Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 6(33): eabb3567, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32851175

RESUMO

Switches between global sleep and wakefulness states are believed to be dictated by top-down influences arising from subcortical nuclei. Using forward genetics and in vivo electrophysiology, we identified a recessive mouse mutant line characterized by a substantially reduced propensity to transition between wake and sleep states with an especially pronounced deficit in initiating rapid eye movement (REM) sleep episodes. The causative mutation, an Ile102Asn substitution in the synaptic vesicular protein, VAMP2, was associated with morphological synaptic changes and specific behavioral deficits, while in vitro electrophysiological investigations with fluorescence imaging revealed a markedly diminished probability of vesicular release in mutants. Our data show that global shifts in the synaptic efficiency across brain-wide networks leads to an altered probability of vigilance state transitions, possibly as a result of an altered excitability balance within local circuits controlling sleep-wake architecture.


Assuntos
Sono REM , Sono , Animais , Encéfalo/fisiologia , Fenômenos Eletrofisiológicos , Camundongos , Sono/genética , Sono REM/genética , Vigília/genética
2.
Nat Commun ; 10(1): 4072, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31492868

RESUMO

The human PKD2 locus encodes Polycystin-2 (PC2), a TRPP channel that localises to several distinct cellular compartments, including the cilium. PKD2 mutations cause Autosomal Dominant Polycystic Kidney Disease (ADPKD) and affect many cellular pathways. Data underlining the importance of ciliary PC2 localisation in preventing PKD are limited because PC2 function is ablated throughout the cell in existing model systems. Here, we dissect the ciliary role of PC2 by analysing mice carrying a non-ciliary localising, yet channel-functional, PC2 mutation. Mutants develop embryonic renal cysts that appear indistinguishable from mice completely lacking PC2. Despite not entering the cilium in mutant cells, mutant PC2 accumulates at the ciliary base, forming a ring pattern consistent with distal appendage localisation. This suggests a two-step model of ciliary entry; PC2 first traffics to the cilium base before TOP domain dependent entry. Our results suggest that PC2 localisation to the cilium is necessary to prevent PKD.


Assuntos
Cílios/metabolismo , Rim/patologia , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Modelos Animais de Doenças , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/metabolismo , Glicosilação , Humanos , Rim/embriologia , Masculino , Camundongos Endogâmicos C57BL , Mutação/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Cátion TRPP/genética
3.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 378-390, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30414501

RESUMO

Autosomal Recessive Polycystic Kidney Disease (ARPKD) is a genetic disorder with an incidence of ~1:20,000 that manifests in a wide range of renal and liver disease severity in human patients and can lead to perinatal mortality. ARPKD is caused by mutations in PKHD1, which encodes the large membrane protein, Fibrocystin, required for normal branching morphogenesis of the ureteric bud during embryonic renal development. The variation in ARPKD phenotype suggests that in addition to PKHD1 mutations, other genes may play a role, acting as modifiers of disease severity. One such pathway involves non-canonical Wnt/Planar Cell Polarity (PCP) signalling that has been associated with other cystic kidney diseases, but has not been investigated in ARPKD. Analysis of the AtminGpg6 mouse showed kidney, liver and lung abnormalities, suggesting it as a novel mouse tool for the study of ARPKD. Further, modulation of Atmin affected Pkhd1 mRNA levels, altered non-canonical Wnt/PCP signalling and impacted cellular proliferation and adhesion, although Atmin does not bind directly to the C-terminus of Fibrocystin. Differences in ATMIN and VANGL2 expression were observed between normal human paediatric kidneys and age-matched ARPKD kidneys. Significant increases in ATMIN, WNT5A, VANGL2 and SCRIBBLE were seen in human ARPKD versus normal kidneys; no substantial differences were seen in DAAM2 or NPHP2. A striking increase in E-cadherin was also detected in ARPKD kidneys. This work indicates a novel role for non-canonical Wnt/PCP signalling in ARPKD and suggests ATMIN as a modulator of PKHD1.


Assuntos
Polaridade Celular , Rim Policístico Autossômico Recessivo/patologia , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Adolescente , Apoptose , Caderinas/metabolismo , Adesão Celular , Linhagem Celular , Proliferação de Células , Criança , Pré-Escolar , Citoesqueleto/metabolismo , Embrião de Mamíferos/metabolismo , Humanos , Lactente , Recém-Nascido , Túbulos Renais Coletores , Fenótipo , Rim Policístico Autossômico Recessivo/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , beta Catenina/metabolismo
4.
Cell Rep ; 25(12): 3315-3328.e6, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566859

RESUMO

Mutations in genes essential for mitochondrial function have pleiotropic effects. The mechanisms underlying these traits yield insights into metabolic homeostasis and potential therapies. Here we report the characterization of a mouse model harboring a mutation in the tryptophanyl-tRNA synthetase 2 (Wars2) gene, encoding the mitochondrial-localized WARS2 protein. This hypomorphic allele causes progressive tissue-specific pathologies, including hearing loss, reduced adiposity, adipose tissue dysfunction, and hypertrophic cardiomyopathy. We demonstrate the tissue heterogeneity arises as a result of variable activation of the integrated stress response (ISR) pathway and the ability of certain tissues to respond to impaired mitochondrial translation. Many of the systemic metabolic effects are likely mediated through elevated fibroblast growth factor 21 (FGF21) following activation of the ISR in certain tissues. These findings demonstrate the potential pleiotropy associated with Wars2 mutations in patients.


Assuntos
Especificidade de Órgãos , Fosforilação Oxidativa , Estresse Fisiológico , Triptofano-tRNA Ligase/genética , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/patologia , Adiposidade , Alelos , Processamento Alternativo/genética , Animais , Sequência de Bases , Peso Corporal , Encéfalo/patologia , Cardiomiopatia Hipertrófica/sangue , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/fisiopatologia , Modelos Animais de Doenças , Transporte de Elétrons , Potenciais Evocados Auditivos do Tronco Encefálico , Éxons/genética , Fatores de Crescimento de Fibroblastos/sangue , Fibroblastos/metabolismo , Perda Auditiva/sangue , Perda Auditiva/complicações , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Camundongos , Camundongos Mutantes , Músculo Esquelético/metabolismo , Mutação/genética , Biogênese de Organelas , Triptofano-tRNA Ligase/metabolismo , Regulação para Cima
5.
Nat Genet ; 47(9): 969-978, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26214591

RESUMO

The function of the majority of genes in the mouse and human genomes remains unknown. The mouse embryonic stem cell knockout resource provides a basis for the characterization of relationships between genes and phenotypes. The EUMODIC consortium developed and validated robust methodologies for the broad-based phenotyping of knockouts through a pipeline comprising 20 disease-oriented platforms. We developed new statistical methods for pipeline design and data analysis aimed at detecting reproducible phenotypes with high power. We acquired phenotype data from 449 mutant alleles, representing 320 unique genes, of which half had no previous functional annotation. We captured data from over 27,000 mice, finding that 83% of the mutant lines are phenodeviant, with 65% demonstrating pleiotropy. Surprisingly, we found significant differences in phenotype annotation according to zygosity. New phenotypes were uncovered for many genes with previously unknown function, providing a powerful basis for hypothesis generation and further investigation in diverse systems.


Assuntos
Estudos de Associação Genética , Animais , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Anotação de Sequência Molecular , Mutação , Fenótipo
6.
Development ; 141(20): 3966-77, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25294941

RESUMO

Initially identified in DNA damage repair, ATM-interactor (ATMIN) further functions as a transcriptional regulator of lung morphogenesis. Here we analyse three mouse mutants, Atmin(gpg6/gpg6), Atmin(H210Q/H210Q) and Dynll1(GT/GT), revealing how ATMIN and its transcriptional target dynein light chain LC8-type 1 (DYNLL1) are required for normal lung morphogenesis and ciliogenesis. Expression screening of ciliogenic genes confirmed Dynll1 to be controlled by ATMIN and further revealed moderately altered expression of known intraflagellar transport (IFT) protein-encoding loci in Atmin mutant embryos. Significantly, Dynll1(GT/GT) embryonic cilia exhibited shortening and bulging, highly similar to the characterised retrograde IFT phenotype of Dync2h1. Depletion of ATMIN or DYNLL1 in cultured cells recapitulated the in vivo ciliogenesis phenotypes and expression of DYNLL1 or the related DYNLL2 rescued the effects of loss of ATMIN, demonstrating that ATMIN primarily promotes ciliogenesis by regulating Dynll1 expression. Furthermore, DYNLL1 as well as DYNLL2 localised to cilia in puncta, consistent with IFT particles, and physically interacted with WDR34, a mammalian homologue of the Chlamydomonas cytoplasmic dynein 2 intermediate chain that also localised to the cilium. This study extends the established Atmin-Dynll1 relationship into a developmental and a ciliary context, uncovering a novel series of interactions between DYNLL1, WDR34 and ATMIN. This identifies potential novel components of cytoplasmic dynein 2 and furthermore provides fresh insights into the molecular pathogenesis of human skeletal ciliopathies.


Assuntos
Cílios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , Fatores de Transcrição/fisiologia , Animais , Chlamydomonas/metabolismo , Cílios/metabolismo , Dineínas do Citoplasma , Dano ao DNA , Dineínas/metabolismo , Marcadores Genéticos , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Mutação , Fenótipo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica
7.
PLoS One ; 9(6): e100447, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24956260

RESUMO

The secreted molecule fibroblast growth factor 9 (FGF9) plays a critical role in testis determination in the mouse. In embryonic gonadal somatic cells it is required for maintenance of SOX9 expression, a key determinant of Sertoli cell fate. Conditional gene targeting studies have identified FGFR2 as the main gonadal receptor for FGF9 during sex determination. However, such studies can be complicated by inefficient and variable deletion of floxed alleles, depending on the choice of Cre deleter strain. Here, we report a novel, constitutive allele of Fgfr2, hobbyhorse (hob), which was identified in an ENU-based forward genetic screen for novel testis-determining loci. Fgr2hob is caused by a C to T mutation in the invariant exon 7, resulting in a polypeptide with a mis-sense mutation at position 263 (Pro263Ser) in the third extracellular immunoglobulin-like domain of FGFR2. Mutant homozygous embryos show severe limb and lung defects and, when on the sensitised C57BL/6J (B6) genetic background, undergo complete XY gonadal sex reversal associated with failure to maintain expression of Sox9. Genetic crosses employing a null mutant of Fgfr2 suggest that Fgr2hob is a hypomorphic allele, affecting both the FGFR2b and FGFR2c splice isoforms of the receptor. We exploited the consistent phenotype of this constitutive mutant by analysing MAPK signalling at the sex-determining stage of gonad development, but no significant abnormalities in mutant embryos were detected.


Assuntos
Extremidades/patologia , Disgenesia Gonadal/genética , Gônadas/anormalidades , Pneumopatias/patologia , Mutação/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Gônadas/metabolismo , Gônadas/patologia , Homozigoto , Humanos , Técnicas Imunoenzimáticas , Pneumopatias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fenótipo , Homologia de Sequência de Aminoácidos
8.
J Clin Invest ; 124(4): 1468-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24614104

RESUMO

The recent identification of multiple dominant mutations in the gene encoding ß-catenin in both humans and mice has enabled exploration of the molecular and cellular basis of ß-catenin function in cognitive impairment. In humans, ß-catenin mutations that cause a spectrum of neurodevelopmental disorders have been identified. We identified de novo ß-catenin mutations in patients with intellectual disability, carefully characterized their phenotypes, and were able to define a recognizable intellectual disability syndrome. In parallel, characterization of a chemically mutagenized mouse line that displays features similar to those of human patients with ß-catenin mutations enabled us to investigate the consequences of ß-catenin dysfunction through development and into adulthood. The mouse mutant, designated batface (Bfc), carries a Thr653Lys substitution in the C-terminal armadillo repeat of ß-catenin and displayed a reduced affinity for membrane-associated cadherins. In association with this decreased cadherin interaction, we found that the mutation results in decreased intrahemispheric connections, with deficits in dendritic branching, long-term potentiation, and cognitive function. Our study provides in vivo evidence that dominant mutations in ß-catenin underlie losses in its adhesion-related functions, which leads to severe consequences, including intellectual disability, childhood hypotonia, progressive spasticity of lower limbs, and abnormal craniofacial features in adults.


Assuntos
Anormalidades Craniofaciais/genética , Deficiência Intelectual/genética , Mutação , beta Catenina/genética , Adolescente , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Encéfalo/patologia , Caderinas/química , Pré-Escolar , Anormalidades Craniofaciais/patologia , DNA/genética , Modelos Animais de Doenças , Feminino , Genes Dominantes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Fenótipo , Homologia de Sequência de Aminoácidos , Síndrome , Adulto Jovem , beta Catenina/química , beta Catenina/metabolismo
9.
PLoS Genet ; 7(4): e1001372, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21533022

RESUMO

Osteoporotic fracture is a major cause of morbidity and mortality worldwide. Low bone mineral density (BMD) is a major predisposing factor to fracture and is known to be highly heritable. Site-, gender-, and age-specific genetic effects on BMD are thought to be significant, but have largely not been considered in the design of genome-wide association studies (GWAS) of BMD to date. We report here a GWAS using a novel study design focusing on women of a specific age (postmenopausal women, age 55-85 years), with either extreme high or low hip BMD (age- and gender-adjusted BMD z-scores of +1.5 to +4.0, n = 1055, or -4.0 to -1.5, n = 900), with replication in cohorts of women drawn from the general population (n = 20,898). The study replicates 21 of 26 known BMD-associated genes. Additionally, we report suggestive association of a further six new genetic associations in or around the genes CLCN7, GALNT3, IBSP, LTBP3, RSPO3, and SOX4, with replication in two independent datasets. A novel mouse model with a loss-of-function mutation in GALNT3 is also reported, which has high bone mass, supporting the involvement of this gene in BMD determination. In addition to identifying further genes associated with BMD, this study confirms the efficiency of extreme-truncate selection designs for quantitative trait association studies.


Assuntos
Densidade Óssea , Fraturas Ósseas/genética , Estudo de Associação Genômica Ampla , N-Acetilgalactosaminiltransferases/genética , Osteoporose Pós-Menopausa/genética , Trombospondinas/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Estudos de Casos e Controles , Canais de Cloreto/genética , Cromossomos Humanos/genética , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Genótipo , Humanos , Sialoproteína de Ligação à Integrina/genética , Proteínas de Ligação a TGF-beta Latente/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Animais , Mutação , Polimorfismo de Nucleotídeo Único , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fatores de Transcrição SOXC/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
10.
Dev Biol ; 307(1): 62-78, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17512925

RESUMO

In the developing CNS alpha- and beta-dystroglycan are highly concentrated in the endfeet of radial neuroepithelial cells at the contact site to the basal lamina. We show that injection of anti-dystroglycan Fab fragments, knockdown of dystroglycan using RNAi, and overexpression of a dominant-negative dystroglycan protein by microelectroporation in neuroepithelial cells of the chick retina and optic tectum in vivo leads to the loss of their radial morphology, to hyperproliferation, to an increased number of postmitotic neurons, and to an altered distribution of several basally concentrated proteins. Moreover, these treatments also altered the oriented growth of axons from retinal ganglion cells and from tectal projection neurons. In contrast, expression of non-cleavable dystroglycan protein in neuroepithelial cells reduced their proliferation and their differentiation to postmitotic neurons. These results demonstrate that dystroglycan plays a key role in maintaining neuroepithelial cell morphology, and that interfering with dystroglycan function influences proliferation and differentiation of neuroepithelial cells. These data also suggest that an impaired dystroglycan function in neuroepithelial cells might be responsible for some of the severe brain abnormalities observed in certain forms of congenital muscular dystrophy.


Assuntos
Sistema Nervoso Central/crescimento & desenvolvimento , Distroglicanas/fisiologia , Células Neuroepiteliais/citologia , Animais , Diferenciação Celular , Proliferação de Células , Forma Celular , Galinhas , Distrofias Musculares/fisiopatologia , Retina/citologia , Colículos Superiores/citologia , Vertebrados
11.
FEBS Lett ; 555(2): 209-16, 2003 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-14644417

RESUMO

Dystroglycan is a component of the dystrophin glycoprotein complex that is cleaved into two polypeptides by an unidentified protease. To determine the role of post-translational processing on dystroglycan synthesis and trafficking we expressed the dystroglycan precursor and mutants thereof in a heterologous system. A point mutant in the processing site, S655A, prevented proteolytic cleavage but had no effect upon the surface localisation of dystroglycan. Mutation of two N-linked glycosylation sites that flank the cleavage site inhibited proteolytic processing of the precursor. Furthermore, chemical inhibition of N- and O-linked glycosylation interfered with the processing of the precursor and reduced the levels of dystroglycan at the cell surface. Dystroglycan processing was also inhibited by the proteasome inhibitor lactacystin. N-linked glycosylation is a prerequisite for efficient proteolytic processing and cleavage and glycosylation are dispensable for cell surface targeting of dystroglycan.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Glicoproteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Transporte Biológico , Células CHO , Células COS , Linhagem Celular , Galinhas , Chlorocebus aethiops , Cricetinae , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Distroglicanas , Inibidores Enzimáticos/farmacologia , Glicosilação/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Peso Molecular , Mutação Puntual , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Alinhamento de Sequência , Transfecção
12.
Hum Mol Genet ; 11(26): 3319-31, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12471058

RESUMO

Two forms of congenital muscular dystrophy (CMD), Fukuyama CMD and CMD type 1C (MDC1C) are caused by mutations in the genes encoding two putative glycosyltransferases, fukutin and fukutin-related protein (FKRP). Additionally, mutations in the FKRP gene also cause limb-girdle muscular dystrophy type 2I (LGMD2I), a considerably milder allelic variant than MDC1C. All of these diseases are associated with secondary changes in muscle alpha-dystroglycan expression. To elucidate the function of FKRP and fukutin and examine the effects of MDC1C patient mutations, we have determined the mechanism for the subcellular location of each protein. FKRP and fukutin are targeted to the medial-Golgi apparatus through their N-termini and transmembrane domains. Overexpression of FKRP in CHO cells alters the post-translational processing of alpha- and beta-dystroglycan inhibiting maturation of the two isoforms. Mutations in the DxD motif in the putative active site of the protein or in the Golgi-targeting sequence, which cause FKRP to be inefficiently trafficked to the Golgi apparatus, did not alter dystroglycan processing in vitro. The P448L mutation in FKRP that causes congenital muscular dystrophy changes a conserved amino acid resulting in the mislocalization of the mutant protein in the cell that is unable to alter dystroglycan processing. Our data show that FKRP and fukutin are Golgi-resident proteins and that FKRP is required for the post-translational modification of dystroglycan. Aberrant processing of dystroglycan caused by a mislocalized FKRP mutant could be a novel mechanism that causes congenital muscular dystrophy.


Assuntos
Complexo de Golgi/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas do Citoesqueleto/metabolismo , Distroglicanas , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutação , Pentosiltransferases , Proteínas/genética , Transferases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...