring one of c.53G>A (p.Cys18Tyr), c.116A>G (p.Asn39Ser), c.218C>T (p.Pro73Leu), and c.470G>A (p.Cys157Tyr) variants, were microcephalic, with head circumferences between -2.5 to -5 SD. In contrast, two individuals with c.151G>A (p.Val51Met) and c.151G>C (p.Val51Leu) alleles were macrocephalic with head circumferences of +4.16 and +4.5 SD. One individual harboring a c.190T>G (p.Tyr64Asp) allele had head circumference in the normal range. Collectively, we observed an extraordinary spread of ∼10 SD of head circumferences orchestrated by distinct mutations in the same gene. In silico modeling, mouse fibroblasts spreading assays, and in vivo overexpression assays using zebrafish as a surrogate model demonstrated that the p.Cys18Tyr and p.Asn39Ser RAC1 variants function as dominant-negative alleles and result in microcephaly, reduced neuronal proliferation, and cerebellar abnormalities in vivo. Conversely, the p.Tyr64Asp substitution is constitutively active. The remaining mutations are probably weakly dominant negative or their effects are context dependent. These findings highlight the importance of RAC1 in neuronal development. Along with TRIO and HACE1, a sub-category of rare developmental disorders is emerging with RAC1 as the central player. We show that ultra-rare disorders caused by private, non-recurrent missense mutations that result in varying phenotypes are challenging to dissect, but can be delineated through focused international collaboration.


Assuntos
Encefalopatias/genética , Deficiências do Desenvolvimento/genética , Microcefalia/genética , Mutação de Sentido Incorreto , Proteínas rac1 de Ligação ao GTP/genética , Adolescente , Sequência de Aminoácidos , Animais , Encefalopatias/patologia , Criança , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Feminino , Humanos , Lactente , Masculino , Camundongos , Microcefalia/patologia , Linhagem , Fenótipo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
3.
Hum Genomics ; 13(1): 19, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30992063

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis [1] is a genetically heterogeneous neurodegenerative disorder, characterized by late-onset degeneration of motor neurons leading to progressive limb and bulbar weakness, as well as of the respiratory muscles, which is the primary cause of disease fatality. To date, over 25 genes have been implicated as causative in ALS with C9orf72, SOD1, FUS, and TARDBP accounting for the majority of genetically positive cases. RESULTS: We identified two patients of Italian and French ancestry with a clinical diagnosis of juvenile-onset ALS who were mutation-negative in any of the known ALS causative genes. Starting with the index case, a consanguineous family of Italian origin, we performed whole-exome sequencing and identified candidate pathogenic mutations in 35 genes, 27 of which were homozygous. We next parsed all candidates against a cohort of 3641 ALS cases; only ATP13A2 was found to harbor recessive changes, in a patient with juvenile-onset ALS, similar to the index case. In vivo complementation of ATP13A2 using a zebrafish surrogate model that focused on the assessment of motor neuron morphology and cerebellar integrity confirmed the role of this gene in central and peripheral nervous system maintenance and corroborated the damaging direction of effect of the change detected in the index case of this study. CONCLUSIONS: We here expand the phenotypic spectrum associated with genetic variants in ATP13A2 that previously comprised Kufor-Rakeb syndrome, spastic paraplegia 78, and neuronal ceroid lipofuscinosis type 12 (CLN12), to also include juvenile-onset ALS, as supported by both genetic and functional data. Our findings highlight the importance of establishing a complete genetic profile towards obtaining an accurate clinical diagnosis.


Assuntos
Esclerose Lateral Amiotrófica/genética , Predisposição Genética para Doença , ATPases Translocadoras de Prótons/genética , Adulto , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios Motores/patologia , Mutação/genética , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/patologia , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/patologia , Linhagem , Fenótipo , Sequenciamento do Exoma , Peixe-Zebra
4.
Hum Mutat ; 40(9): 1474-1485, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31260570

RESUMO

The CAGI-5 pericentriolar material 1 (PCM1) challenge aimed to predict the effect of 38 transgenic human missense mutations in the PCM1 protein implicated in schizophrenia. Participants were provided with 16 benign variants (negative controls), 10 hypomorphic, and 12 loss of function variants. Six groups participated and were asked to predict the probability of effect and standard deviation associated to each mutation. Here, we present the challenge assessment. Prediction performance was evaluated using different measures to conclude in a final ranking which highlights the strengths and weaknesses of each group. The results show a great variety of predictions where some methods performed significantly better than others. Benign variants played an important role as negative controls, highlighting predictors biased to identify disease phenotypes. The best predictor, Bromberg lab, used a neural-network-based method able to discriminate between neutral and non-neutral single nucleotide polymorphisms. The CAGI-5 PCM1 challenge allowed us to evaluate the state of the art techniques for interpreting the effect of novel variants for a difficult target protein.


Assuntos
Autoantígenos/genética , Proteínas de Ciclo Celular/genética , Biologia Computacional/métodos , Mutação de Sentido Incorreto , Esquizofrenia/genética , Bases de Dados Genéticas , Predisposição Genética para Doença , Humanos , Redes Neurais de Computação , Fenótipo , Polimorfismo de Nucleotídeo Único
5.
Am J Hum Genet ; 96(5): 816-25, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25865493

RESUMO

Transcription factors operate in developmental processes to mediate inductive events and cell competence, and perturbation of their function or regulation can dramatically affect morphogenesis, organogenesis, and growth. We report that a narrow spectrum of amino-acid substitutions within the transactivation domain of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a leucine zipper-containing transcription factor of the AP1 superfamily, profoundly affect development. Seven different de novo missense mutations involving conserved residues of the four GSK3 phosphorylation motifs were identified in eight unrelated individuals. The distinctive clinical phenotype, for which we propose the eponym Aymé-Gripp syndrome, is not limited to lens and eye defects as previously reported for MAF/Maf loss of function but includes sensorineural deafness, intellectual disability, seizures, brachycephaly, distinctive flat facial appearance, skeletal anomalies, mammary gland hypoplasia, and reduced growth. Disease-causing mutations were demonstrated to impair proper MAF phosphorylation, ubiquitination and proteasomal degradation, perturbed gene expression in primary skin fibroblasts, and induced neurodevelopmental defects in an in vivo model. Our findings nosologically and clinically delineate a previously poorly understood recognizable multisystem disorder, provide evidence for MAF governing a wider range of developmental programs than previously appreciated, and describe a novel instance of protein dosage effect severely perturbing development.


Assuntos
Catarata/genética , Surdez/genética , Quinase 3 da Glicogênio Sintase/genética , Deficiência Intelectual/genética , Proteínas Proto-Oncogênicas c-maf/genética , Catarata/patologia , Síndrome de Down/genética , Síndrome de Down/patologia , Humanos , Deficiência Intelectual/patologia , Mutação , Fenótipo , Fosforilação , Convulsões/genética , Convulsões/patologia
6.
Am J Hum Genet ; 96(2): 245-57, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25597510

RESUMO

We studied a group of individuals with elevated urinary excretion of 3-methylglutaconic acid, neutropenia that can develop into leukemia, a neurological phenotype ranging from nonprogressive intellectual disability to a prenatal encephalopathy with progressive brain atrophy, movement disorder, cataracts, and early death. Exome sequencing of two unrelated individuals and subsequent Sanger sequencing of 16 individuals with an overlapping phenotype identified a total of 14 rare, predicted deleterious alleles in CLPB in 14 individuals from 9 unrelated families. CLPB encodes caseinolytic peptidase B homolog ClpB, a member of the AAA+ protein family. To evaluate the relevance of CLPB in the pathogenesis of this syndrome, we developed a zebrafish model and an in vitro assay to measure ATPase activity. Suppression of clpb in zebrafish embryos induced a central nervous system phenotype that was consistent with cerebellar and cerebral atrophy that could be rescued by wild-type, but not mutant, human CLPB mRNA. Consistent with these data, the loss-of-function effect of one of the identified variants (c.1222A>G [p.Arg408Gly]) was supported further by in vitro evidence with the mutant peptides abolishing ATPase function. Additionally, we show that CLPB interacts biochemically with ATP2A2, known to be involved in apoptotic processes in severe congenital neutropenia (SCN) 3 (Kostmann disease [caused by HAX1 mutations]). Taken together, mutations in CLPB define a syndrome with intellectual disability, congenital neutropenia, progressive brain atrophy, movement disorder, cataracts, and 3-methylglutaconic aciduria.


Assuntos
Anormalidades Múltiplas/genética , Encéfalo/patologia , Endopeptidase Clp/genética , Deficiência Intelectual/genética , Erros Inatos do Metabolismo/genética , Anormalidades Múltiplas/patologia , Adenosina Trifosfatases/metabolismo , Animais , Atrofia/genética , Atrofia/patologia , Sequência de Bases , Catarata/genética , Catarata/patologia , Endopeptidase Clp/metabolismo , Exoma/genética , Humanos , Deficiência Intelectual/patologia , Erros Inatos do Metabolismo/patologia , Dados de Sequência Molecular , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/patologia , Neutropenia/genética , Neutropenia/patologia , Polimorfismo de Nucleotídeo Único/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Análise de Sequência de DNA , Peixe-Zebra
7.
Am J Hum Genet ; 97(6): 922-32, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26637982

RESUMO

We describe an X-linked genetic syndrome associated with mutations in TAF1 and manifesting with global developmental delay, intellectual disability (ID), characteristic facial dysmorphology, generalized hypotonia, and variable neurologic features, all in male individuals. Simultaneous studies using diverse strategies led to the identification of nine families with overlapping clinical presentations and affected by de novo or maternally inherited single-nucleotide changes. Two additional families harboring large duplications involving TAF1 were also found to share phenotypic overlap with the probands harboring single-nucleotide changes, but they also demonstrated a severe neurodegeneration phenotype. Functional analysis with RNA-seq for one of the families suggested that the phenotype is associated with downregulation of a set of genes notably enriched with genes regulated by E-box proteins. In addition, knockdown and mutant studies of this gene in zebrafish have shown a quantifiable, albeit small, effect on a neuronal phenotype. Our results suggest that mutations in TAF1 play a critical role in the development of this X-linked ID syndrome.


Assuntos
Deficiências do Desenvolvimento/genética , Histona Acetiltransferases/genética , Deficiência Intelectual/genética , Doenças Neurodegenerativas/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Adolescente , Animais , Criança , Pré-Escolar , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Elementos E-Box , Fácies , Família , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Humanos , Lactente , Padrões de Herança , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Mutação , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Linhagem , Fenótipo , Transdução de Sinais , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Adulto Jovem , Peixe-Zebra
8.
Genome Res ; 25(2): 155-66, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25561519

RESUMO

RNA polymerase III (Pol III) synthesizes tRNAs and other small noncoding RNAs to regulate protein synthesis. Dysregulation of Pol III transcription has been linked to cancer, and germline mutations in genes encoding Pol III subunits or tRNA processing factors cause neurogenetic disorders in humans, such as hypomyelinating leukodystrophies and pontocerebellar hypoplasia. Here we describe an autosomal recessive disorder characterized by cerebellar hypoplasia and intellectual disability, as well as facial dysmorphic features, short stature, microcephaly, and dental anomalies. Whole-exome sequencing revealed biallelic missense alterations of BRF1 in three families. In support of the pathogenic potential of the discovered alleles, suppression or CRISPR-mediated deletion of brf1 in zebrafish embryos recapitulated key neurodevelopmental phenotypes; in vivo complementation showed all four candidate mutations to be pathogenic in an apparent isoform-specific context. BRF1 associates with BDP1 and TBP to form the transcription factor IIIB (TFIIIB), which recruits Pol III to target genes. We show that disease-causing mutations reduce Brf1 occupancy at tRNA target genes in Saccharomyces cerevisiae and impair cell growth. Moreover, BRF1 mutations reduce Pol III-related transcription activity in vitro. Taken together, our data show that BRF1 mutations that reduce protein activity cause neurodevelopmental anomalies, suggesting that BRF1-mediated Pol III transcription is required for normal cerebellar and cognitive development.


Assuntos
Anormalidades Múltiplas/genética , Deficiência Intelectual/genética , Mutação , RNA Polimerase III/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Transcrição Gênica , Anormalidades Múltiplas/diagnóstico , Adolescente , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Encéfalo/patologia , Proliferação de Células , Criança , Pré-Escolar , Exoma , Fácies , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Deficiência Intelectual/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Linhagem , Fenótipo , Conformação Proteica , Isoformas de Proteínas , Irmãos , Síndrome , Fatores Associados à Proteína de Ligação a TATA/química , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Peixe-Zebra
9.
Brain ; 140(5): 1267-1279, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28335020

RESUMO

Progressive encephalopathy with oedema, hypsarrhythmia, and optic atrophy (PEHO) syndrome is an early childhood onset, severe autosomal recessive encephalopathy characterized by extreme cerebellar atrophy due to almost total granule neuron loss. By combining homozygosity mapping in Finnish families with Sanger sequencing of positional candidate genes and with exome sequencing a homozygous missense substitution of leucine for serine at codon 31 in ZNHIT3 was identified as the primary cause of PEHO syndrome. ZNHIT3 encodes a nuclear zinc finger protein previously implicated in transcriptional regulation and in small nucleolar ribonucleoprotein particle assembly and thus possibly to pre-ribosomal RNA processing. The identified mutation affects a highly conserved amino acid residue in the zinc finger domain of ZNHIT3. Both knockdown and genome editing of znhit3 in zebrafish embryos recapitulate the patients' cerebellar defects, microcephaly and oedema. These phenotypes are rescued by wild-type, but not mutant human ZNHIT3 mRNA, suggesting that the patient missense substitution causes disease through a loss-of-function mechanism. Transfection of cell lines with ZNHIT3 expression vectors showed that the PEHO syndrome mutant protein is unstable. Immunohistochemical analysis of mouse cerebellar tissue demonstrated ZNHIT3 to be expressed in proliferating granule cell precursors, in proliferating and post-mitotic granule cells, and in Purkinje cells. Knockdown of Znhit3 in cultured mouse granule neurons and ex vivo cerebellar slices indicate that ZNHIT3 is indispensable for granule neuron survival and migration, consistent with the zebrafish findings and patient neuropathology. These results suggest that loss-of-function of a nuclear regulator protein underlies PEHO syndrome and imply that establishment of its spatiotemporal interaction targets will be the basis for developing therapeutic approaches and for improved understanding of cerebellar development.


Assuntos
Edema Encefálico/genética , Edema Encefálico/patologia , Cerebelo/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Atrofia Óptica/genética , Atrofia Óptica/patologia , Espasmos Infantis/genética , Espasmos Infantis/patologia , Animais , Complexo do Signalossomo COP9 , Movimento Celular/genética , Movimento Celular/fisiologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Cerebelo/metabolismo , Edema/complicações , Edema/genética , Exoma/genética , Edição de Genes , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Microcefalia/complicações , Microcefalia/genética , Mutação de Sentido Incorreto/genética , Mutação de Sentido Incorreto/fisiologia , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Análise de Sequência de DNA , Fatores de Transcrição/biossíntese , Peixe-Zebra
10.
Neuropediatrics ; 49(4): 256-261, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29801191

RESUMO

Alexander disease (AxD) is a genetic leukodystrophy caused by GFAP mutations leading to astrocyte dysfunction. Neonatal AxD is a rare phenotype with onset in the first month of life. The proband, belonging to a large pedigree with dominantly inherited benign familial neonatal epilepsy (BFNE), had a phenotype distinct from the rest of the family, with hypotonia and macrocephaly in addition to drug-resistant neonatal seizures. The patient deteriorated and passed away at 6 weeks of age. The pathological and neuroimaging data were consistent with the diagnosis of AxD. Genetic analysis of the proband identified a novel de novo GFAP missense mutation and a KCNQ2 splice site mutation segregating with the BFNE phenotype in the family. The GFAP mutation was located in the coil 2B region of GFAP protein, similar to most neonatal-onset AxD cases with an early death. The clinical and neuroradiological features of the previously published neonatal AxD patients are presented. This study further supports the classification of neonatal-onset AxD as a distinct phenotype based on the age of onset.


Assuntos
Doença de Alexander/genética , Proteína Glial Fibrilar Ácida/genética , Mutação , Doença de Alexander/diagnóstico por imagem , Doença de Alexander/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Evolução Fatal , Humanos , Lactente , Masculino , Fenótipo
11.
Hum Mol Genet ; 24(20): 5677-86, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26188006

RESUMO

Essential tremor (ET) is a common movement disorder with an estimated prevalence of 5% of the population aged over 65 years. In spite of intensive efforts, the genetic architecture of ET remains unknown. We used a combination of whole-exome sequencing and targeted resequencing in three ET families. In vitro and in vivo experiments in oligodendrocyte precursor cells and zebrafish were performed to test our findings. Whole-exome sequencing revealed a missense mutation in TENM4 segregating in an autosomal-dominant fashion in an ET family. Subsequent targeted resequencing of TENM4 led to the discovery of two novel missense mutations. Not only did these two mutations segregate with ET in two additional families, but we also observed significant over transmission of pathogenic TENM4 alleles across the three families. Consistent with a dominant mode of inheritance, in vitro analysis in oligodendrocyte precursor cells showed that mutant proteins mislocalize. Finally, expression of human mRNA harboring any of three patient mutations in zebrafish embryos induced defects in axon guidance, confirming a dominant-negative mode of action for these mutations. Our genetic and functional data, which is corroborated by the existence of a Tenm4 knockout mouse displaying an ET phenotype, implicates TENM4 in ET. Together with previous studies of TENM4 in model organisms, our studies intimate that processes regulating myelination in the central nervous system and axon guidance might be significant contributors to the genetic burden of this disorder.


Assuntos
Axônios/patologia , Tremor Essencial/genética , Glicoproteínas de Membrana/genética , Mutação de Sentido Incorreto , Oligodendroglia/patologia , Adulto , Animais , Análise Mutacional de DNA , Tremor Essencial/metabolismo , Tremor Essencial/fisiopatologia , Exoma , Feminino , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Linhagem , Transporte Proteico , Adulto Jovem , Peixe-Zebra/metabolismo
12.
Am J Hum Genet ; 95(1): 85-95, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24995868

RESUMO

Restless legs syndrome (RLS) is a common neurologic condition characterized by nocturnal dysesthesias and an urge to move, affecting the legs. RLS is a complex trait, for which genome-wide association studies (GWASs) have identified common susceptibility alleles of modest (OR 1.2-1.7) risk at six genomic loci. Among these, variants in MEIS1 have emerged as the largest risk factors for RLS, suggesting that perturbations in this transcription factor might be causally related to RLS susceptibility. To establish this causality, direction of effect, and total genetic burden of MEIS1, we interrogated 188 case subjects and 182 control subjects for rare alleles not captured by previous GWASs, followed by genotyping of ∼3,000 case subjects and 3,000 control subjects, and concluded with systematic functionalization of all discovered variants using a previously established in vivo model of neurogenesis. We observed a significant excess of rare MEIS1 variants in individuals with RLS. Subsequent assessment of all nonsynonymous variants by in vivo complementation revealed an excess of loss-of-function alleles in individuals with RLS. Strikingly, these alleles compromised the function of the canonical MEIS1 splice isoform but were irrelevant to an isoform known to utilize an alternative 3' sequence. Our data link MEIS1 loss of function to the etiopathology of RLS, highlight how combined sequencing and systematic functional annotation of rare variation at GWAS loci can detect risk burden, and offer a plausible explanation for the specificity of phenotypic expressivity of loss-of-function alleles at a locus broadly necessary for neurogenesis and neurodevelopment.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Neoplasias/genética , Síndrome das Pernas Inquietas/genética , Animais , Teste de Complementação Genética , Genótipo , Humanos , Hibridização In Situ , Espectrometria de Massas , Proteína Meis1 , Peixe-Zebra/embriologia
13.
Proc Natl Acad Sci U S A ; 111(11): 4197-202, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24591628

RESUMO

Signaling through the store-operated Ca(2+) release-activated Ca(2+) (CRAC) channel regulates critical cellular functions, including gene expression, cell growth and differentiation, and Ca(2+) homeostasis. Loss-of-function mutations in the CRAC channel pore-forming protein ORAI1 or the Ca(2+) sensing protein stromal interaction molecule 1 (STIM1) result in severe immune dysfunction and nonprogressive myopathy. Here, we identify gain-of-function mutations in the cytoplasmic domain of STIM1 (p.R304W) associated with thrombocytopenia, bleeding diathesis, miosis, and tubular myopathy in patients with Stormorken syndrome, and in ORAI1 (p.P245L), associated with a Stormorken-like syndrome of congenital miosis and tubular aggregate myopathy but without hematological abnormalities. Heterologous expression of STIM1 p.R304W results in constitutive activation of the CRAC channel in vitro, and spontaneous bleeding accompanied by reduced numbers of thrombocytes in zebrafish embryos, recapitulating key aspects of Stormorken syndrome. p.P245L in ORAI1 does not make a constitutively active CRAC channel, but suppresses the slow Ca(2+)-dependent inactivation of the CRAC channel, thus also functioning as a gain-of-function mutation. These data expand our understanding of the phenotypic spectrum of dysregulated CRAC channel signaling, advance our knowledge of the molecular function of the CRAC channel, and suggest new therapies aiming at attenuating store-operated Ca(2+) entry in the treatment of patients with Stormorken syndrome and related pathologic conditions.


Assuntos
Transtornos Plaquetários/genética , Canais de Cálcio/genética , Sinalização do Cálcio/genética , Dislexia/genética , Ictiose/genética , Proteínas de Membrana/genética , Transtornos de Enxaqueca/genética , Miose/genética , Miopatias Congênitas Estruturais/genética , Proteínas de Neoplasias/genética , Baço/anormalidades , Animais , Sequência de Bases , Criança , Primers do DNA/genética , Eritrócitos Anormais , Feminino , Humanos , Dados de Sequência Molecular , Fadiga Muscular/genética , Mutagênese Sítio-Dirigida , Mutação/genética , Proteína ORAI1 , Técnicas de Patch-Clamp , Linhagem , Análise de Sequência de DNA , Molécula 1 de Interação Estromal , Peixe-Zebra
14.
N Engl J Med ; 368(21): 1992-2003, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23656588

RESUMO

BACKGROUND: The combination of ataxia and hypogonadism was first described more than a century ago, but its genetic basis has remained elusive. METHODS: We performed whole-exome sequencing in a patient with ataxia and hypogonadotropic hypogonadism, followed by targeted sequencing of candidate genes in similarly affected patients. Neurologic and reproductive endocrine phenotypes were characterized in detail. The effects of sequence variants and the presence of an epistatic interaction were tested in a zebrafish model. RESULTS: Digenic homozygous mutations in RNF216 and OTUD4, which encode a ubiquitin E3 ligase and a deubiquitinase, respectively, were found in three affected siblings in a consanguineous family. Additional screening identified compound heterozygous truncating mutations in RNF216 in an unrelated patient and single heterozygous deleterious mutations in four other patients. Knockdown of rnf216 or otud4 in zebrafish embryos induced defects in the eye, optic tectum, and cerebellum; combinatorial suppression of both genes exacerbated these phenotypes, which were rescued by nonmutant, but not mutant, human RNF216 or OTUD4 messenger RNA. All patients had progressive ataxia and dementia. Neuronal loss was observed in cerebellar pathways and the hippocampus; surviving hippocampal neurons contained ubiquitin-immunoreactive intranuclear inclusions. Defects were detected at the hypothalamic and pituitary levels of the reproductive endocrine axis. CONCLUSIONS: The syndrome of hypogonadotropic hypogonadism, ataxia, and dementia can be caused by inactivating mutations in RNF216 or by the combination of mutations in RNF216 and OTUD4. These findings link disordered ubiquitination to neurodegeneration and reproductive dysfunction and highlight the power of whole-exome sequencing in combination with functional studies to unveil genetic interactions that cause disease. (Funded by the National Institutes of Health and others.).


Assuntos
Ataxia/genética , Demência/genética , Hipogonadismo/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Animais , Consanguinidade , Exoma , Feminino , Humanos , Masculino , Linhagem , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
16.
J Med Genet ; 49(6): 391-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22693283

RESUMO

BACKGROUND: The progressive myoclonus epilepsies (PMEs) comprise a group of clinically and genetically heterogeneous disorders characterised by myoclonus, epilepsy, and neurological deterioration. This study aimed to identify the underlying gene(s) in childhood onset PME patients with unknown molecular genetic background. METHODS: Homozygosity mapping was applied on genome-wide single nucleotide polymorphism data of 18 Turkish patients. The potassium channel tetramerisation domain-containing 7 (KCTD7) gene, previously associated with PME in a single inbred family, was screened for mutations. The spatiotemporal expression of KCTD7 was assessed in cellular cultures and mouse brain tissue. RESULTS: Overlapping homozygosity in 8/18 patients defined a 1.5 Mb segment on 7q11.21 as the major candidate locus. Screening of the positional candidate gene KCTD7 revealed homozygous missense mutations in two of the eight cases. Screening of KCTD7 in a further 132 PME patients revealed four additional mutations (two missense, one in-frame deletion, and one frameshift-causing) in five families. Eight patients presented with myoclonus and epilepsy and one with ataxia, the mean age of onset being 19 months. Within 2 years after onset, progressive loss of mental and motor skills ensued leading to severe dementia and motor handicap. KCTD7 showed cytosolic localisation and predominant neuronal expression, with widespread expression throughout the brain. None of three polypeptides carrying patient missense mutations affected the subcellular distribution of KCTD7. DISCUSSION: These data confirm the causality of KCTD7 defects in PME, and imply that KCTD7 mutation screening should be considered in PME patients with onset around 2 years of age followed by rapid mental and motor deterioration.


Assuntos
Mutação , Epilepsias Mioclônicas Progressivas/genética , Canais de Potássio/genética , Animais , Western Blotting , Química Encefálica , Células Cultivadas , Mapeamento Cromossômico , Homozigoto , Humanos , Espaço Intracelular , Camundongos , Microscopia de Fluorescência , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Turquia
17.
Hum Mutat ; 33(1): 42-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21990111

RESUMO

The neuronal ceroid lipofuscinoses (NCLs) are clinically and genetically heterogeneous neurodegenerative disorders. Most are autosomal recessively inherited. Clinical features include a variable age of onset, motor and mental decline, epilepsy, visual loss, and premature death. Mutations in eight genes (PPT1/CLN1, TPP1/CLN2, CLN3, CLN5, CLN6, MFSD8/CLN7, CLN8) have been identified and several more are predicted to exist, including two provisionally named CLN4 and CLN9. Despite excessive in vitro and in vivo studies, the precise functions of the NCL proteins and the disease mechanisms remain elusive. To date 365 NCL-causing mutations are known, with 91 novel disease-causing mutations reported. These are reviewed with an emphasis on their complex correlation to phenotypes. Different mutations within the NCL spectrum can cause variable disease severity. The NCLs exemplify both phenotypic convergence or mimicry and phenotypic divergence. For example, mutations in CLN5, CLN6, MFSD8, or CLN8 can underlie the clinically similar late infantile variant NCL disease. Phenotypic divergence is exemplified by different CLN8 mutations giving rise to two very different diseases, the mild CLN8 disease, EPMR (progressive epilepsy with mental retardation), and the more severe CLN8 disease, late infantile variant. The increase in the genetic understanding of the NCLs has led to improved diagnostic approaches, and the recent proposal of a new nomenclature.


Assuntos
Cegueira/genética , Epilepsia/genética , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , Mutação , Lipofuscinoses Ceroides Neuronais/genética , Adolescente , Adulto , Cegueira/diagnóstico , Cegueira/metabolismo , Epilepsia/diagnóstico , Epilepsia/metabolismo , Éxons , Variação Genética , Genótipo , Humanos , Lactente , Íntrons , Glicoproteínas de Membrana/isolamento & purificação , Chaperonas Moleculares/isolamento & purificação , Mortalidade Prematura , Lipofuscinoses Ceroides Neuronais/diagnóstico , Lipofuscinoses Ceroides Neuronais/epidemiologia , Lipofuscinoses Ceroides Neuronais/metabolismo , Fenótipo , Índice de Gravidade de Doença , Tripeptidil-Peptidase 1
18.
Brain ; 132(Pt 3): 810-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19201763

RESUMO

The neuronal ceroid lipofuscinoses (NCLs), the most common neurodegenerative disorders of childhood, are characterized by the accumulation of autofluorescent storage material mainly in neurons. Although clinically rather uniform, variant late-infantile onset NCL (vLINCL) is genetically heterogeneous with four major underlying genes identified so far. We evaluated the genetic background underlying vLINCL in 119 patients, and specifically analysed the recently reported CLN7/MFSD8 gene for mutations in 80 patients. Clinical data were collected from the CLN7/MFSD8 mutation positive patients. Eight novel CLN7/MFSD8 mutations and seven novel mutations in the CLN1/PPT1, CLN2/TPP1, CLN5, CLN6 and CLN8 genes were identified in patients of various ethnic origins. A significant group of Roma patients originating from the former Czechoslovakia was shown to bear the c.881C>A (p.Thr294Lys) mutation in CLN7/MFSD8, possibly due to a founder effect. With one exception, the CLN7/MFSD8 mutation positive patients present a phenotype indistinguishable from the other vLINCL forms. In one patient with an in-frame amino acid substitution mutation in CLN7/MFSD8, the disease onset was later and the disease course less aggressive than in variant late-infantile NCL. Our findings raise the total number of CLN7/MFSD8 mutations to 14 with the majority of families having private mutations. Our study confirms that CLN7/MFSD8 defects are not restricted to the Turkish population, as initially anticipated, but are a relatively common cause of NCL in different populations. CLN7/MFSD8 should be considered a diagnostic alternative not only in variant late-infantile but also later onset NCL forms with a more protracted disease course. A significant number of NCL patients in Turkey exist, in which the underlying genetic defect remains to be determined.


Assuntos
Proteínas de Membrana Transportadoras/genética , Mutação , Lipofuscinoses Ceroides Neuronais/genética , Idade de Início , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA/métodos , Feminino , Haplótipos , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Tripeptidil-Peptidase 1
19.
Nat Commun ; 11(1): 5903, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33214552

RESUMO

The neuronal primary cilium and centriolar satellites have functions in neurogenesis, but little is known about their roles in the postnatal brain. We show that ablation of pericentriolar material 1 in the mouse leads to progressive ciliary, anatomical, psychomotor, and cognitive abnormalities. RNAseq reveals changes in amine- and G-protein coupled receptor pathways. The physiological relevance of this phenotype is supported by decreased available dopamine D2 receptor (D2R) levels and the failure of antipsychotic drugs to rescue adult behavioral defects. Immunoprecipitations show an association with Pcm1 and D2Rs. Finally, we sequence PCM1 in two human cohorts with severe schizophrenia. Systematic modeling of all discovered rare alleles by zebrafish in vivo complementation reveals an enrichment for pathogenic alleles. Our data emphasize a role for the pericentriolar material in the postnatal brain, with progressive degenerative ciliary and behavioral phenotypes; and they support a contributory role for PCM1 in some individuals diagnosed with schizophrenia.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Cílios/patologia , Predisposição Genética para Doença/genética , Esquizofrenia/genética , Adulto , Idoso , Alelos , Aminas/metabolismo , Animais , Antipsicóticos/uso terapêutico , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Resistência a Medicamentos/genética , Humanos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Fenótipo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Esquizofrenia/tratamento farmacológico , Esquizofrenia/patologia , Esquizofrenia/fisiopatologia , Transdução de Sinais , Adulto Jovem , Peixe-Zebra
20.
Nat Genet ; 52(11): 1145-1150, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33046855

RESUMO

The influence of genetic background on driver mutations is well established; however, the mechanisms by which the background interacts with Mendelian loci remain unclear. We performed a systematic secondary-variant burden analysis of two independent cohorts of patients with Bardet-Biedl syndrome (BBS) with known recessive biallelic pathogenic mutations in one of 17 BBS genes for each individual. We observed a significant enrichment of trans-acting rare nonsynonymous secondary variants in patients with BBS compared with either population controls or a cohort of individuals with a non-BBS diagnosis and recessive variants in the same gene set. Strikingly, we found a significant over-representation of secondary alleles in chaperonin-encoding genes-a finding corroborated by the observation of epistatic interactions involving this complex in vivo. These data indicate a complex genetic architecture for BBS that informs the biological properties of disease modules and presents a model for secondary-variant burden analysis in recessive disorders.


Assuntos
Síndrome de Bardet-Biedl/genética , Variação Genética , Alelos , Estudos de Coortes , Exoma , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA