Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
EMBO Mol Med ; 9(12): 1711-1731, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29084757

RESUMO

Defects of CIB2, calcium- and integrin-binding protein 2, have been reported to cause isolated deafness, DFNB48 and Usher syndrome type-IJ, characterized by congenital profound deafness, balance defects and blindness. We report here two new nonsense mutations (pGln12* and pTyr110*) in CIB2 patients displaying nonsyndromic profound hearing loss, with no evidence of vestibular or retinal dysfunction. Also, the generated CIB2-/- mice display an early onset profound deafness and have normal balance and retinal functions. In these mice, the mechanoelectrical transduction currents are totally abolished in the auditory hair cells, whilst they remain unchanged in the vestibular hair cells. The hair bundle morphological abnormalities of CIB2-/- mice, unlike those of mice defective for the other five known USH1 proteins, begin only after birth and lead to regression of the stereocilia and rapid hair-cell death. This essential role of CIB2 in mechanotransduction and cell survival that, we show, is restricted to the cochlea, probably accounts for the presence in CIB2-/- mice and CIB2 patients, unlike in Usher syndrome, of isolated hearing loss without balance and vision deficits.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Surdez/diagnóstico , Células Ciliadas Auditivas Internas/metabolismo , Mecanotransdução Celular/fisiologia , Animais , Limiar Auditivo , Comportamento Animal , Proteínas de Ligação ao Cálcio/deficiência , Sobrevivência Celular , Surdez/genética , Modelos Animais de Doenças , Olho/diagnóstico por imagem , Olho/patologia , Feminino , Células Ciliadas Auditivas Internas/patologia , Humanos , Masculino , Aprendizagem em Labirinto , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linhagem , Polimorfismo de Nucleotídeo Único , Retina/patologia , Retina/fisiologia
2.
J Cell Biol ; 216(6): 1849-1864, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28495838

RESUMO

Usher syndrome type 1 (USH1) causes combined hearing and sight defects, but how mutations in USH1 genes lead to retinal dystrophy in patients remains elusive. The USH1 protein complex is associated with calyceal processes, which are microvilli of unknown function surrounding the base of the photoreceptor outer segment. We show that in Xenopus tropicalis, these processes are connected to the outer-segment membrane by links composed of protocadherin-15 (USH1F protein). Protocadherin-15 deficiency, obtained by a knockdown approach, leads to impaired photoreceptor function and abnormally shaped photoreceptor outer segments. Rod basal outer disks displayed excessive outgrowth, and cone outer segments were curved, with lamellae of heterogeneous sizes, defects also observed upon knockdown of Cdh23, encoding cadherin-23 (USH1D protein). The calyceal processes were virtually absent in cones and displayed markedly reduced F-actin content in rods, suggesting that protocadherin-15-containing links are essential for their development and/or maintenance. We propose that calyceal processes, together with their associated links, control the sizing of rod disks and cone lamellae throughout their daily renewal.


Assuntos
Caderinas/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo da Célula Bastonete/metabolismo , Síndromes de Usher/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Caderinas/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Larva/genética , Larva/metabolismo , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Segmento Externo das Células Fotorreceptoras da Retina/ultraestrutura , Segmento Externo da Célula Bastonete/ultraestrutura , Síndromes de Usher/genética , Síndromes de Usher/patologia , Xenopus/embriologia , Xenopus/genética , Proteínas de Xenopus/genética
3.
Am J Hum Genet ; 98(6): 1266-1270, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27259055

RESUMO

By genetic linkage analysis in a large consanguineous Iranian family with eleven individuals affected by severe to profound congenital deafness, we were able to define a 2.8 Mb critical interval (at chromosome 1p21.2-1p21.1) for an autosomal-recessive nonsyndromic deafness locus (DFNB). Whole-exome sequencing allowed us to identify a CDC14A biallelic nonsense mutation, c.1126C>T (p.Arg376(∗)), which was present in the eight clinically affected individuals still alive. Subsequent screening of 115 unrelated individuals affected by severe or profound congenital deafness of unknown genetic cause led us to identify another CDC14A biallelic nonsense mutation, c.1015C>T (p.Arg339(∗)), in an individual originating from Mauritania. CDC14A encodes a protein tyrosine phosphatase. Immunofluorescence analysis of the protein distribution in the mouse inner ear showed a strong labeling of the hair cells' kinocilia. By using a morpholino strategy to knockdown cdc14a in zebrafish larvae, we found that the length of the kinocilia was reduced in inner-ear hair cells. Therefore, deafness caused by loss-of-function mutations in CDC14A probably arises from a morphogenetic defect of the auditory sensory cells' hair bundles, whose differentiation critically depends on the proper growth of their kinocilium.


Assuntos
Cílios/patologia , Células Ciliadas Auditivas/patologia , Perda Auditiva Neurossensorial/etiologia , Mutação/genética , Monoéster Fosfórico Hidrolases/genética , Índice de Gravidade de Doença , Adulto , Idoso , Animais , Cílios/metabolismo , Feminino , Imunofluorescência , Células Ciliadas Auditivas/enzimologia , Perda Auditiva Neurossensorial/patologia , Humanos , Larva/genética , Larva/crescimento & desenvolvimento , Masculino , Camundongos , Pessoa de Meia-Idade , Linhagem , Proteínas Tirosina Fosfatases , Adulto Jovem , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
4.
J Cell Biol ; 212(2): 231-44, 2016 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-26754646

RESUMO

The precise architecture of hair bundles, the arrays of mechanosensitive microvilli-like stereocilia crowning the auditory hair cells, is essential to hearing. Myosin IIIa, defective in the late-onset deafness form DFNB30, has been proposed to transport espin-1 to the tips of stereocilia, thereby promoting their elongation. We show that Myo3a(-/-)Myo3b(-/-) mice lacking myosin IIIa and myosin IIIb are profoundly deaf, whereas Myo3a-cKO Myo3b(-/-) mice lacking myosin IIIb and losing myosin IIIa postnatally have normal hearing. Myo3a(-/-)Myo3b(-/-) cochlear hair bundles display robust mechanoelectrical transduction currents with normal kinetics but show severe embryonic abnormalities whose features rapidly change. These include abnormally tall and numerous microvilli or stereocilia, ungraded stereocilia bundles, and bundle rounding and closure. Surprisingly, espin-1 is properly targeted to Myo3a(-/-)Myo3b(-/-) stereocilia tips. Our results uncover the critical role that class III myosins play redundantly in hair-bundle morphogenesis; they unexpectedly limit the elongation of stereocilia and of subsequently regressing microvilli, thus contributing to the early hair bundle shaping.


Assuntos
Células Ciliadas Auditivas/fisiologia , Microvilosidades/fisiologia , Cadeias Pesadas de Miosina/fisiologia , Miosina Tipo III/fisiologia , Estereocílios/fisiologia , Sequência de Aminoácidos , Animais , Padronização Corporal , Surdez/genética , Células HEK293 , Células Ciliadas Auditivas/ultraestrutura , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteínas dos Microfilamentos/metabolismo , Microvilosidades/ultraestrutura , Dados de Sequência Molecular , Estereocílios/ultraestrutura
5.
Cell ; 163(4): 894-906, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26544938

RESUMO

A deficiency in pejvakin, a protein of unknown function, causes a strikingly heterogeneous form of human deafness. Pejvakin-deficient (Pjvk(-/-)) mice also exhibit variable auditory phenotypes. Correlation between their hearing thresholds and the number of pups per cage suggest a possible harmful effect of pup vocalizations. Direct sound or electrical stimulation show that the cochlear sensory hair cells and auditory pathway neurons of Pjvk(-/-) mice and patients are exceptionally vulnerable to sound. Subcellular analysis revealed that pejvakin is associated with peroxisomes and required for their oxidative-stress-induced proliferation. Pjvk(-/-) cochleas display features of marked oxidative stress and impaired antioxidant defenses, and peroxisomes in Pjvk(-/-) hair cells show structural abnormalities after the onset of hearing. Noise exposure rapidly upregulates Pjvk cochlear transcription in wild-type mice and triggers peroxisome proliferation in hair cells and primary auditory neurons. Our results reveal that the antioxidant activity of peroxisomes protects the auditory system against noise-induced damage.


Assuntos
Perda Auditiva Provocada por Ruído/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peroxissomos/metabolismo , Proteínas/metabolismo , Animais , Vias Auditivas , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Perda Auditiva Provocada por Ruído/patologia , Humanos , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Estresse Oxidativo , Proteínas/genética
6.
J Cell Biol ; 199(2): 381-99, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23045546

RESUMO

The mechanisms underlying retinal dystrophy in Usher syndrome type I (USH1) remain unknown because mutant mice lacking any of the USH1 proteins-myosin VIIa, harmonin, cadherin-23, protocadherin-15, sans-do not display retinal degeneration. We found here that, in macaque photoreceptor cells, all USH1 proteins colocalized at membrane interfaces (i) between the inner and outer segments in rods and (ii) between the microvillus-like calyceal processes and the outer segment basolateral region in rods and cones. This pattern, conserved in humans and frogs, was mediated by the formation of an USH1 protein network, which was associated with the calyceal processes from the early embryonic stages of outer segment growth onwards. By contrast, mouse photoreceptors lacked calyceal processes and had no USH1 proteins at the inner-outer segment interface. We suggest that USH1 proteins form an adhesion belt around the basolateral region of the photoreceptor outer segment in humans, and that defects in this structure cause the retinal degeneration in USH1 patients.


Assuntos
Junções Intercelulares/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura , Síndromes de Usher/metabolismo , Animais , Anuros , Proteínas Relacionadas a Caderinas , Caderinas/deficiência , Caderinas/genética , Caderinas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Humanos , Junções Intercelulares/ultraestrutura , Macaca fascicularis , Camundongos , Miosina VIIa , Miosinas/deficiência , Miosinas/genética , Miosinas/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Precursores de Proteínas/deficiência , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Retina/metabolismo , Retina/ultraestrutura , Distrofias Retinianas/patologia , Suínos , Síndromes de Usher/patologia
7.
Hum Mol Genet ; 21(17): 3835-44, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22678063

RESUMO

We report a consanguineous Iranian family affected by congenital profound sensorineural deafness segregating in an autosomal recessive mode. Auditory tests implicated at least a cochlear defect in these patients. We mapped the deafness, autosomal recessive (DFNB) locus involved by linkage analysis to a 4.8 Mb region at chromosome 21q22.3-qter. Exclusion of the DFNB8/10 gene TMPRSS3, located in this chromosomal interval, led us to identify a new deafness locus, DFNB98. Whole exome sequencing allowed us to identify a homozygous frame-shifting mutation (c.1726G>T+c.1728delC) in the gene TSPEAR (thrombospondin-type laminin G domain and EAR repeats). This truncating mutation (p.V576LfsX37) impeded the secretion of the encoded protein by cells transfected with the mutated gene. Alternative splicing of TSPEAR transcripts predict two protein isoforms, 522 and 669 amino acids in length, both of which would be affected by the mutation. These isoforms are composed of a thrombospondin-type laminin G (TSP) domain followed by seven tandemly organized epilepsy-associated repeats (EARs), probably forming a ß-propeller domain. Tspear is expressed in a variety of murine tissues. Only the larger Tspear transcript was found in the cochlea, and the protein was detected by immunofluorescence at the surface of the hair bundles of sensory cells. The mammalian EAR protein family includes six known members. Defects in four of them, i.e. Lgi1, Lgi2, Vlgr1 and, we show here, TSPEAR, cause disorders with auditory features: epilepsy, which can include auditory features in humans; audiogenic seizures in animals; and/or hearing impairments in humans and mice. These observations demonstrate that EAR-containing proteins are essential for the development and function of the auditory system.


Assuntos
Surdez/genética , Loci Gênicos/genética , Proteínas/química , Proteínas/genética , Sequências Repetitivas de Aminoácidos/genética , Adulto , Animais , Audiometria , Sequência de Bases , Segregação de Cromossomos/genética , Cromossomos Humanos Par 21/genética , Cóclea/metabolismo , Feminino , Mutação da Fase de Leitura/genética , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Linhagem , Estrutura Terciária de Proteína , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
8.
J Neurosci ; 28(11): 2827-36, 2008 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-18337413

RESUMO

The mitochondrial metalloprotease AFG3L2 assembles with the homologous protein paraplegin to form a supracomplex in charge of the essential protein quality control within mitochondria. Mutations of paraplegin cause a specific axonal degeneration of the upper motoneuron and, therefore, hereditary spastic paraplegia. Here we present two Afg3l2 murine models: a newly developed null and a spontaneous mutant that we found carrier of a missense mutation. Contrasting with the mild and late onset axonal degeneration of paraplegin-deficient mouse, Afg3l2 models display a marked impairment of axonal development with delayed myelination and poor axonal radial growth leading to lethality at P16. The increased severity of the Afg3l2 mutants is explained by two main molecular features that differentiate AFG3L2 from paraplegin: its higher neuronal expression and its versatile ability to support both hetero-oligomerization and homo-oligomerization. Our data assign to AFG3L2 a crucial role by linking mitochondrial metabolism and axonal development. Moreover, we propose AFG3L2 as an excellent candidate for motoneuron and cerebellar diseases with early onset unknown etiology.


Assuntos
Adenosina Trifosfatases/biossíntese , Axônios/enzimologia , Mitocôndrias/enzimologia , Proteínas Mitocondriais/biossíntese , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Axônios/patologia , Axônios/fisiologia , Camundongos , Camundongos Mutantes , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Dados de Sequência Molecular
9.
J Biol Chem ; 283(23): 15638-46, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18364348

RESUMO

Although the D-glucarate degradation pathway is well characterized in Escherichia coli, genetic and biochemical information concerning the alternative pathway proposed in Pseudomonas species and Bacillus subtilis remains incomplete. Acinetobacter baylyi ADP1 is a Gram-negative soil bacterium possessing the alternative pathway and able to grow using D-glucarate as the only carbon source. Based on the annotation of its sequenced genome (1), we have constructed a complete collection of singlegene deletion mutants (2). High throughput profiling for growth on a minimal medium containing D-glucarate as the only carbon source for approximately 2450 mutants led to the identification of the genes involved in D-glucarate degradation. Protein purification after recombinant production in E. coli allowed us to reconstitute the enzymatic pathway in vitro. We describe here the kinetic characterization of D-glucarate dehydratase, d-5-keto-4-deoxyglucarate dehydratase, and of cooperative alpha-ketoglutarate semialdehyde dehydrogenase. Transcription and expression analyses of the genes involved in D-glucarate metabolism within a single organism made it possible to access information regarding the regulation of this pathway for the first time.


Assuntos
Acinetobacter/enzimologia , Proteínas de Bactérias/biossíntese , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Glutaratos/metabolismo , Hidroliases/biossíntese , Acinetobacter/genética , Proteínas de Bactérias/genética , Deleção de Genes , Genoma Bacteriano/fisiologia , Hidroliases/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transcrição Gênica/fisiologia
10.
Int J Mol Med ; 18(4): 593-600, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964410

RESUMO

We describe a novel spontaneous autosomal recessive mutation, cervelet-4 (crv4), which arose in a BALB/c strain. Mice homozygous for the mutation exhibit principally a reduced body size, a congenital neurological phenotype characterized by ataxic gait and intention tremor, with no gross anomalies observed in brain or cerebellum, and skeletal anomalies. Using linkage analysis, we mapped the crv4 locus to the proximal region of chromosome 10, at the location of the Grm1 gene. Genetic complementation crosses between crv4 and Grm1 KO mice confirmed that crv4 is a new allele of Grm1. Molecular analysis of the Grm1 gene in mutant mice revealed the insertion of a 190-bp LTR fragment in intron 4. Our results also indicated that the presence of the LTR fragment caused the disruption of the Grm1 normal splicing process and complete absence of the wild-type protein. crv4 is an interesting model to extend the study of Grm1 function and the pathological effects of Grm1 deficiency in vivo.


Assuntos
Ataxia/genética , Cifose/genética , Mutação/genética , Splicing de RNA , Receptores de Glutamato Metabotrópico/genética , Escoliose/genética , Animais , Ataxia/etiologia , Sequência de Bases , Western Blotting , Mapeamento Cromossômico/métodos , Modelos Animais de Doenças , Feminino , Genótipo , Humanos , Íntrons/genética , Cifose/complicações , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Dados de Sequência Molecular , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Retroelementos/genética , Escoliose/complicações , Homologia de Sequência do Ácido Nucleico
11.
Nat Genet ; 38(7): 770-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16804542

RESUMO

Auditory neuropathy is a particular type of hearing impairment in which neural transmission of the auditory signal is impaired, while cochlear outer hair cells remain functional. Here we report on DFNB59, a newly identified gene on chromosome 2q31.1-q31.3 mutated in four families segregating autosomal recessive auditory neuropathy. DFNB59 encodes pejvakin, a 352-residue protein. Pejvakin is a paralog of DFNA5, a protein of unknown function also involved in deafness. By immunohistofluorescence, pejvakin is detected in the cell bodies of neurons of the afferent auditory pathway. Furthermore, Dfnb59 knock-in mice, homozygous for the R183W variant identified in one DFNB59 family, show abnormal auditory brainstem responses indicative of neuronal dysfunction along the auditory pathway. Unlike previously described sensorineural deafness genes, all of which underlie cochlear cell pathologies, DFNB59 is the first human gene implicated in nonsyndromic deafness due to a neuronal defect.


Assuntos
Vias Auditivas/metabolismo , Perda Auditiva Neurossensorial/genética , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Sequência de Aminoácidos , Animais , Vias Auditivas/patologia , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Humanos Par 2/genética , DNA/genética , Orelha Interna/metabolismo , Orelha Interna/patologia , Feminino , Genes Recessivos , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Linhagem
12.
Genet. mol. biol ; 28(2): 201-204, 2005. ilus
Artigo em Inglês | LILACS | ID: lil-416284

RESUMO

The Paralysé mutation is a spontaneous neuromuscular mutation, first observed in 1980 at the Pasteur Institute, which is transmitted by the autosomal recessive par allele. Affected homozygote par/par mice rarely survive beyond 16 days of age and at the end of their life they are emaciated and completely paralyzed. Several concordant histological and physiological observations indicate that mutant mice might be good models for studying early-onset human motor neuron diseases such as spinal muscular atrophy. Linkage analysis using a set of molecular markers and two F2 crosses indicate that the mutation maps to mouse chromosome 18 in a region spanning 4 cM (or 9 megabase pairs, Mbp) between the microsatellites D18Mit140 and D18Mit33. These results positioned the par locus in a region homologous to human chromosome 18p11.22 to 18q21.32.


Assuntos
Camundongos , Doenças Neuromusculares , Camundongos
13.
Eur J Hum Genet ; 11(10): 816-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14512974

RESUMO

We report on a novel localization for a recessive form of deafness (DFNB), by linkage analysis in an Iranian consanguineous family. Affected individuals suffer from prelingual profound sensorineural hearing loss. Genome-wide analysis led to the characterization of a new locus, DFNB40, which maps to an approximately 9 Mb interval between markers D22S427 and D22S1144 at chromosome 22q11.21-12.1. Maximum lod score of 3.09 was obtained with D22S1174. Since the Bronx waltzer (bv) mouse mutant, characterized by waltzing behavior, deafness, and degeneration of cochlear inner hair cells, has been mapped to the syntenic region on murine chromosome 5, we suggest that DFNB40 and bv may result from orthologous gene defects.


Assuntos
Cromossomos Humanos Par 22 , Genes Recessivos , Perda Auditiva Neurossensorial/genética , Adolescente , Adulto , Criança , Mapeamento Cromossômico , Feminino , Marcadores Genéticos , Genótipo , Humanos , Escore Lod , Masculino , Mutação , Linhagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...