Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 161(5): 1012-1025, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-25959774

RESUMO

Mammalian genomes are organized into megabase-scale topologically associated domains (TADs). We demonstrate that disruption of TADs can rewire long-range regulatory architecture and result in pathogenic phenotypes. We show that distinct human limb malformations are caused by deletions, inversions, or duplications altering the structure of the TAD-spanning WNT6/IHH/EPHA4/PAX3 locus. Using CRISPR/Cas genome editing, we generated mice with corresponding rearrangements. Both in mouse limb tissue and patient-derived fibroblasts, disease-relevant structural changes cause ectopic interactions between promoters and non-coding DNA, and a cluster of limb enhancers normally associated with Epha4 is misplaced relative to TAD boundaries and drives ectopic limb expression of another gene in the locus. This rewiring occurred only if the variant disrupted a CTCF-associated boundary domain. Our results demonstrate the functional importance of TADs for orchestrating gene expression via genome architecture and indicate criteria for predicting the pathogenicity of human structural variants, particularly in non-coding regions of the human genome.


Assuntos
Modelos Animais de Doenças , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Animais , Extremidades/anatomia & histologia , Extremidades/crescimento & desenvolvimento , Humanos , Deformidades Congênitas dos Membros/genética , Camundongos , Regiões Promotoras Genéticas , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Receptor EphA4/genética
2.
Hum Genomics ; 18(1): 23, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38448978

RESUMO

BACKGROUND/OBJECTIVES: Rare genetic disorders causing specific congenital developmental abnormalities often manifest in single families. Investigation of disease-causing molecular features are most times lacking, although these investigations may open novel therapeutic options for patients. In this study, we aimed to identify the genetic cause in an Iranian patient with severe skeletal dysplasia and to model its molecular function in zebrafish embryos. RESULTS: The proband displays short stature and multiple skeletal abnormalities, including mesomelic dysplasia of the arms with complete humero-radio-ulna synostosis, arched clavicles, pelvic dysplasia, short and thin fibulae, proportionally short vertebrae, hyperlordosis and mild kyphosis. Exome sequencing of the patient revealed a novel homozygous c.374G > T, p.(Arg125Leu) missense variant in MSGN1 (NM_001105569). MSGN1, a basic-Helix-Loop-Helix transcription factor, plays a crucial role in formation of presomitic mesoderm progenitor cells/mesodermal stem cells during early developmental processes in vertebrates. Initial in vitro experiments show protein stability and correct intracellular localization of the novel variant in the nucleus and imply retained transcription factor function. To test the pathogenicity of the detected variant, we overexpressed wild-type and mutant msgn1 mRNA in zebrafish embryos and analyzed tbxta (T/brachyury/ntl). Overexpression of wild-type or mutant msgn1 mRNA significantly reduces tbxta expression in the tailbud compared to control embryos. Mutant msgn1 mRNA injected embryos depict a more severe effect, implying a gain-of-function mechanism. In vivo analysis on embryonic development was performed by clonal msgn1 overexpression in zebrafish embryos further demonstrated altered cell compartments in the presomitic mesoderm, notochord and pectoral fin buds. Detection of ectopic tbx6 and bmp2 expression in these embryos hint to affected downstream signals due to Msgn1 gain-of-function. CONCLUSION: In contrast to loss-of-function effects described in animal knockdown models, gain-of-function of MSGN1 explains the only mildly affected axial skeleton of the proband and rather normal vertebrae. In this context we observed notochord bending and potentially disruption of pectoral fin buds/upper extremity after overexpression of msgn1 in zebrafish embryos. The latter might result from Msgn1 function on mesenchymal stem cells or on chondrogenesis in these regions. In addition, we detected ectopic tbx6 and bmp2a expression after gain of Msgn1 function in zebrafish, which are interconnected to short stature, congenital scoliosis, limb shortening and prominent skeletal malformations in patients. Our findings highlight a rare, so far undescribed skeletal dysplasia syndrome associated with a gain-of-function mutation in MSGN1 and hint to its molecular downstream effectors.


Assuntos
Anormalidades Múltiplas , Nanismo , Osteocondrodisplasias , Animais , Feminino , Humanos , Gravidez , Mutação com Ganho de Função , Irã (Geográfico) , RNA Mensageiro , Proteínas com Domínio T/genética , Fatores de Transcrição , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
J Inherit Metab Dis ; 47(4): 805-817, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38618884

RESUMO

Fabry disease (FD) is an X-linked multiorgan disorder caused by variants in the alpha-galactosidase A gene (GLA). Depending on the variant, disease phenotypes range from benign to life-threatening. More than 1000 GLA variants are known, but a link between genotype and phenotype in FD has not yet been established for all. p.A143T, p.D313Y, and p.S126G are frequent examples of variants of unknown significance (VUS). We have investigated the potential pathogenicity of these VUS combining clinical data with data obtained in human cellular in vitro systems. We have analyzed four different male subject-derived cell types for alpha-galactosidase A enzyme (GLA) activity and intracellular Gb3 load. Additionally, Gb3 load in skin tissue as well as clinical data were studied for correlates of disease manifestations. A reduction of GLA activity was observed in cells carrying p.A143T compared with controls (p < 0.05). In cells carrying the p.D313Y variant, a reduced GLA activity was found only in endothelial cells (p < 0.01) compared with controls. No pathological changes were observed in cells carrying the p.S126G variant. None of the VUS investigated caused intracellular Gb3 accumulation in any cell type. Our data of aberrant GLA activity in cells of p.A143T hemizygotes and overall normal cellular phenotypes in cells of p.D313Y and p.S126G hemizygotes contribute a basic science perspective to the clinically highly relevant discussion on VUS in GLA.


Assuntos
Doença de Fabry , Fenótipo , alfa-Galactosidase , Humanos , Doença de Fabry/genética , Doença de Fabry/patologia , Doença de Fabry/enzimologia , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo , Masculino , Adulto , Variação Genética , Triexosilceramidas/metabolismo , Pessoa de Meia-Idade , Pele/patologia , Células Endoteliais/patologia , Células Endoteliais/metabolismo , Mutação , Glicolipídeos/metabolismo , Esfingolipídeos
4.
Cytogenet Genome Res ; 160(11-12): 659-663, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33472199

RESUMO

The 22q11.2 microduplication syndrome shows variable phenotypes with reduced penetrance compared to the 22q11.2 deletion syndrome. We report a woman with overgrowth and macrocephaly, mild mental retardation, heart defect, kidney anomalies, and dysmorphic features. Array-CGH analysis revealed a 246-kb duplication at the 22q11.2 region. No additional clinically significant CNVs were found. The case resembles a previously published case also showing overgrowth and macrocephaly with an almost identical 22q11.2 duplication of 252 kb.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Duplicação Cromossômica/genética , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patologia , Testes Genéticos , Adolescente , Adulto , Criança , Cromossomos Humanos Par 22/genética , Cromossomos Humanos Par 8/genética , Variações do Número de Cópias de DNA , Feminino , Humanos , Lactente , Masculino , Linhagem , Fenótipo , Síndrome
5.
Clin Genet ; 98(4): 418-419, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33294970

RESUMO

The clinical impact of duplications affecting the 11p15.5 region is difficult to predict, and depends on the parent-of-origin of the affected allele as well as on the type (deletion, duplication), the extent and genomic content of the variant. Three unrelated families with inheritance of duplications affecting the IC1 region in 11p15.5 through two generations but different phenotypes (Beckwith-Wiedemann and Silver-Russell syndromes, normal phenotype) are reported. The inconsistent phenotypic patterns of carriers of the same variant strongly indicate the impact of cis- and/or trans-acting modifiers on the clinical outcome of IC1 duplication carriers.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Predisposição Genética para Doença , Fator de Crescimento Insulin-Like II/genética , RNA Longo não Codificante/genética , Síndrome de Silver-Russell/genética , Alelos , Síndrome de Beckwith-Wiedemann/patologia , Criança , Pré-Escolar , Deleção Cromossômica , Duplicação Cromossômica/genética , Cromossomos Humanos Par 11/genética , Feminino , Impressão Genômica/genética , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/patologia
6.
Pediatr Blood Cancer ; 67(2): e28078, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31724816

RESUMO

Defects of platelet intracellular signaling can result in severe platelet dysfunction. Several mutations in each of the linked genes FERMT3 and RASGRP2 on chromosome 11 causing a Glanzmann-like bleeding phenotype have been identified so far. We report on novel variants in two unrelated pediatric patients with severe bleeding diathesis-one with leukocyte adhesion deficiency type III due to a homozygous frameshift in FERMT3 and the other with homozygous variants in both, FERMT3 and RASGRP2. We focus on the challenging genetic and functional variant assessment and aim to accentuate the risk of obtaining misleading results due to the phenomenon of genetic linkage.


Assuntos
Transtornos Plaquetários/patologia , Fatores de Troca do Nucleotídeo Guanina/genética , Transtornos Hemorrágicos/patologia , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Adolescente , Transtornos Plaquetários/genética , Criança , Feminino , Ligação Genética , Transtornos Hemorrágicos/genética , Homozigoto , Humanos , Masculino , Linhagem , Fenótipo , Prognóstico
7.
Z Kinder Jugendpsychiatr Psychother ; 48(6): 478-489, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33172359

RESUMO

Objective: Developmental dyslexia is a highly heritable specific reading and writing disability. To identify a possible new locus and candidate gene for this disability, we investigated a four-generation pedigree where transmission of dyslexia is consistent with an autosomal dominant inheritance pattern. Methods: We performed genome wide array-based SNP genotyping and parametric linkage analysis and sequencing analysis of protein-coding exons, exon-intron boundaries and conserved extragenic regions within the haplotype cosegregating with dyslexia in DNA from one affected and one unaffected family member. Cosegregation was confirmed by sequencing all available family members. Additionally, we analyzed 96 dyslexic individuals who had previously shown positive LOD scores on chromosome 4q28 as well as an even larger sample (n = 2591). Results: We found a single prominent linkage interval on chromosome 4q, where sequence analysis revealed a nucleotide variant in the 3' UTR of brain expressed SPRY1 in the dyslexic family member that cosegregated with dyslexia. This sequence alteration might affect the binding efficiency of the IGF2BP1 RNA-binding protein and thus influence the expression level of the SPRY1 gene product. An analysis of 96 individuals from a cohort of dyslexic individuals revealed a second heterozygous variant in this gene, which was absent in the unaffected sister of the proband. An investigation of the region in a much larger sample further found a nominal p-value of 0.0016 for verbal short-term memory (digit span) in 2,591 individuals for a neighboring SNV. After correcting for the local number of analyzed SNVs, and after taking into account linkage disequilibrium, we found this corresponds to a p-value of 0.0678 for this phenotype. Conclusions: We describe a new locus for familial dyslexia and discuss the possibility that SPRY1 might play a role in the etiology of a monogenic form of dyslexia.


Assuntos
Cromossomos Humanos Par 4/genética , Dislexia/genética , Regiões 3' não Traduzidas/genética , Saúde da Família , Humanos , Escore Lod , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linhagem , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo
8.
Hum Mutat ; 40(8): 1101-1114, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30924982

RESUMO

Dominant mutations in the MYH7 and MYBPC3 genes are common causes of inherited cardiomyopathies, which often demonstrate variable phenotypic expression and incomplete penetrance across family members. Biallelic inheritance is rare but allows gaining insights into the genetic mode of action of single variants. Here, we present three cases carrying a loss-of-function (LoF) variant in a compound heterozygous state with a missense variant in either MYH7 or MYBPC3 leading to severe cardiomyopathy with left ventricular noncompaction. Most likely, MYH7 haploinsufficiency due to one LoF allele results in a clinical phenotype only in compound heterozygous form with a missense variant. In contrast, haploinsufficiency in MYBPC3 results in a severe early-onset ventricular noncompaction phenotype requiring heart transplantation when combined with a de novo missense variant on the second allele. In addition, the missense variant may lead to an unstable protein, as overall only 20% of the MYBPC3 protein remain detectable in affected cardiac tissue compared to control tissue. In conclusion, in patients with early disease onset and atypical clinical course, biallelic inheritance or more complex variants including copy number variations and de novo mutations should be considered. In addition, the pathogenic consequence of variants may differ in heterozygous versus compound heterozygous state.


Assuntos
Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Miocárdio Ventricular não Compactado Isolado/genética , Mutação com Perda de Função , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/genética , Adolescente , Adulto , Feminino , Haploinsuficiência , Transplante de Coração , Humanos , Lactente , Miocárdio Ventricular não Compactado Isolado/terapia , Masculino , Linhagem , Fenótipo , Adulto Jovem
9.
Int J Cancer ; 145(4): 1020-1032, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30873613

RESUMO

Merkel cell carcinoma (MCC), an aggressive neuroendocrine skin tumor, is a polyomavirus-induced human cancer. To study the causal relationship of MCC carcinogenesis with the integrated Merkel cell polyomavirus (MCPyV) in detail, well-characterized MCC cell lines are needed. Consequently, in the current study, we established and characterized six MCPyV-positive MCC cell lines. Microarray-based comparative genomic hybridization revealed a stable genome carrying only a limited number of chromosomal gains and deletions. All cell lines expressed MCC markers Keratin-20 and neuron-specific enolase as well as truncated MCPyV-encoded large T antigen (LT). For five cell lines, we were able to identify the MCPyV-integration sites in introns of different genes. The LT-truncating stop codon mutations and integration sites were affirmed in the respective clinical patient samples. Inverse PCR suggested that three of the cell lines contained MCPyV genomes as concatemers. This notion was confirmed for the two cell lines with known integration sites. Importantly, our observation of distinct stop codon mutations in cell lines with concatemeric MCPyV integration indicates that these LT-truncating mutations occur before integration. In summary, we provide the detailed characterization of six MCPyV-positive MCC cell lines, which are likely to serve as valuable tools in future MCC research.


Assuntos
Antígenos Virais de Tumores/genética , Carcinoma de Célula de Merkel/genética , Poliomavírus das Células de Merkel/genética , Infecções por Polyomavirus/genética , Infecções Tumorais por Vírus/genética , Animais , Carcinoma de Célula de Merkel/virologia , Linhagem Celular Tumoral , Códon de Terminação/genética , Genoma Viral/genética , Humanos , Camundongos , Mutação/genética , Infecções por Polyomavirus/virologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/virologia , Infecções Tumorais por Vírus/virologia
10.
Genet Med ; 20(6): 599-607, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29236091

RESUMO

PurposeCopy-number variants (CNVs) are generally interpreted by linking the effects of gene dosage with phenotypes. The clinical interpretation of noncoding CNVs remains challenging. We investigated the percentage of disease-associated CNVs in patients with congenital limb malformations that affect noncoding cis-regulatory sequences versus genes sensitive to gene dosage effects.MethodsWe applied high-resolution copy-number analysis to 340 unrelated individuals with isolated limb malformation. To investigate novel candidate CNVs, we re-engineered human CNVs in mice using clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing.ResultsOf the individuals studied, 10% harbored CNVs segregating with the phenotype in the affected families. We identified 31 CNVs previously associated with congenital limb malformations and four novel candidate CNVs. Most of the disease-associated CNVs (57%) affected the noncoding cis-regulatory genome, while only 43% included a known disease gene and were likely to result from gene dosage effects. In transgenic mice harboring four novel candidate CNVs, we observed altered gene expression in all cases, indicating that the CNVs had a regulatory effect either by changing the enhancer dosage or altering the topological associating domain architecture of the genome.ConclusionOur findings suggest that CNVs affecting noncoding regulatory elements are a major cause of congenital limb malformations.


Assuntos
DNA Intergênico/genética , Deformidades Congênitas dos Membros/genética , Animais , Variações do Número de Cópias de DNA/genética , Feminino , Dosagem de Genes/genética , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Linhagem , Fenótipo
11.
Hum Mol Genet ; 24(11): 3119-32, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25712132

RESUMO

Chordin-Like 1 (CHRDL1) mutations cause non-syndromic X-linked megalocornea (XMC) characterized by enlarged anterior eye segments. Mosaic corneal degeneration, presenile cataract and secondary glaucoma are associated with XMC. Beside that CHRDL1 encodes Ventroptin, a secreted bone morphogenetic protein (BMP) antagonist, the molecular mechanism of XMC is not well understood yet. In a family with broad phenotypic variability of XMC, we identified the novel CHRDL1 frameshift mutation c.807_808delTC [p.H270Wfs*22] presumably causing CHRDL1 loss of function. Using Xenopus laevis as model organism, we demonstrate that chrdl1 is specifically expressed in the ocular tissue at late developmental stages. The chrdl1 knockdown directly resembles the human XMC phenotype and confirms CHRDL1 deficiency to cause XMC. Interestingly, secondary to this bmp4 is down-regulated in the Xenopus eyes. Moreover, phospho-SMAD1/5 is altered and BMP receptor 1A is reduced in a XMC patient. Together, we classify these observations as negative-feedback regulation due to the deficient BMP antagonism in XMC. As CHRDL1 is preferentially expressed in the limbal stem cell niche of adult human cornea, we assume that CHRDL1 plays a key role in cornea homeostasis. In conclusion, we provide novel insights into the molecular mechanism of XMC as well as into the specific role of CHRDL1 during cornea organogenesis, among others by the establishment of the first XMC in vivo model. We show that unravelling monogenic cornea disorders like XMC-with presumably disturbed cornea growth and differentiation-contribute to the identification of potential limbal stem cell niche factors that are promising targets for regenerative therapies of corneal injuries.


Assuntos
Oftalmopatias Hereditárias/genética , Proteínas do Olho/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Proteínas do Tecido Nervoso/genética , Adolescente , Animais , Sequência de Bases , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Córnea/patologia , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Expressão Gênica , Estudos de Associação Genética , Humanos , Masculino , Linhagem , Transdução de Sinais , Xenopus laevis
12.
Pediatr Blood Cancer ; 64(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27917594

RESUMO

BACKGROUND: Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disease characterized by oculocutaneous albinism and platelet dysfunction. We report on a novel HPS6 homozygous frameshift variant (c.1919_1920delTC; p.Val640Glyfs*29) in a nonconsanguineous Caucasian family with two affected siblings (index patients) who presented with oculocutaneous albinism at birth and a mild bleeding phenotype during childhood and adolescence. PROCEDURE: Genetic analysis was conducted by panel-based next-generation sequencing (NGS) and Sanger sequencing. Platelets of the index patients, their parents, and the unaffected sister were then comprehensively evaluated by luminoaggregometry, whole blood flow cytometry, immunoblotting, immunofluorescence, and transmission electron microscopy. RESULTS: The homozygous frameshift variant in HPS6 gene detected by panel-based NGS and its segregation in the family was confirmed by Sanger sequencing. Flow cytometric analysis of the patients' platelets revealed a substantially decreased mepacrine uptake and release upon activation with a thrombin receptor agonist. Electron microscopy of resting platelets confirmed diminished dense granule content and enhanced vacuolization. Reduced release of adenosine triphosphate and CD63 neoexposition upon activation indicated not only a lack of dense granule content, but even an impairment of dense granule release. CONCLUSIONS: Our results demonstrate that the novel loss-of-function variant in the HPS6 subunit of biogenesis of lysosome-related organelles complex 2 is pathologic and leads to a reduced platelet dense granules and their release. The findings are compatible with an impaired platelet function and hence an enhanced bleeding risk. In future, a valid genotype-phenotype correlation may translate into best supportive care, especially regarding elective surgery or trauma management.


Assuntos
Antineoplásicos/metabolismo , Plaquetas/metabolismo , Síndrome de Hermanski-Pudlak/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Quinacrina/metabolismo , Trifosfato de Adenosina/metabolismo , Adolescente , Sequência de Bases , Transporte Biológico/genética , Plaquetas/citologia , Criança , Feminino , Citometria de Fluxo , Mutação da Fase de Leitura/genética , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Microscopia Eletrônica , Análise de Sequência de DNA , Deleção de Sequência/genética , Tetraspanina 30/metabolismo
13.
Nat Genet ; 40(9): 1065-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19165920

RESUMO

CASK is a multi-domain scaffolding protein that interacts with the transcription factor TBR1 and regulates expression of genes involved in cortical development such as RELN. Here we describe a previously unreported X-linked brain malformation syndrome caused by mutations of CASK. All five affected individuals with CASK mutations had congenital or postnatal microcephaly, disproportionate brainstem and cerebellar hypoplasia, and severe mental retardation.


Assuntos
Tronco Encefálico/anormalidades , Cerebelo/anormalidades , Doenças Genéticas Ligadas ao Cromossomo X/genética , Guanilato Quinases/genética , Microcefalia/genética , Mutação , Pré-Escolar , Orelha/anormalidades , Feminino , Humanos , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína Reelina , Síndrome
14.
Am J Hum Genet ; 93(1): 67-77, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23768516

RESUMO

Deletion 1p36 syndrome is recognized as the most common terminal deletion syndrome. Here, we describe the loss of a gene within the deletion that is responsible for the cardiomyopathy associated with monosomy 1p36, and we confirm its role in nonsyndromic left ventricular noncompaction cardiomyopathy (LVNC) and dilated cardiomyopathy (DCM). With our own data and publically available data from array comparative genomic hybridization (aCGH), we identified a minimal deletion for the cardiomyopathy associated with 1p36del syndrome that included only the terminal 14 exons of the transcription factor PRDM16 (PR domain containing 16), a gene that had previously been shown to direct brown fat determination and differentiation. Resequencing of PRDM16 in a cohort of 75 nonsyndromic individuals with LVNC detected three mutations, including one truncation mutant, one frameshift null mutation, and a single missense mutant. In addition, in a series of cardiac biopsies from 131 individuals with DCM, we found 5 individuals with 4 previously unreported nonsynonymous variants in the coding region of PRDM16. None of the PRDM16 mutations identified were observed in more than 6,400 controls. PRDM16 has not previously been associated with cardiac disease but is localized in the nuclei of cardiomyocytes throughout murine and human development and in the adult heart. Modeling of PRDM16 haploinsufficiency and a human truncation mutant in zebrafish resulted in both contractile dysfunction and partial uncoupling of cardiomyocytes and also revealed evidence of impaired cardiomyocyte proliferative capacity. In conclusion, mutation of PRDM16 causes the cardiomyopathy in 1p36 deletion syndrome as well as a proportion of nonsyndromic LVNC and DCM.


Assuntos
Cardiomiopatia Dilatada/genética , Transtornos Cromossômicos/genética , Mapeamento Cromossômico/métodos , Proteínas de Ligação a DNA/genética , Miocárdio Ventricular não Compactado Isolado/genética , Fatores de Transcrição/genética , Animais , Cardiomiopatia Dilatada/patologia , Estudos de Casos e Controles , Proliferação de Células , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Hibridização Genômica Comparativa , Éxons , Mutação da Fase de Leitura , Humanos , Mutação de Sentido Incorreto , Contração Miocárdica , Miócitos Cardíacos , Peixe-Zebra/embriologia , Peixe-Zebra/genética
15.
Mamm Genome ; 27(3-4): 111-21, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26803617

RESUMO

We performed exome sequencing for mutation discovery of an ENU (N-ethyl-N-nitrosourea)-derived mouse model characterized by significant elevated plasma alkaline phosphatase (ALP) activities in female and male mutant mice, originally named BAP014 (bone screen alkaline phosphatase #14). We identified a novel loss-of-function mutation within the Fam46a (family with sequence similarity 46, member A) gene (NM_001160378.1:c.469G>T, NP_001153850.1:p.Glu157*). Heterozygous mice of this mouse line (renamed Fam46a (E157*Mhda)) had significantly high ALP activities and apparently no other differences in morphology compared to wild-type mice. In contrast, homozygous Fam46a (E157*Mhda) mice showed severe morphological and skeletal abnormalities including short stature along with limb, rib, pelvis, and skull deformities with minimal trabecular bone and reduced cortical bone thickness in long bones. ALP activities of homozygous mutants were almost two-fold higher than in heterozygous mice. Fam46a is weakly expressed in most adult and embryonic tissues with a strong expression in mineralized tissues as calvaria and femur. The FAM46A protein is computationally predicted as a new member of the superfamily of nucleotidyltransferase fold proteins, but little is known about its function. Fam46a (E157*Mhda) mice are the first mouse model for a mutation within the Fam46a gene.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Códon sem Sentido , Exoma , Fosfatase Alcalina/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Análise Mutacional de DNA , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Expressão Gênica , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Camundongos Knockout , Fenótipo
16.
J Med Genet ; 52(7): 476-83, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26032025

RESUMO

INTRODUCTION: Mesomelic dysplasias are a group of skeletal disorders characterised by shortness of the middle limb segments (mesomelia). They are divided into 11 different categories. Among those without known molecular basis is mesomelic dysplasia Savarirayan type, characterised by severe shortness of the middle segment of the lower limb. OBJECTIVE: To identify the molecular cause of mesomelic dysplasia Savarirayan type. METHODS AND RESULTS: We performed array comparative genomic hybridisation in three unrelated patients with mesomelic dysplasia Savarirayan type and identified 2 Mb overlapping de novo microdeletions on chromosome 6p22.3. The deletions encompass four known genes: MBOAT1, E2F3, CDKAL1 and SOX4. All patients showed mesomelia of the lower limbs with hypoplastic tibiae and fibulae. We identified a fourth patient with intellectual disability and an overlapping slightly larger do novo deletion also encompassing the flanking gene ID4. Given the fact that the fourth patient had no skeletal abnormalities and none of the genes in the deleted interval are known to be associated with abnormalities in skeletal development, other mutational mechanisms than loss of function of the deleted genes have to be considered. Analysis of the genomic region showed that the deletion removes two regulatory boundaries and brings several potential limb enhancers into close proximity of ID4. Thus, the deletion could result in the aberrant activation and misexpression of ID4 in the limb bud, thereby causing the mesomelic dysplasia. CONCLUSIONS: Our data indicate that the distinct deletion 6p22.3 is associated with mesomelic dysplasia Savarirayan type featuring hypoplastic, triangular-shaped tibiae and abnormally shaped or hypoplastic fibulae.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 6/genética , Fíbula/anormalidades , Proteínas Inibidoras de Diferenciação/metabolismo , Perna (Membro)/anormalidades , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Rádio (Anatomia)/anormalidades , Deleção de Sequência/genética , Tíbia/anormalidades , Ulna/anormalidades , Acetiltransferases/genética , Sequência de Bases , Hibridização Genômica Comparativa , Quinase 5 Dependente de Ciclina/genética , Fator de Transcrição E2F3/genética , Fíbula/patologia , Humanos , Proteínas Inibidoras de Diferenciação/genética , Proteínas de Membrana/genética , Dados de Sequência Molecular , Rádio (Anatomia)/patologia , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXC , Análise de Sequência de DNA , Tíbia/patologia , Ulna/patologia , tRNA Metiltransferases
17.
Am J Hum Genet ; 91(4): 629-35, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23022097

RESUMO

The study of homeotic-transformation mutants in model organisms such as Drosophila revolutionized the field of developmental biology, but how these mutants relate to human developmental defects remains to be elucidated. Here, we show that Liebenberg syndrome, an autosomal-dominant upper-limb malformation, shows features of a homeotic limb transformation in which the arms have acquired morphological characteristics of a leg. Using high-resolution array comparative genomic hybridization and paired-end whole-genome sequencing, we identified two deletions and a translocation 5' of PITX1. The structural changes are likely to remove active PITX1 forelimb suppressor and/or insulator elements and thereby move active enhancer elements in the vicinity of the PITX1 regulatory landscape. We generated transgenic mice in which PITX1 was misexpressed under the control of a nearby enhancer and were able to recapitulate the Liebenberg phenotype.


Assuntos
Braquidactilia/genética , Rearranjo Gênico , Genes Homeobox , Loci Gênicos , Deformidades Congênitas da Mão/genética , Fatores de Transcrição Box Pareados/genética , Sinostose/genética , Transformação Genética , Animais , Ossos do Carpo/anormalidades , Hibridização Genômica Comparativa/métodos , Articulação do Cotovelo/anormalidades , Feminino , Dedos/anormalidades , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Genoma Humano , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Análise de Sequência de DNA/métodos , Translocação Genética , Articulação do Punho/anormalidades
18.
Blood ; 122(7): 1312-5, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23733340

RESUMO

To ascertain the genetic basis of a paroxysmal nocturnal hemoglobinuria (PNH) case without somatic mutations in PIGA, we performed deep next-generation sequencing on all exons of known genes of the glycosylphosphatidylinositol (GPI) anchor synthesis pathway. We identified a heterozygous germline splice site mutation in PIGT and a somatic 8-MB deletion in granulocytes affecting the other copy of PIGT. PIGA is essential for GPI anchor synthesis, whereas PIGT is essential for attachment of the preassembled GPI anchor to proteins. Although a single mutation event in the X-chromosomal gene PIGA is known to cause GPI-anchored protein deficiency, 2 such hits are required in the autosomal gene PIGT. Our data indicate that PNH can occur even in the presence of fully assembled GPI if its transfer to proteins is defective in hematopoietic stem cells.


Assuntos
Aciltransferases/genética , Mutação em Linhagem Germinativa/genética , Hemoglobinúria Paroxística/genética , Mutação/genética , Adulto , Processamento Alternativo/genética , Animais , Células CHO , Estudos de Casos e Controles , Hibridização Genômica Comparativa , Cricetulus , Éxons/genética , Feminino , Citometria de Fluxo , Genes Ligados ao Cromossomo X , Humanos , Hibridização in Situ Fluorescente , Análise de Sequência de DNA , Deleção de Sequência
19.
Artigo em Inglês | MEDLINE | ID: mdl-21756107

RESUMO

Whereas single-nucleotide polymorphisms and their role in predisposition to disease have been studied extensively, the analysis of structural variants--genomic changes such as insertions, deletions, inversions, duplications, and translocations--is still in its infancy. Changes in copy number, also known as copy-number variations (CNVs), constitute one such group of these structural variants. CNVs are structural genomic variants that arise from deletions (loss) or duplications (gain), and as a consequence result in a copy-number change of the respective genomic region. CNVs may include entire genes or regions of transcribed sequence, or, indeed, comprise only nontranscribed sequences. Whereas the duplication or deletion of a gene can be expected to have an effect on gene dosage, the consequences of CNVs in nontranscribed sequences are less obvious. Here we review CNVs that involve regulatory nontranscribed regions of the genome, describe the associated human phenotypes, and discuss possible disease mechanisms.


Assuntos
Variações do Número de Cópias de DNA , Doença/genética , Elementos Reguladores de Transcrição , Animais , Dosagem de Genes , Regulação da Expressão Gênica , Humanos
20.
Am J Hum Genet ; 88(1): 70-5, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21167467

RESUMO

Indian hedgehog (IHH) is a secreted signaling molecule of the hedgehog family known to play important roles in the regulation of chondrocyte differentiation, cortical bone formation, and the development of joints. Here, we describe that copy-number variations of the IHH locus involving conserved noncoding elements (CNEs) are associated with syndactyly and craniosynostosis. These CNEs are able to drive reporter gene expression in a pattern highly similar to wild-type Ihh expression. We postulate that the observed duplications lead to a misexpression and/or overexpression of IHH and by this affect the complex regulatory signaling network during digit and skull development.


Assuntos
Craniossinostoses/genética , Variações do Número de Cópias de DNA , Duplicação Gênica , Loci Gênicos , Proteínas Hedgehog/genética , Sindactilia/genética , Animais , Sequência Conservada/genética , Feminino , Dedos/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Crânio/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa