Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Neurol ; 24(1): 119, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38605286

RESUMO

BACKGROUND: Ischemic stroke in young adults can be caused by a variety of etiologies including the monogenic disorders. Visceral heterotaxy is a condition caused by abnormal left-right determinations during embryonic development. We aimed to determine the cause of a young ischemic stroke patient with visceral heterotaxy. CASE PRESENTATION: We performed neurological, radiological, and genetic evaluations in a 17-year-old male patient presenting ischemic stroke and visceral heterotaxy to determine the underlying cause of this rare disease combination. Brain magnetic resonance imaging (MRI) showed evidence of embolic stroke, abdominal computed tomography (CT) showed visceral heterotaxy, and echocardiogram showed cardiac anomaly with right-to-left-shunt (RLS). Whole genome sequencing (WGS) revealed a heterozygous missense variant (NM_018055.5: c.1016 T > C, p.(Met339Val)) in the NODAL gene, which is essential to the determination of the left-right body axis. CONCLUSIONS: Our study highlights the importance of evaluating genetic etiology in young ischemic stroke and the need for stroke risk management in visceral heterotaxy patients with RLS. To the best of our knowledge, we report the first genetically-confirmed case of visceral heterotaxy with young embolic stroke reported to date.


Assuntos
AVC Embólico , Síndrome de Heterotaxia , Adolescente , Humanos , Masculino , Anormalidades Cardiovasculares , Síndrome de Heterotaxia/genética , Imageamento por Ressonância Magnética , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/genética
2.
Hum Mol Genet ; 30(7): 552-563, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33693705

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is an inherited muscle disease caused by misexpression of the DUX4 gene in skeletal muscle. DUX4 is a transcription factor, which is normally expressed in the cleavage-stage embryo and regulates gene expression involved in early embryonic development. Recent studies revealed that DUX4 also activates the transcription of repetitive elements such as endogenous retroviruses (ERVs), mammalian apparent long terminal repeat (LTR)-retrotransposons and pericentromeric satellite repeats (Human Satellite II). DUX4-bound ERV sequences also create alternative promoters for genes or long non-coding RNAs, producing fusion transcripts. To further understand transcriptional regulation by DUX4, we performed nanopore long-read direct RNA sequencing (dRNA-seq) of human muscle cells induced by DUX4, because long reads show whole isoforms with greater confidence. We successfully detected differential expression of known DUX4-induced genes and discovered 61 differentially expressed repeat loci, which are near DUX4-ChIP peaks. We also identified 247 gene-ERV fusion transcripts, of which 216 were not reported previously. In addition, long-read dRNA-seq clearly shows that RNA splicing is a common event in DUX4-activated ERV transcripts. Long-read analysis showed non-LTR transposons including Alu elements are also transcribed from LTRs. Our findings revealed further complexity of DUX4-induced ERV transcripts. This catalogue of DUX4-activated repetitive elements may provide useful information to elucidate the pathology of FSHD. Also, our results indicate that nanopore dRNA-seq has complementary strengths to conventional short-read complementary DNA sequencing.


Assuntos
Proteínas de Homeodomínio/genética , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Nanoporos , Sequências Repetitivas de Ácido Nucleico/genética , Análise de Sequência de RNA/métodos , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Células Musculares/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Isoformas de Proteínas/genética , Isoformas de RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA/estatística & dados numéricos
3.
Am J Hum Genet ; 106(4): 549-558, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32169168

RESUMO

De novo variants (DNVs) cause many genetic diseases. When DNVs are examined in the whole coding regions of genes in next-generation sequencing analyses, pathogenic DNVs often cluster in a specific region. One such region is the last exon and the last 50 bp of the penultimate exon, where truncating DNVs cause escape from nonsense-mediated mRNA decay [NMD(-) region]. Such variants can have dominant-negative or gain-of-function effects. Here, we first developed a resource of rates of truncating DNVs in NMD(-) regions under the null model of DNVs. Utilizing this resource, we performed enrichment analysis of truncating DNVs in NMD(-) regions in 346 developmental and epileptic encephalopathy (DEE) trios. We observed statistically significant enrichment of truncating DNVs in semaphorin 6B (SEMA6B) (p value: 2.8 × 10-8; exome-wide threshold: 2.5 × 10-6). The initial analysis of the 346 individuals and additional screening of 1,406 and 4,293 independent individuals affected by DEE and developmental disorders collectively identified four truncating DNVs in the SEMA6B NMD(-) region in five individuals who came from unrelated families (p value: 1.9 × 10-13) and consistently showed progressive myoclonic epilepsy. RNA analysis of lymphoblastoid cells established from an affected individual showed that the mutant allele escaped NMD, indicating stable production of the truncated protein. Importantly, heterozygous truncating variants in the NMD(+) region of SEMA6B are observed in general populations, and SEMA6B is most likely loss-of-function tolerant. Zebrafish expressing truncating variants in the NMD(-) region of SEMA6B orthologs displayed defective development of brain neurons and enhanced pentylenetetrazole-induced seizure behavior. In summary, we show that truncating DNVs in the final exon of SEMA6B cause progressive myoclonic epilepsy.


Assuntos
Exoma/genética , Éxons/genética , Predisposição Genética para Doença/genética , Variação Genética/genética , Epilepsias Mioclônicas Progressivas/genética , Semaforinas/genética , Adolescente , Adulto , Alelos , Animais , Feminino , Heterozigoto , Humanos , Masculino , Degradação do RNAm Mediada por Códon sem Sentido/genética , Convulsões/genética , Adulto Jovem , Peixe-Zebra/genética
4.
Am J Hum Genet ; 106(1): 13-25, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31839203

RESUMO

MN1 was originally identified as a tumor-suppressor gene. Knockout mouse studies have suggested that Mn1 is associated with craniofacial development. However, no MN1-related phenotypes have been established in humans. Here, we report on three individuals who have de novo MN1 variants that lead to a protein lacking the carboxyl (C) terminus and who presented with severe developmental delay, craniofacial abnormalities with specific facial features, and structural abnormalities in the brain. An in vitro study revealed that the deletion of the C-terminal region led to increased protein stability, an inhibitory effect on cell proliferation, and enhanced MN1 aggregation in nuclei compared to what occurred in the wild type, suggesting that a gain-of-function mechanism is involved in this disease. Considering that C-terminal deletion increases the fraction of intrinsically disordered regions of MN1, it is possible that altered phase separation could be involved in the mechanism underlying the disease. Our data indicate that MN1 participates in transcriptional regulation of target genes through interaction with the transcription factors PBX1, PKNOX1, and ZBTB24 and that mutant MN1 impairs the binding with ZBTB24 and RING1, which is an E3 ubiquitin ligase. On the basis of our findings, we propose the model that C-terminal deletion interferes with MN1's interaction molecules related to the ubiquitin-mediated proteasome pathway, including RING1, and increases the amount of the mutant protein; this increase leads to the dysregulation of MN1 target genes by inhibiting rapid MN1 protein turnover.


Assuntos
Encefalopatias/etiologia , Anormalidades Craniofaciais/etiologia , Mutação com Ganho de Função , Regulação da Expressão Gênica , Deleção de Sequência , Transativadores/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Encefalopatias/patologia , Proliferação de Células , Criança , Pré-Escolar , Anormalidades Craniofaciais/patologia , Feminino , Células HeLa , Humanos , Masculino , Proteólise , Síndrome , Transativadores/metabolismo , Transcriptoma , Proteínas Supressoras de Tumor/metabolismo
5.
J Hum Genet ; 67(12): 739-742, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35945270

RESUMO

In genetic testing of Mendelian diseases, it is a bioinformatics challenge to effectively prioritize disease-causing candidate genes listed from massively parallel sequencing. Tissue specificity of the gene expression levels may give a clue because it may reflect tissue-specific disease manifestation. However, considering poor correlations between mRNA and protein expression in some genes, it is not clear whether transcriptomics- or proteomics-based tissue specificity should be used to prioritize candidate genes. Therefore, we compared the efficiency of tissue-specific scores (TS scores) obtained from transcriptome and proteome data in prioritizing candidate genes for whole exome sequencing (WES) analysis of Mendelian disease patients. We show that both Protein and RNA TS scores are useful in prioritizing candidate genes in WES analysis, although diseases like coagulopathies get more benefit from Protein TS score. This study may provide useful evidence in developing new methods to effectively identify novel disease-causing genes.


Assuntos
Exoma , Testes Genéticos , Humanos , Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Expressão Gênica
6.
Brain ; 144(4): 1103-1117, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33791773

RESUMO

A pentanucleotide TTTCA repeat insertion into a polymorphic TTTTA repeat element in SAMD12 causes benign adult familial myoclonic epilepsy. Although the precise determination of the entire SAMD12 repeat sequence is important for molecular diagnosis and research, obtaining this sequence remains challenging when using conventional genomic/genetic methods, and even short-read and long-read next-generation sequencing technologies have been insufficient. Incomplete information regarding expanded repeat sequences may hamper our understanding of the pathogenic roles played by varying numbers of repeat units, genotype-phenotype correlations, and mutational mechanisms. Here, we report a new approach for the precise determination of the entire expanded repeat sequence and present a workflow designed to improve the diagnostic rates in various repeat expansion diseases. We examined 34 clinically diagnosed benign adult familial myoclonic epilepsy patients, from 29 families using repeat-primed PCR, Southern blot, and long-read sequencing with Cas9-mediated enrichment. Two cases with questionable results from repeat-primed PCR and/or Southern blot were confirmed as pathogenic using long-read sequencing with Cas9-mediated enrichment, resulting in the identification of pathogenic SAMD12 repeat expansions in 76% of examined families (22/29). Importantly, long-read sequencing with Cas9-mediated enrichment was able to provide detailed information regarding the sizes, configurations, and compositions of the expanded repeats. The inserted TTTCA repeat size and the proportion of TTTCA sequences among the overall repeat sequences were highly variable, and a novel repeat configuration was identified. A genotype-phenotype correlation study suggested that the insertion of even short (TTTCA)14 repeats contributed to the development of benign adult familial myoclonic epilepsy. However, the sizes of the overall TTTTA and TTTCA repeat units are also likely to be involved in the pathology of benign adult familial myoclonic epilepsy. Seven unsolved SAMD12-negative cases were investigated using whole-genome long-read sequencing, and infrequent, disease-associated, repeat expansions were identified in two cases. The strategic workflow resolved two questionable SAMD12-positive cases and two previously SAMD12-negative cases, increasing the diagnostic yield from 69% (20/29 families) to 83% (24/29 families). This study indicates the significant utility of long-read sequencing technologies to explore the pathogenic contributions made by various repeat units in complex repeat expansions and to improve the overall diagnostic rate.


Assuntos
Expansão das Repetições de DNA/genética , Epilepsias Mioclônicas/genética , Proteínas do Tecido Nervoso/genética , Análise de Sequência de DNA/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Feminino , Estudos de Associação Genética , Humanos , Masculino , Repetições de Microssatélites , Pessoa de Meia-Idade
7.
Hum Mol Genet ; 28(14): 2319-2329, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30985895

RESUMO

Disorders of sex development (DSDs) are defined as congenital conditions in which chromosomal, gonadal or anatomical sex is atypical. In many DSD cases, genetic causes remain to be elucidated. Here, we performed a case-control exome sequencing study comparing gene-based burdens of rare damaging variants between 26 DSD cases and 2625 controls. We found exome-wide significant enrichment of rare heterozygous truncating variants in the MYRF gene encoding myelin regulatory factor, a transcription factor essential for oligodendrocyte development. All three variants occurred de novo. We identified an additional 46,XY DSD case of a de novo damaging missense variant in an independent cohort. The clinical symptoms included hypoplasia of Müllerian derivatives and ovaries in 46,XX DSD patients, defective development of Sertoli and Leydig cells in 46,XY DSD patients and congenital diaphragmatic hernia in one 46,XY DSD patient. As all of these cells and tissues are or partly consist of coelomic epithelium (CE)-derived cells (CEDC) and CEDC developed from CE via proliferaiton and migration, MYRF might be related to these processes. Consistent with this hypothesis, single-cell RNA sequencing of foetal gonads revealed high expression of MYRF in CE and CEDC. Reanalysis of public chromatin immunoprecipitation sequencing data for rat Myrf showed that genes regulating proliferation and migration were enriched among putative target genes of Myrf. These results suggested that MYRF is a novel causative gene of 46,XY and 46,XX DSD and MYRF is a transcription factor regulating CD and/or CEDC proliferation and migration, which is essential for development of multiple organs.


Assuntos
Transtornos 46, XX do Desenvolvimento Sexual/genética , Transtorno 46,XY do Desenvolvimento Sexual/genética , Proteínas de Membrana/genética , Fatores de Transcrição/genética , Transtornos 46, XX do Desenvolvimento Sexual/patologia , Adolescente , Estudos de Casos e Controles , Movimento Celular , Proliferação de Células , Pré-Escolar , Estudos de Coortes , Biologia Computacional , Transtorno 46,XY do Desenvolvimento Sexual/patologia , Feminino , Ontologia Genética , Gônadas/crescimento & desenvolvimento , Haploinsuficiência , Humanos , Masculino , Proteínas de Membrana/metabolismo , Mutação , Mutação de Sentido Incorreto , Análise de Célula Única , Fatores de Transcrição/metabolismo , Sequenciamento do Exoma , Adulto Jovem
8.
J Hum Genet ; 66(7): 697-705, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33510365

RESUMO

Whole-exome sequencing (WES) can detect not only single-nucleotide variants in causal genes, but also pathogenic copy-number variations using several methods. However, there may be overlooked pathogenic variations in the out of target genome regions of WES analysis (e.g., promoters), leaving many patients undiagnosed. Whole-genome sequencing (WGS) can potentially analyze such regions. We applied long-read nanopore WGS and our recently developed analysis pipeline "dnarrange" to a patient who was undiagnosed by trio-based WES analysis, and identified a heterozygous 97-kb deletion partially involving 5'-untranslated exons of MBD5, which was outside the WES target regions. The phenotype of the patient, a 32-year-old male, was consistent with haploinsufficiency of MBD5. The transcript level of MBD5 in the patient's lymphoblastoid cells was reduced. We therefore concluded that the partial MBD5 deletion is the culprit for this patient. Furthermore, we found other rare structural variations (SVs) in this patient, i.e., a large inversion and a retrotransposon insertion, which were not seen in 33 controls. Although we considered that they are benign SVs, this finding suggests that our pipeline using long-read WGS is useful for investigating various types of potentially pathogenic SVs. In conclusion, we identified a 97-kb deletion, which causes haploinsufficiency of MBD5 in a patient with neurodevelopmental disorder, demonstrating that long-read WGS is a powerful technique to discover pathogenic SVs.


Assuntos
Proteínas de Ligação a DNA/genética , Predisposição Genética para Doença , Transtornos do Neurodesenvolvimento/genética , Adulto , Exoma/genética , Haploinsuficiência/genética , Humanos , Masculino , Mutagênese Insercional/genética , Transtornos do Neurodesenvolvimento/patologia , Retroelementos/genética , Sequenciamento Completo do Genoma
9.
J Hum Genet ; 66(5): 499-507, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33144663

RESUMO

The objective of this study was to evaluate the efficacy of whole exome sequencing (WES) for the genetic diagnosis of cases presenting with fetal structural anomalies detected by ultrasonography. WES was performed on 19 cases with prenatal structural anomalies. Genomic DNA was extracted from umbilical cords or umbilical blood obtained shortly after birth. WES data were analyzed on prenatal phenotypes alone, and the data were re-analyzed after information regarding the postnatal phenotype was obtained. Based solely on the fetal phenotype, pathogenic, or likely pathogenic, single nucleotide variants were identified in 5 of 19 (26.3%) cases. Moreover, we detected trisomy 21 in two cases by WES-based copy number variation analysis. The overall diagnostic rate was 36.8% (7/19). They were all compatible with respective fetal structural anomalies. By referring to postnatal phenotype information, another candidate variant was identified by a postnatal clinical feature that was not detected in prenatal screening. As detailed phenotyping is desirable for better diagnostic rates in WES analysis, we should be aware that fetal phenotype is a useful, but sometimes limited source of information for comprehensive genetic analysis. It is important to amass more data of genotype-phenotype correlations, especially to appropriately assess the validity of WES in prenatal settings.


Assuntos
Anormalidades Congênitas/genética , Sequenciamento do Exoma , Feto/anormalidades , Ultrassonografia Pré-Natal , Aborto Eugênico , Adulto , Cesárea , Anormalidades Congênitas/diagnóstico por imagem , Anormalidades Congênitas/embriologia , DNA/sangue , DNA/genética , Variações do Número de Cópias de DNA , Síndrome de Down/diagnóstico por imagem , Síndrome de Down/embriologia , Síndrome de Down/genética , Feminino , Sangue Fetal/química , Morte Fetal/etiologia , Idade Gestacional , Humanos , Leucócitos/química , Leucócitos/ultraestrutura , Polimorfismo de Nucleotídeo Único , Gravidez , Resultado da Gravidez
10.
J Hum Genet ; 66(4): 401-407, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33040083

RESUMO

Pontocerebellar hypoplasia (PCH) is currently classified into 13 subgroups and many gene variants associated with PCH have been identified by next generation sequencing. PCH type 1 is a rare heterogeneous neurodegenerative disorder. The clinical presentation includes early-onset severe developmental delay, progressive motor neuronopathy, and cerebellar and pontine atrophy. Recently two variants in the EXOSC9 gene (MIM: 606180), NM_001034194.1: c.41T>C (p.Leu14Pro) and c.481C>T (p.Arg161*) were identified in four unrelated patients with PCH type 1D (PCH1D) (MIM: 618065). EXOSC9 encodes a component of the exosome complex, which is essential for correct processing and degradation of RNA. We report here two PCH1D families with biallelic EXOSC9 variants: c.239T>G (p.Leu80Arg) and c.484dupA (p.Arg162Lysfs*3) in one family and c.151G>C (p.Gly51Arg) in the other family. Although the patients studied here showed similar clinical features as previously described for PCH1D, relatively greater intellectual development (although still highly restricted) and normal pontine structure were recognized. Our findings expand the clinical consequences of biallelic EXOSC9 variants.


Assuntos
Atrofia/patologia , Doenças Cerebelares/patologia , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Doença dos Neurônios Motores/patologia , Atrofia Muscular Espinal/patologia , Mutação , Atrofias Olivopontocerebelares/patologia , Proteínas de Ligação a RNA/genética , Atrofia/complicações , Atrofia/genética , Doenças Cerebelares/complicações , Doenças Cerebelares/genética , Feminino , Estudos de Associação Genética , Humanos , Lactente , Masculino , Doença dos Neurônios Motores/complicações , Doença dos Neurônios Motores/genética , Atrofia Muscular Espinal/complicações , Atrofia Muscular Espinal/genética , Atrofias Olivopontocerebelares/complicações , Atrofias Olivopontocerebelares/genética , Linhagem
11.
Int J Mol Sci ; 22(9)2021 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-33925887

RESUMO

(1) Background: The ERVPb1 gene in humans is derived from an envelope (Env) gene of a human endogenous retrovirus group, HERV-P(b). The ERVPb1 gene reportedly has a conserved open reading frame (ORF) in Old World monkeys. Although its forced expression led to cell-fusion in an ex vivo cell culture system, like other Env-derived genes such as syncytin-1 and -2, its mRNA expression is not placenta-specific, but almost ubiquitous, albeit being quite low in human tissues and organs, implying a distinct role for ERVPb1. (2) Methods: To elucidate the cell lineage(s) in which the ERVPb1 protein is translated in human development, we developed a novel, highly sensitive system for detecting HERV-derived proteins/peptides expressed in the tissue differentiation process of human induced pluripotent stem cells (iPSCs). (3) Results: We first determined that ERVPb1 is also conserved in New World monkeys. Then, we showed that the ERVPb1 protein is translated from a uniquely spliced ERVPb1 transcript in hematopoietic cell lineages, including a subset of macrophages, and further showed that its mRNA expression is upregulated by lipopolysaccharide (LPS) stimulation in primary human monocytes. (4) Conclusions: ERVPb1 is unique to Simiiformes and actually translated in hematopoietic cell lineages, including a subset of macrophages.


Assuntos
Retrovirus Endógenos , Haplorrinos/virologia , Macrófagos/virologia , Animais , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem Celular , Retrovirus Endógenos/genética , Retrovirus Endógenos/isolamento & purificação , Retrovirus Endógenos/metabolismo , Corantes Fluorescentes , Edição de Genes/métodos , Genes Virais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Macrófagos/metabolismo , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
12.
Hum Mol Genet ; 27(23): 4024-4035, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30107443

RESUMO

Double homeobox 4 (DUX4), the causative gene of facioscapulohumeral muscular dystrophy (FSHD), is ectopically expressed in the skeletal muscle cells of FSHD patients because of chromatin relaxation at 4q35. The diminished heterochromatic state at 4q35 is caused by either large genome contractions [FSHD type 1 (FSHD1)] or mutations in genes encoding chromatin regulators, such as SMCHD1 [FSHD type 2 (FSHD2)]. However, the mechanism by which DUX4 expression is regulated remains largely unknown. Here, using a myocyte model developed from patient-derived induced pluripotent stem cells, we determined that DUX4 expression was increased by oxidative stress (OS), a common environmental stress in skeletal muscle, in both FSHD1 and FSHD2 myocytes. We generated FSHD2-derived isogenic control clones with SMCHD1 mutation corrected by clustered regularly interspaced short palindromic repeats (CRISPR)/ CRISPR associated 9 (Cas9) and homologous recombination and found in the myocytes obtained from these clones that DUX4 basal expression and the OS-induced upregulation were markedly suppressed due to an increase in the heterochromatic state at 4q35. We further found that DNA damage response (DDR) was involved in OS-induced DUX4 increase and identified ataxia-telangiectasia mutated, a DDR regulator, as a mediator of this effect. Our results suggest that the relaxed chromatin state in FSHD muscle cells permits aberrant access of OS-induced DDR signaling, thus increasing DUX4 expression. These results suggest OS could represent an environmental risk factor that promotes FSHD progression.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Sistemas CRISPR-Cas/genética , Cromatina/genética , Cromossomos Humanos Par 4/genética , Dano ao DNA/genética , Regulação da Expressão Gênica , Humanos , Células Musculares/metabolismo , Células Musculares/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Mutação , Estresse Oxidativo/genética
13.
Hum Mol Genet ; 27(8): 1421-1433, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29432562

RESUMO

Calcineurin is a calcium (Ca2+)/calmodulin-regulated protein phosphatase that mediates Ca2+-dependent signal transduction. Here, we report six heterozygous mutations in a gene encoding the alpha isoform of the calcineurin catalytic subunit (PPP3CA). Notably, mutations were observed in different functional domains: in addition to three catalytic domain mutations, two missense mutations were found in the auto-inhibitory (AI) domain. One additional frameshift insertion that caused premature termination was also identified. Detailed clinical evaluation of the six individuals revealed clinically unexpected consequences of the PPP3CA mutations. First, the catalytic domain mutations and frameshift mutation were consistently found in patients with nonsyndromic early onset epileptic encephalopathy. In contrast, the AI domain mutations were associated with multiple congenital abnormalities including craniofacial dysmorphism, arthrogryposis and short stature. In addition, one individual showed severe skeletal developmental defects, namely, severe craniosynostosis and gracile bones (severe bone slenderness and perinatal fractures). Using a yeast model system, we showed that the catalytic and AI domain mutations visibly result in decreased and increased calcineurin signaling, respectively. These findings indicate that different functional effects of PPP3CA mutations are associated with two distinct disorders and suggest that functional approaches using a simple cellular system provide a tool for resolving complex genotype-phenotype correlations.


Assuntos
Artrogripose/genética , Calcineurina/genética , Anormalidades Craniofaciais/genética , Nanismo/genética , Mutação com Ganho de Função , Mutação com Perda de Função , Espasmos Infantis/genética , Sequência de Aminoácidos , Artrogripose/metabolismo , Artrogripose/patologia , Sequência de Bases , Calcineurina/química , Calcineurina/metabolismo , Criança , Pré-Escolar , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Nanismo/metabolismo , Nanismo/patologia , Feminino , Regulação da Expressão Gênica , Estudos de Associação Genética , Humanos , Masculino , Modelos Moleculares , Linhagem , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Espasmos Infantis/metabolismo , Espasmos Infantis/patologia , Adulto Jovem
14.
Am J Hum Genet ; 100(1): 169-178, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-28017374

RESUMO

Nemaline myopathy (NM) is a common form of congenital nondystrophic skeletal muscle disease characterized by muscular weakness of proximal dominance, hypotonia, and respiratory insufficiency but typically not cardiac dysfunction. Wide variation in severity has been reported. Intranuclear rod myopathy is a subtype of NM in which rod-like bodies are seen in the nucleus, and it often manifests as a severe phenotype. Although ten mutant genes are currently known to be associated with NM, only ACTA1 is associated with intranuclear rod myopathy. In addition, the genetic cause remains unclear in approximately 25%-30% of individuals with NM. We performed whole-exome sequencing on individuals with histologically confirmed but genetically unsolved NM. Our study included individuals with milder, later-onset NM and identified biallelic loss-of-function mutations in myopalladin (MYPN) in four families. Encoded MYPN is a sarcomeric protein exclusively localized in striated muscle in humans. Individuals with identified MYPN mutations in all four of these families have relatively mild, childhood- to adult-onset NM with slowly progressive muscle weakness. Walking difficulties were recognized around their forties. Decreased respiratory function, cardiac involvement, and intranuclear rods in biopsied muscle were observed in two individuals. MYPN was localized at the Z-line in control skeletal muscles but was absent from affected individuals. Homozygous knockin mice with a nonsense mutation in Mypn showed Z-streaming and nemaline-like bodies adjacent to a disorganized Z-line on electron microscopy, recapitulating the disease. Our results suggest that MYPN screening should be considered in individuals with mild NM, especially when cardiac problems or intranuclear rods are present.


Assuntos
Alelos , Progressão da Doença , Proteínas Musculares/genética , Mutação , Miopatias da Nemalina/genética , Adulto , Idade de Início , Animais , Criança , Pré-Escolar , Feminino , Técnicas de Introdução de Genes , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Miopatias da Nemalina/patologia , Linhagem
15.
J Hum Genet ; 65(1): 11-19, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31558760

RESUMO

During the past decade, the search for pathogenic mutations in rare human genetic diseases has involved huge efforts to sequence coding regions, or the entire genome, using massively parallel short-read sequencers. However, the approximate current diagnostic rate is <50% using these approaches, and there remain many rare genetic diseases with unknown cause. There may be many reasons for this, but one plausible explanation is that the responsible mutations are in regions of the genome that are difficult to sequence using conventional technologies (e.g., tandem-repeat expansion or complex chromosomal structural aberrations). Despite the drawbacks of high cost and a shortage of standard analytical methods, several studies have analyzed pathogenic changes in the genome using long-read sequencers. The results of these studies provide hope that further application of long-read sequencers to identify the causative mutations in unsolved genetic diseases may expand our understanding of the human genome and diseases. Such approaches may also be applied to molecular diagnosis and therapeutic strategies for patients with genetic diseases in the future.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Doenças Raras/genética , Análise de Sequência de DNA/métodos , Aberrações Cromossômicas , Elementos de DNA Transponíveis/genética , Genoma Humano , Humanos , Doenças Raras/diagnóstico , Deleção de Sequência/genética , Inversão de Sequência/genética , Sequências de Repetição em Tandem/genética
16.
J Hum Genet ; 65(5): 481-485, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32005903

RESUMO

p21-activated kinases (PAKs) are protein serine/threonine kinases stimulated by Rho-family p21 GTPases such as CDC42 and RAC. PAKs have been implicated in several human disorders, with pathogenic variants in PAK3 associated with intellectual disability and several PAK members, especially PAK1 and PAK4, overexpressed in human cancer. Recently, de novo PAK1 variants were reported to be causative of neurodevelopmental disorder (ND) with secondary macrocephaly in three patients. We herein report a fourth patient with ND, epilepsy, and macrocephaly caused by a de novo PAK1 missense variant. Two previously reported missense PAK1 variants functioned as activating alleles by reducing PAK1 homodimerization. To examine the pathogenicity of the identified novel p.Ser110Thr variant, we carried out in silico structural analysis. Our findings suggest that this variant also prevents PAK1 homodimerization, leading to constitutive PAK1 activation.


Assuntos
Epilepsia , Megalencefalia , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento , Multimerização Proteica , Quinases Ativadas por p21 , Substituição de Aminoácidos , Criança , Ativação Enzimática/genética , Epilepsia/enzimologia , Epilepsia/genética , Humanos , Masculino , Megalencefalia/enzimologia , Megalencefalia/genética , Transtornos do Neurodesenvolvimento/enzimologia , Transtornos do Neurodesenvolvimento/genética , Domínios Proteicos , Quinases Ativadas por p21/química , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
17.
J Hum Genet ; 65(8): 667-674, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32296131

RESUMO

Chromothripsis is a type of chaotic complex genomic rearrangement caused by a single event of chromosomal shattering and repair processes. Chromothripsis is known to cause rare congenital diseases when it occurs in germline cells, however, current genome analysis technologies have difficulty in detecting and deciphering chromothripsis. It is possible that this type of complex rearrangement may be overlooked in rare-disease patients whose genetic diagnosis is unsolved. We applied long read nanopore sequencing and our recently developed analysis pipeline dnarrange to a patient who has a reciprocal chromosomal translocation t(8;18)(q22;q21) as a result of chromothripsis between the two chromosomes, and fully characterize the complex rearrangements at the translocation site. The patient genome was evidently shattered into 19 fragments, and rejoined into derivative chromosomes in a random order and orientation. The reconstructed patient genome indicates loss of five genomic regions, which all overlap with microarray-detected copy number losses. We found that two disease-related genes RAD21 and EXT1 were lost by chromothripsis. These two genes could fully explain the disease phenotype with facial dysmorphisms and bone abnormality, which is likely a contiguous gene syndrome, Cornelia de Lange syndrome type IV (CdLs-4) and atypical Langer-Giedion syndrome (LGS), also known as trichorhinophalangeal syndrome type II (TRPSII). This provides evidence that our approach based on long read sequencing can fully characterize chromothripsis in a patient's genome, which is important for understanding the phenotype of disease caused by complex genomic rearrangement.


Assuntos
Proteínas de Ciclo Celular/genética , Cromotripsia , Proteínas de Ligação a DNA/genética , Síndrome de Cornélia de Lange/genética , Síndrome de Langer-Giedion/genética , N-Acetilglucosaminiltransferases/genética , Criança , Deleção Cromossômica , Síndrome de Cornélia de Lange/diagnóstico , Síndrome de Cornélia de Lange/fisiopatologia , Genoma , Humanos , Síndrome de Langer-Giedion/diagnóstico , Síndrome de Langer-Giedion/fisiopatologia , Masculino , Sequenciamento por Nanoporos , Fenótipo , Análise de Sequência de DNA , Translocação Genética
18.
J Hum Genet ; 65(9): 751-757, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32405030

RESUMO

Inborn errors of metabolism can cause epileptic encephalopathies. Biallelic loss-of-function variants in the ITPA gene, encoding inosine triphosphate pyrophosphatase (ITPase), have been reported in epileptic encephalopathies with lack of myelination of the posterior limb of the internal capsule, brainstem tracts, and tracts to the primary visual and motor cortices (MIM:616647). ITPase plays an important role in purine metabolism. In this study, we identified two novel homozygous ITPA variants, c.264-1 G > A and c.489-1 G > A, in two unrelated consanguineous families. The probands had epilepsy, microcephaly with characteristic magnetic resonance imaging findings (T2 hyperintensity signals in the pyramidal tracts of the internal capsule, delayed myelination, and thin corpus callosum), hypotonia, and developmental delay; both died in early infancy. Our report expands the knowledge of clinical consequences of biallelic ITPA variants.


Assuntos
Encefalopatias/genética , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Predisposição Genética para Doença , Insuficiência de Múltiplos Órgãos/genética , Hipotonia Muscular/genética , Pirofosfatases/genética , Encefalopatias/complicações , Encefalopatias/enzimologia , Encefalopatias/mortalidade , Corpo Caloso/diagnóstico por imagem , Corpo Caloso/patologia , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/enzimologia , Deficiências do Desenvolvimento/mortalidade , Epilepsia/complicações , Epilepsia/enzimologia , Epilepsia/mortalidade , Feminino , Genótipo , Homozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Insuficiência de Múltiplos Órgãos/complicações , Insuficiência de Múltiplos Órgãos/enzimologia , Insuficiência de Múltiplos Órgãos/mortalidade , Hipotonia Muscular/complicações , Hipotonia Muscular/enzimologia , Hipotonia Muscular/mortalidade , Mutação , Linhagem , Tratos Piramidais/diagnóstico por imagem , Tratos Piramidais/patologia , Sequenciamento do Exoma
19.
J Hum Genet ; 65(9): 727-734, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32341456

RESUMO

The ubiquitin-proteasome system is the principal system for protein degradation mediated by ubiquitination and is involved in various cellular processes. Cullin-RING ligases (CRL) are one class of E3 ubiquitin ligases that mediate polyubiquitination of specific target proteins, leading to decomposition of the substrate. Cullin 3 (CUL3) is a member of the Cullin family proteins, which act as scaffolds of CRL. Here we describe three cases of global developmental delays, with or without epilepsy, who had de novo CUL3 variants. One missense variant c.854T>C, p.(Val285Ala) and two frameshift variants c.137delG, p.(Arg46Leufs*32) and c.1239del, p.(Asp413Glufs*42) were identified by whole-exome sequencing. The Val285 residue located in the Cullin N-terminal domain and p.Val285Ala CUL3 mutant showed significantly weaker interactions to the BTB domain proteins than wild-type CUL3. Our findings suggest that de novo CUL3 variants may cause structural instability of the CRL complex and impairment of the ubiquitin-proteasome system, leading to diverse neuropsychiatric disorders.


Assuntos
Proteínas Culina/genética , Proteínas Culina/metabolismo , Deficiências do Desenvolvimento/genética , Predisposição Genética para Doença , Espasmos Infantis/genética , Criança , Pré-Escolar , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Mutação da Fase de Leitura , Células HEK293 , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto , Ligação Proteica , Espasmos Infantis/complicações , Ubiquitina-Proteína Ligases/metabolismo , Sequenciamento do Exoma
20.
J Hum Genet ; 65(9): 811, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32536687

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA