Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 140(5): 678-91, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20211137

RESUMO

The incorporation of histone H3 variants has been implicated in the epigenetic memory of cellular state. Using genome editing with zinc-finger nucleases to tag endogenous H3.3, we report genome-wide profiles of H3 variants in mammalian embryonic stem cells and neuronal precursor cells. Genome-wide patterns of H3.3 are dependent on amino acid sequence and change with cellular differentiation at developmentally regulated loci. The H3.3 chaperone Hira is required for H3.3 enrichment at active and repressed genes. Strikingly, Hira is not essential for localization of H3.3 at telomeres and many transcription factor binding sites. Immunoaffinity purification and mass spectrometry reveal that the proteins Atrx and Daxx associate with H3.3 in a Hira-independent manner. Atrx is required for Hira-independent localization of H3.3 at telomeres and for the repression of telomeric RNA. Our data demonstrate that multiple and distinct factors are responsible for H3.3 localization at specific genomic locations in mammalian cells.


Assuntos
Histonas/análise , Telômero/química , Animais , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células-Tronco Embrionárias/metabolismo , Genoma , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Telômero/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sítio de Iniciação de Transcrição
2.
Kidney Int ; 101(4): 720-732, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35090878

RESUMO

To guide the development of therapeutic interventions for acute kidney injury, elucidating the deleterious pathways of this global health problem is highly warranted. Emerging evidence has indicated a pivotal role of endothelial dysfunction in the etiology of this disease. We found that the class III semaphorin SEMA3C was ectopically upregulated with full length protein excreted into the blood and truncated protein secreted into the urine upon kidney injury and hypothesized a role for SEAM3C in acute kidney injury. Sema3c was genetically abrogated during acute kidney injury and subsequent kidney morphological and functional defects in two well-characterized models of acute kidney injury; warm ischemia/reperfusion and folic acid injection were analyzed. Employing a beta actin-dependent, inducible knockout of Sema3c, we demonstrate that in acute kidney injury SEMA3C promotes interstitial edema, leucocyte infiltration and tubular injury. Additionally, intravital microscopy combined with Evans Blue dye extravasation and primary culture of magnetically sorted peritubular endothelial cells identified a novel role for SEMA3C in promoting vascular permeability. Thus, our study points to microvascular permeability as an important driver of injury in acute kidney injury, and to SEMA3C as a novel permeability factor and potential target for therapeutic intervention.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Semaforinas , Injúria Renal Aguda/genética , Injúria Renal Aguda/prevenção & controle , Animais , Permeabilidade Capilar , Células Endoteliais/metabolismo , Feminino , Humanos , Rim/metabolismo , Masculino , Camundongos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/prevenção & controle , Semaforinas/genética , Semaforinas/metabolismo
3.
Angiogenesis ; 24(2): 271-288, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33825109

RESUMO

Lymphatic vessels have critical roles in both health and disease and their study is a rapidly evolving area of vascular biology. The consensus on how the first lymphatic vessels arise in the developing embryo has recently shifted. Originally, they were thought to solely derive by sprouting from veins. Since then, several studies have uncovered novel cellular mechanisms and a diversity of contributing cell lineages in the formation of organ lymphatic vasculature. Here, we review the key mechanisms and cell lineages contributing to lymphatic development, discuss the advantages and limitations of experimental techniques used for their study and highlight remaining knowledge gaps that require urgent attention. Emerging technologies should accelerate our understanding of how lymphatic vessels develop normally and how they contribute to disease.


Assuntos
Linhagem da Célula , Células Endoteliais/metabolismo , Linfangiogênese , Vasos Linfáticos/embriologia , Animais , Humanos
4.
Hum Mol Genet ; 27(3): 529-545, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29228333

RESUMO

DNAAF1 (LRRC50) is a cytoplasmic protein required for dynein heavy chain assembly and cilia motility, and DNAAF1 mutations cause primary ciliary dyskinesia (PCD; MIM 613193). We describe four families with DNAAF1 mutations and complex congenital heart disease (CHD). In three families, all affected individuals have typical PCD phenotypes. However, an additional family demonstrates isolated CHD (heterotaxy) in two affected siblings, but no clinical evidence of PCD. We identified a homozygous DNAAF1 missense mutation, p.Leu191Phe, as causative for heterotaxy in this family. Genetic complementation in dnaaf1-null zebrafish embryos demonstrated the rescue of normal heart looping with wild-type human DNAAF1, but not the p.Leu191Phe variant, supporting the conserved pathogenicity of this DNAAF1 missense mutation. This observation points to a phenotypic continuum between CHD and PCD, providing new insights into the pathogenesis of isolated CHD. In further investigations of the function of DNAAF1 in dynein arm assembly, we identified interactions with members of a putative dynein arm assembly complex. These include the ciliary intraflagellar transport protein IFT88 and the AAA+ (ATPases Associated with various cellular Activities) family proteins RUVBL1 (Pontin) and RUVBL2 (Reptin). Co-localization studies support these findings, with the loss of RUVBL1 perturbing the co-localization of DNAAF1 with IFT88. We show that RUVBL1 orthologues have an asymmetric left-sided distribution at both the mouse embryonic node and the Kupffer's vesicle in zebrafish embryos, with the latter asymmetry dependent on DNAAF1. These results suggest that DNAAF1-RUVBL1 biochemical and genetic interactions have a novel functional role in symmetry breaking and cardiac development.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Transporte/metabolismo , Cílios/metabolismo , DNA Helicases/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Animais , Proteínas de Transporte/genética , Cílios/fisiologia , DNA Helicases/genética , Feminino , Genótipo , Células HEK293 , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/genética , Mutação de Sentido Incorreto/genética , Linhagem , Fenótipo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Sequenciamento do Exoma/métodos , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
J Mol Cell Cardiol ; 114: 29-37, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29061339

RESUMO

Physiological changes during embryonic development are associated with changes in the isoform expression of both myocyte sarcomeric proteins and of erythrocyte haemoglobins. Cell type-specific isoform expression of these genes also occurs. Although these changes appear to be coordinated, it is unclear how changes in these disparate cell types may be linked. The transcription factor Hic2 is required for normal cardiac development and the mutant is embryonic lethal. Hic2 embryos exhibit precocious expression of the definitive-lineage haemoglobin Hbb-bt in circulating primitive erythrocytes and of foetal isoforms of cardiomyocyte genes (creatine kinase, Ckm, and eukaryotic elongation factor Eef1a2) as well as ectopic cardiac expression of fast-twitch skeletal muscle troponin isoforms. We propose that HIC2 regulates a switching event within both the contractile machinery of cardiomyocytes and the oxygen carrying systems during the developmental period where demands on cardiac loading change rapidly.


Assuntos
Sistema Cardiovascular/embriologia , Sistema Cardiovascular/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Perda do Embrião/patologia , Eritrócitos/metabolismo , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hemoglobinas/metabolismo , Fatores de Transcrição Kruppel-Like/sangue , Camundongos , Mutação/genética , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Fatores de Tempo , Troponina I/metabolismo , Proteínas Supressoras de Tumor/sangue
7.
Kidney Int ; 93(4): 903-920, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29398135

RESUMO

The Wilms' tumor suppressor gene, WT1, encodes a zinc finger protein that regulates podocyte development and is highly expressed in mature podocytes. Mutations in the WT1 gene are associated with the development of renal failure due to the formation of scar tissue within glomeruli, the mechanisms of which are poorly understood. Here, we used a tamoxifen-based CRE-LoxP system to induce deletion of Wt1 in adult mice to investigate the mechanisms underlying evolution of glomerulosclerosis. Podocyte apoptosis was evident as early as the fourth day post-induction and increased during disease progression, supporting a role for Wt1 in mature podocyte survival. Podocyte Notch activation was evident at disease onset with upregulation of Notch1 and its transcriptional targets, including Nrarp. There was repression of podocyte FoxC2 and upregulation of Hey2 supporting a role for a Wt1/FoxC2/Notch transcriptional network in mature podocyte injury. The expression of cleaved Notch1 and HES1 proteins in podocytes of mutant mice was confirmed in early disease. Furthermore, induction of podocyte HES1 expression was associated with upregulation of genes implicated in epithelial mesenchymal transition, thereby suggesting that HES1 mediates podocyte EMT. Lastly, early pharmacological inhibition of Notch signaling ameliorated glomerular scarring and albuminuria. Thus, loss of Wt1 in mature podocytes modulates podocyte Notch activation, which could mediate early events in WT1-related glomerulosclerosis.


Assuntos
Glomerulonefrite/metabolismo , Podócitos/metabolismo , Receptor Notch1/metabolismo , Proteínas Repressoras/metabolismo , Albuminúria/genética , Albuminúria/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Glomerulonefrite/genética , Glomerulonefrite/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Endogâmicos C57BL , Camundongos Knockout , Podócitos/patologia , Proteínas/genética , Proteínas/metabolismo , Receptor Notch1/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Transdução de Sinais , Transcrição Gênica , Proteínas WT1
8.
Am J Med Genet A ; 176(10): 2070-2081, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30380194

RESUMO

The 22q11.2 deletion syndrome (22q11.2DS) is a congenital malformation and neuropsychiatric disorder caused by meiotic chromosome rearrangements. One of the goals of this review is to summarize the current state of basic research studies of 22q11.2DS. It highlights efforts to understand the mechanisms responsible for the 22q11.2 deletion that occurs in meiosis. This mechanism involves the four sets of low copy repeats (LCR22) that are dispersed in the 22q11.2 region and the deletion is mediated by nonallelic homologous recombination events. This review also highlights selected genes mapping to the 22q11.2 region that may contribute to the typical clinical findings associated with the disorder and explain that mutations in genes on the remaining allele can uncover rare recessive conditions. Another important aspect of 22q11.2DS is the existence of phenotypic heterogeneity. While some patients are mildly affected, others have severe medical, cognitive, and/or psychiatric challenges. Variability may be due in part to the presence of genetic modifiers. This review discusses current genome-wide efforts to identify such modifiers that could shed light on molecular pathways required for normal human development, cognition or behavior.


Assuntos
Anormalidades Múltiplas/genética , Cromossomos Humanos Par 22 , Síndrome de DiGeorge/genética , Mutação , Deleção Cromossômica , Síndrome de DiGeorge/etiologia , Genes Recessivos , Testes Genéticos , Humanos , Meiose
9.
Mol Biol Rep ; 45(5): 1001-1011, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30030774

RESUMO

HIRA is a histone chaperone known to modulate gene expression through the deposition of H3.3. Conditional knockout of Hira in embryonic mouse hearts leads to cardiac septal defects. Loss of function mutation in HIRA, together with other chromatin modifiers, was found in patients with congenital heart diseases. However, the effects of HIRA on gene expression at earlier stages of cardiogenic mesoderm differentiation have not yet been studied. Differentiation of mouse embryonic stem cells (mESCs) towards cardiomyocytes mimics some of these early events and is an accepted model of these early stages. We performed RNA-Seq and H3.3-HA ChIP-seq on both WT and Hira-null mESCs and early cardiomyocyte progenitors of both genotypes. Analysis of RNA-seq data showed differential down regulation of cardiovascular development-related genes in Hira-null cardiomyocytes compared to WT cardiomyocytes. We found HIRA-dependent H3.3 deposition at these genes. In particular, we observed that HIRA influenced directly the expression of the transcription factors Gata6, Meis1 and Tbx2, essential for cardiac septation, through H3.3 deposition. We therefore identified new direct targets of HIRA during cardiac differentiation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Chaperonas de Histonas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Miócitos Cardíacos/citologia , Análise de Sequência de RNA/métodos , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Linhagem Celular , Regulação para Baixo , Elementos Facilitadores Genéticos , Fator de Transcrição GATA6/genética , Defeitos dos Septos Cardíacos/embriologia , Defeitos dos Septos Cardíacos/metabolismo , Histonas/metabolismo , Mutação com Perda de Função , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Proteína Meis1/genética , Miócitos Cardíacos/metabolismo , Proteínas com Domínio T/genética , Fatores de Transcrição/metabolismo
10.
Am J Med Genet C Semin Med Genet ; 175(4): 487-495, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29088513

RESUMO

Heart defects caused by loss-of-function mutations in CHD7 are a frequent cause of morbidity and mortality in CHARGE syndrome. Here we review the clinical and molecular aspects of CHD7 that are related to the cardiovascular manifestations of the syndrome. The types of heart defects found in patients with CHD7 mutations are variable, with an overrepresentation of atrioventricular septal defect and outflow tract defect including aortic arch anomalies compared to nonsyndromic heart defects. Chd7 haploinsufficiency in mouse is a good model for studying the heart effects seen in CHARGE syndrome, and mouse models reveal a role for Chd7 in multiple lineages during heart development. Formation of the great vessels requires Chd7 expression in the pharyngeal surface ectoderm, and this expression likely has an non-autonomous effect on neural crest cells. In the cardiogenic mesoderm, Chd7 is required for atrioventricular cushion development and septation of the outflow tract. Emerging knowledge about the function of CHD7 in the heart indicates that it may act in concert with transcription factors such as TBX1 and SMADs to regulate genes such as p53 and the cardiac transcription factor NKX2.5.


Assuntos
DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Estudos de Associação Genética , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Mutação , Fenótipo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Síndrome CHARGE/diagnóstico , Síndrome CHARGE/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Especificidade de Órgãos/genética , Organogênese/genética , Transdução de Sinais
11.
Dev Biol ; 405(1): 82-95, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26102480

RESUMO

CHARGE syndrome is caused by spontaneous loss-of-function mutations to the ATP-dependant chromatin remodeller chromodomain-helicase-DNA-binding protein 7 (CHD7). It is characterised by a distinct pattern of congenital anomalies, including cardiovascular malformations. Disruption to the neural crest lineage has previously been emphasised in the aetiology of this developmental disorder. We present evidence for an additional requirement for CHD7 activity in the Mesp1-expressing anterior mesoderm during heart development. Conditional ablation of Chd7 in this lineage results in major structural cardiovascular defects akin to those seen in CHARGE patients, as well as a striking loss of cardiac innervation and embryonic lethality. Genome-wide transcriptional analysis identified aberrant expression of key components of the Class 3 Semaphorin and Slit-Robo signalling pathways in Chd7(fl/fl);Mesp1-Cre mutant hearts. CHD7 localises at the Sema3c promoter in vivo, with alteration of the local chromatin structure seen following Chd7 ablation, suggestive of direct transcriptional regulation. Furthermore, we uncover a novel role for CHD7 activity upstream of critical calcium handling genes, and demonstrate an associated functional defect in the ability of cardiomyocytes to undergo excitation-contraction coupling. This work therefore reveals the importance of CHD7 in the cardiogenic mesoderm for multiple processes during cardiovascular development.


Assuntos
Sistema Cardiovascular/embriologia , Sistema Cardiovascular/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/patologia , Sinalização do Cálcio/genética , Sistema Cardiovascular/inervação , Cruzamentos Genéticos , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Endocárdio/anormalidades , Endocárdio/patologia , Acoplamento Excitação-Contração/genética , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Integrases/metabolismo , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Semaforinas/metabolismo
12.
J Biol Chem ; 290(21): 13053-63, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25847244

RESUMO

RUNX1 (Runt-related transcription factor 1) is indispensable for the generation of hemogenic endothelium. However, the regulation of RUNX1 during this developmental process is poorly understood. We investigated the role of the histone chaperone HIRA (histone cell cycle regulation-defective homolog A) from this perspective and report that HIRA significantly contributes toward the regulation of RUNX1 in the transition of differentiating mouse embryonic stem cells from hemogenic to hematopoietic stage. Direct interaction of HIRA and RUNX1 activates the downstream targets of RUNX1 implicated in generation of hematopoietic stem cells. At the molecular level, HIRA-mediated incorporation of histone H3.3 variant within the Runx1 +24 mouse conserved noncoding element is essential for the expression of Runx1 during endothelial to hematopoietic transition. An inactive chromatin at the intronic enhancer of Runx1 in absence of HIRA significantly repressed the transition of cells from hemogenic to hematopoietic fate. We expect that the HIRA-RUNX1 axis might open up a novel approach in understanding leukemogenesis in future.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Chaperonas de Histonas/fisiologia , Fatores de Transcrição/fisiologia , Animais , Western Blotting , Proteínas de Ciclo Celular/antagonistas & inibidores , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Imunofluorescência , Células-Tronco Hematopoéticas/metabolismo , Chaperonas de Histonas/antagonistas & inibidores , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/antagonistas & inibidores , Saco Vitelino/citologia , Saco Vitelino/metabolismo
13.
Trends Genet ; 29(12): 700-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24035368

RESUMO

The immense challenge of annotating the entire mouse genome has stimulated the development of cutting-edge imaging technologies in a drive for novel information. These techniques promise to improve understanding of the genes involved in embryo development, at least one third of which have been shown to be essential. Aligning advanced imaging technologies with biological needs will be fundamental to maximising the number of phenotypes discovered in the coming years. International efforts are underway to meet this challenge through an integrated and sophisticated approach to embryo phenotyping. We review rapid advances made in the imaging field over the past decade and provide a comprehensive examination of the relative merits of current and emerging techniques. The aim of this review is to provide a guide to state-of-the-art embryo imaging that will enable informed decisions as to which technology to use and fuel conversations between expert imaging laboratories, researchers, and core mouse production facilities.


Assuntos
Diagnóstico por Imagem , Desenvolvimento Embrionário , Animais , Camundongos
14.
Am J Hum Genet ; 92(1): 88-98, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23261303

RESUMO

Defects in motile cilia and sperm flagella cause primary ciliary dyskinesia (PCD), characterized by chronic airway disease, infertility, and left-right laterality disturbances, usually as a result of loss of the outer dynein arms (ODAs) that power cilia/flagella beating. Here, we identify loss-of-function mutations in CCDC114 causing PCD with laterality malformations involving complex heart defects. CCDC114 is homologous to DCC2, an ODA microtubule-docking complex component of the biflagellate alga Chlamydomonas. We show that CCDC114 localizes along the entire length of human cilia and that its deficiency causes a complete absence of ciliary ODAs, resulting in immotile cilia. Thus, CCDC114 is an essential ciliary protein required for microtubular attachment of ODAs in the axoneme. Fertility is apparently not greatly affected by CCDC114 deficiency, and qPCR shows that this may explained by low transcript expression in testis compared to ciliated respiratory epithelium. One CCDC114 mutation, c.742G>A, dating back to at least the 1400s, presents an important diagnostic and therapeutic target in the isolated Dutch Volendam population.


Assuntos
Axonema/genética , Síndrome de Kartagener/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação , Sítios de Splice de RNA , Sequência de Bases , Dineínas , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Linhagem
15.
Am J Hum Genet ; 93(3): 515-23, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23910462

RESUMO

Short-rib polydactyly syndromes (SRPS I-V) are a group of lethal congenital disorders characterized by shortening of the ribs and long bones, polydactyly, and a range of extraskeletal phenotypes. A number of other disorders in this grouping, including Jeune and Ellis-van Creveld syndromes, have an overlapping but generally milder phenotype. Collectively, these short-rib dysplasias (with or without polydactyly) share a common underlying defect in primary cilium function and form a subset of the ciliopathy disease spectrum. By using whole-exome capture and massive parallel sequencing of DNA from an affected Australian individual with SRPS type III, we detected two novel heterozygous mutations in WDR60, a relatively uncharacterized gene. These mutations segregated appropriately in the unaffected parents and another affected family member, confirming compound heterozygosity, and both were predicted to have a damaging effect on the protein. Analysis of an additional 54 skeletal ciliopathy exomes identified compound heterozygous mutations in WDR60 in a Spanish individual with Jeune syndrome of relatively mild presentation. Of note, these two families share one novel WDR60 missense mutation, although haplotype analysis suggested no shared ancestry. We further show that WDR60 localizes at the base of the primary cilium in wild-type human chondrocytes, and analysis of fibroblasts from affected individuals revealed a defect in ciliogenesis and aberrant accumulation of the GLI2 transcription factor at the centrosome or basal body in the absence of an obvious axoneme. These findings show that WDR60 mutations can cause skeletal ciliopathies and suggest a role for WDR60 in ciliogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Síndrome de Ellis-Van Creveld/genética , Mutação/genética , Síndrome de Costela Curta e Polidactilia/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Pré-Escolar , Condrócitos/metabolismo , Condrócitos/patologia , Segregação de Cromossomos/genética , Cílios/metabolismo , Síndrome de Ellis-Van Creveld/diagnóstico por imagem , Evolução Fatal , Feminino , Feto/diagnóstico por imagem , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Linhagem , Gravidez , Radiografia , Síndrome de Costela Curta e Polidactilia/diagnóstico por imagem
16.
Am J Hum Genet ; 93(5): 932-44, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24183451

RESUMO

Bidirectional (anterograde and retrograde) motor-based intraflagellar transport (IFT) governs cargo transport and delivery processes that are essential for primary cilia growth and maintenance and for hedgehog signaling functions. The IFT dynein-2 motor complex that regulates ciliary retrograde protein transport contains a heavy chain dynein ATPase/motor subunit, DYNC2H1, along with other less well functionally defined subunits. Deficiency of IFT proteins, including DYNC2H1, underlies a spectrum of skeletal ciliopathies. Here, by using exome sequencing and a targeted next-generation sequencing panel, we identified a total of 11 mutations in WDR34 in 9 families with the clinical diagnosis of Jeune syndrome (asphyxiating thoracic dystrophy). WDR34 encodes a WD40 repeat-containing protein orthologous to Chlamydomonas FAP133, a dynein intermediate chain associated with the retrograde intraflagellar transport motor. Three-dimensional protein modeling suggests that the identified mutations all affect residues critical for WDR34 protein-protein interactions. We find that WDR34 concentrates around the centrioles and basal bodies in mammalian cells, also showing axonemal staining. WDR34 coimmunoprecipitates with the dynein-1 light chain DYNLL1 in vitro, and mining of proteomics data suggests that WDR34 could represent a previously unrecognized link between the cytoplasmic dynein-1 and IFT dynein-2 motors. Together, these data show that WDR34 is critical for ciliary functions essential to normal development and survival, most probably as a previously unrecognized component of the mammalian dynein-IFT machinery.


Assuntos
Proteínas de Transporte/genética , Dineínas do Citoplasma/genética , Síndrome de Ellis-Van Creveld/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Animais , Povo Asiático/genética , Axonema/genética , Criança , Chlamydomonas/genética , Cílios/genética , Cílios/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Síndrome de Ellis-Van Creveld/patologia , Exoma , Éxons , Humanos , Lactente , Recém-Nascido , Mutação , Conformação Proteica , Proteômica , População Branca/genética
17.
Am J Hum Genet ; 93(5): 915-25, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24140113

RESUMO

Intraflagellar transport (IFT) depends on two evolutionarily conserved modules, subcomplexes A (IFT-A) and B (IFT-B), to drive ciliary assembly and maintenance. All six IFT-A components and their motor protein, DYNC2H1, have been linked to human skeletal ciliopathies, including asphyxiating thoracic dystrophy (ATD; also known as Jeune syndrome), Sensenbrenner syndrome, and Mainzer-Saldino syndrome (MZSDS). Conversely, the 14 subunits in the IFT-B module, with the exception of IFT80, have unknown roles in human disease. To identify additional IFT-B components defective in ciliopathies, we independently performed different mutation analyses: candidate-based sequencing of all IFT-B-encoding genes in 1,467 individuals with a nephronophthisis-related ciliopathy or whole-exome resequencing in 63 individuals with ATD. We thereby detected biallelic mutations in the IFT-B-encoding gene IFT172 in 12 families. All affected individuals displayed abnormalities of the thorax and/or long bones, as well as renal, hepatic, or retinal involvement, consistent with the diagnosis of ATD or MZSDS. Additionally, cerebellar aplasia or hypoplasia characteristic of Joubert syndrome was present in 2 out of 12 families. Fibroblasts from affected individuals showed disturbed ciliary composition, suggesting alteration of ciliary transport and signaling. Knockdown of ift172 in zebrafish recapitulated the human phenotype and demonstrated a genetic interaction between ift172 and ift80. In summary, we have identified defects in IFT172 as a cause of complex ATD and MZSDS. Our findings link the group of skeletal ciliopathies to an additional IFT-B component, IFT172, similar to what has been shown for IFT-A.


Assuntos
Ataxia Cerebelar/genética , Síndrome de Ellis-Van Creveld/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Retinose Pigmentar/genética , Alelos , Sequência de Aminoácidos , Animais , Povo Asiático/genética , Osso e Ossos/anormalidades , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Ataxia Cerebelar/patologia , Craniossinostoses/genética , Craniossinostoses/patologia , Dineínas do Citoplasma/genética , Dineínas do Citoplasma/metabolismo , Dineínas/genética , Dineínas/metabolismo , Displasia Ectodérmica/genética , Displasia Ectodérmica/patologia , Síndrome de Ellis-Van Creveld/patologia , Epistasia Genética , Feminino , Fibroblastos/patologia , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Masculino , Dados de Sequência Molecular , Mutação , Fenótipo , Retinose Pigmentar/patologia , População Branca/genética , Peixe-Zebra/genética
18.
Stem Cells ; 33(1): 196-210, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25183173

RESUMO

Neural stem/progenitor cells (NSCs) in the hippocampus produce new neurons throughout adult life. NSCs are maintained in a state of reversible quiescence and the failure to maintain the quiescent state can result in the premature depletion of the stem cell pool. The epigenetic mechanisms that maintain this quiescent state have not been identified. Using an inducible knockout mouse model, we show that the chromatin remodeling factor chromodomain-helicase-DNA-binding protein 7 (CHD7) is essential for maintaining NSC quiescence. CHD7 inactivation in adult NSCs results in a loss of stem cell quiescence in the hippocampus, a transient increase in cell divisions, followed by a significant decline in neurogenesis. This loss of NSC quiescence is associated with the premature loss of NSCs in middle-aged mice. We find that CHD7 represses the transcription of several positive regulators of cell cycle progression and is required for full induction of the Notch target gene Hes5 in quiescent NSCs. These findings directly link CHD7 to pathways involved in NSC quiescence and identify the first chromatin-remodeling factor with a role in NSC quiescence and maintenance. As CHD7 haplo-insufficiency is associated with a range of cognitive disabilities in CHARGE syndrome, our observations may have implications for understanding the basis of these deficits.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Hipocampo/citologia , Células-Tronco Neurais/citologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , DNA Helicases/biossíntese , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Hipocampo/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia
19.
Circ Res ; 115(1): 23-31, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24748541

RESUMO

RATIONALE: 22q11 deletion syndrome arises from recombination between low-copy repeats on chromosome 22. Typical deletions result in hemizygosity for TBX1 associated with congenital cardiovascular disease. Deletions distal to the typically deleted region result in a similar cardiac phenotype but lack in extracardiac features of the syndrome, suggesting that a second haploinsufficient gene maps to this interval. OBJECTIVE: The transcription factor HIC2 is lost in most distal deletions, as well as in a minority of typical deletions. We used mouse models to test the hypothesis that HIC2 hemizygosity causes congenital heart disease. METHODS AND RESULTS: We created a genetrap mouse allele of Hic2. The genetrap reporter was expressed in the heart throughout the key stages of cardiac morphogenesis. Homozygosity for the genetrap allele was embryonic lethal before embryonic day E10.5, whereas the heterozygous condition exhibited a partially penetrant late lethality. One third of heterozygous embryos had a cardiac phenotype. MRI demonstrated a ventricular septal defect with over-riding aorta. Conditional targeting indicated a requirement for Hic2 within the Nkx2.5+ and Mesp1+ cardiovascular progenitor lineages. Microarray analysis revealed increased expression of Bmp10. CONCLUSIONS: Our results demonstrate a novel role for Hic2 in cardiac development. Hic2 is the first gene within the distal 22q11 interval to have a demonstrated haploinsufficient cardiac phenotype in mice. Together our data suggest that HIC2 haploinsufficiency likely contributes to the cardiac defects seen in distal 22q11 deletion syndrome.


Assuntos
Síndrome da Deleção 22q11/etiologia , Coração/embriologia , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Síndrome da Deleção 22q11/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Cardiopatias Congênitas/etiologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Morfogênese , Mutagênese , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/fisiologia , Proteínas Supressoras de Tumor/genética
20.
Nat Genet ; 39(6): 727-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468754

RESUMO

Jeune asphyxiating thoracic dystrophy, an autosomal recessive chondrodysplasia, often leads to death in infancy because of a severely constricted thoracic cage and respiratory insufficiency; retinal degeneration, cystic renal disease and polydactyly may be complicating features. We show that IFT80 mutations underlie a subset of Jeune asphyxiating thoracic dystrophy cases, establishing the first association of a defective intraflagellar transport (IFT) protein with human disease. Knockdown of ift80 in zebrafish resulted in cystic kidneys, and knockdown in Tetrahymena thermophila produced shortened or absent cilia.


Assuntos
Asfixia/genética , Doenças do Desenvolvimento Ósseo/genética , Proteínas de Transporte/genética , Doenças Renais Císticas/genética , Mutação/genética , Tetrahymena thermophila/genética , Doenças Torácicas/genética , Peixe-Zebra/genética , Animais , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Polidactilia/genética , Tetrahymena thermophila/crescimento & desenvolvimento , Peixe-Zebra/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA