Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Toxicol Lett ; 349: 115-123, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34089817

RESUMO

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.


Assuntos
Inibidores Enzimáticos/farmacologia , Células Ciliadas Auditivas/efeitos dos fármacos , Perda Auditiva/prevenção & controle , Audição/efeitos dos fármacos , NAD/biossíntese , Ototoxicidade/prevenção & controle , Sirtuína 1/metabolismo , Animais , Animais Geneticamente Modificados , Carboxiliases/antagonistas & inibidores , Carboxiliases/metabolismo , Cisplatino , Modelos Animais de Doenças , Ativação Enzimática , Células Ciliadas Auditivas/enzimologia , Células Ciliadas Auditivas/patologia , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Sistema da Linha Lateral/efeitos dos fármacos , Sistema da Linha Lateral/enzimologia , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Ototoxicidade/enzimologia , Ototoxicidade/etiologia , Ototoxicidade/fisiopatologia , Peixe-Zebra
2.
Nat Commun ; 10(1): 4150, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515474

RESUMO

Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.


Assuntos
Cisplatino/efeitos adversos , Glutationa Transferase/metabolismo , Ototoxicidade/enzimologia , Animais , Limiar Auditivo/efeitos dos fármacos , Capilares/patologia , Cóclea/enzimologia , Cóclea/patologia , Cóclea/fisiopatologia , Cruzamentos Genéticos , Dano ao DNA/genética , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Transferase/deficiência , Perda Auditiva/complicações , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Masculino , Camundongos Endogâmicos CBA , Ototoxicidade/complicações , Ototoxicidade/patologia , Ototoxicidade/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia
3.
Nat Genet ; 47(3): 263-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25665007

RESUMO

Taking a genome-wide association study approach, we identified inherited genetic variations in ACYP2 associated with cisplatin-related ototoxicity (rs1872328: P = 3.9 × 10(-8), hazard ratio = 4.5) in 238 children with newly diagnosed brain tumors, with independent replication in 68 similarly treated children. The ACYP2 risk variant strongly predisposed these patients to precipitous hearing loss and was related to ototoxicity severity. These results point to new biology underlying the ototoxic effects of platinum agents.


Assuntos
Hidrolases Anidrido Ácido/genética , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Perda Auditiva/induzido quimicamente , Perda Auditiva/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Pré-Escolar , Ensaios Clínicos como Assunto , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Perda Auditiva/enzimologia , Humanos , Lactente , Masculino , Polimorfismo de Nucleotídeo Único , Acilfosfatase
4.
Otol Neurotol ; 35(5): e169-77, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24608370

RESUMO

HYPOTHESIS: To investigate whether curcumin may have in vivo protective effects against cisplatin ototoxicity by its direct scavenger activity and/or by curcumin-mediated upregulation of HO-1. BACKGROUND: Cisplatin-induced ototoxicity is a major dose-limiting side effect in anticancer chemotherapy. A protective approach to decrease cisplatin ototoxicity without compromising its therapeutic efficacy remains a critical goal for anticancer therapy. Recent evidences indicate that curcumin exhibits antioxidant, anti-inflammatory, and chemosensitizer activities. METHODS: In male adult Wistar rats, a curcumin dose of 200 mg/kg, selected from a dose-response curve, was injected 1 hour before cisplatin administration and once daily for the following 3 days. A single dose of cisplatin (16 mg/kg) was administered intraperitoneally. Rats were divided as follows: 1) control, 2) curcumin control, 3) vehicle control, 4) cisplatin, 5) cisplatin+ vehicle, and 6) curcumin+cisplatin. ABRs were measured before and at Days 3 and 5 after cisplatin administration. Rhodamine-phalloidin staining, 4-hydroxy-2-nonenal and heme-oxigenase-1 immunostainings, and Western blot analyses were performed to assess and quantify OHC loss, lipid peroxidation, and the endogenous response to cisplatin-induced damage and to curcumin protection. RESULTS: Curcumin treatment attenuated hearing loss induced by cisplatin, increased OHC survival, decreased 4-HNE expression, and increased HO-1 expression. CONCLUSION: This preclinical study demonstrates that systemic curcumin attenuates ototoxicity and provides molecular evidence for a role of HO-1 as an additional mediator in attenuating cisplatin-induced damage.


Assuntos
Curcumina/uso terapêutico , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Perda Auditiva/tratamento farmacológico , Heme Oxigenase-1/metabolismo , Animais , Cisplatino , Curcumina/farmacologia , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Ratos , Ratos Wistar
5.
Hum Mol Genet ; 23(12): 3289-98, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24482543

RESUMO

Cyclic AMP (cAMP) production, which is important for mechanotransduction within the inner ear, is catalyzed by adenylate cyclases (AC). However, knowledge of the role of ACs in hearing is limited. Previously, a novel autosomal recessive non-syndromic hearing impairment locus DFNB44 was mapped to chromosome 7p14.1-q11.22 in a consanguineous family from Pakistan. Through whole-exome sequencing of DNA samples from hearing-impaired family members, a nonsense mutation c.3112C>T (p.Arg1038*) within adenylate cyclase 1 (ADCY1) was identified. This stop-gained mutation segregated with hearing impairment within the family and was not identified in ethnically matched controls or within variant databases. This mutation is predicted to cause the loss of 82 amino acids from the carboxyl tail, including highly conserved residues within the catalytic domain, plus a calmodulin-stimulation defect, both of which are expected to decrease enzymatic efficiency. Individuals who are homozygous for this mutation had symmetric, mild-to-moderate mixed hearing impairment. Zebrafish adcy1b morphants had no FM1-43 dye uptake and lacked startle response, indicating hair cell dysfunction and gross hearing impairment. In the mouse, Adcy1 expression was observed throughout inner ear development and maturation. ADCY1 was localized to the cytoplasm of supporting cells and hair cells of the cochlea and vestibule and also to cochlear hair cell nuclei and stereocilia. Ex vivo studies in COS-7 cells suggest that the carboxyl tail of ADCY1 is essential for localization to actin-based microvilli. These results demonstrate that ADCY1 has an evolutionarily conserved role in hearing and that cAMP signaling is important to hair cell function within the inner ear.


Assuntos
Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/patologia , Adenilil Ciclases/química , Animais , Células COS , Chlorocebus aethiops , Códon sem Sentido , Citoplasma/metabolismo , Orelha Interna/crescimento & desenvolvimento , Feminino , Perda Auditiva/enzimologia , Humanos , Células Labirínticas de Suporte/metabolismo , Masculino , Camundongos , Peixe-Zebra/genética
6.
Dis Model Mech ; 6(2): 434-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23065636

RESUMO

Mutations in the ATP6V0A4 gene lead to autosomal recessive distal renal tubular acidosis in patients, who often show sensorineural hearing impairment. A first Atp6v0a4 knockout mouse model that recapitulates the loss of H(+)-ATPase function seen in humans has been generated and recently reported (Norgett et al., 2012). Here, we present the first detailed analysis of the structure and function of the auditory system in Atp6v0a4(-/-) knockout mice. Measurements of the auditory brainstem response (ABR) showed significantly elevated thresholds in homozygous mutant mice, which indicate severe hearing impairment. Heterozygote thresholds were normal. Analysis of paint-filled inner ears and sections from E16.5 embryos revealed a marked expansion of cochlear and endolymphatic ducts in Atp6v0a4(-/-) mice. A regulatory link between Atp6v0a4, Foxi1 and Pds has been reported and we found that the endolymphatic sac of Atp6v0a4(-/-) mice expresses both Foxi1 and Pds, which suggests a downstream position of Atp6v0a4. These mutants also showed a lack of endocochlear potential, suggesting a functional defect of the stria vascularis on the lateral wall of the cochlear duct. However, the main K(+) channels involved in the generation of endocochlear potential, Kcnj10 and Kcnq1, are strongly expressed in Atp6v0a4(-/-) mice. Our results lead to a better understanding of the role of this proton pump in hearing function.


Assuntos
Orelha Interna/enzimologia , Orelha Interna/patologia , Endolinfa/enzimologia , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Subunidades Proteicas/deficiência , ATPases Translocadoras de Prótons/deficiência , Animais , Animais Recém-Nascidos , Proteínas de Transporte de Ânions/metabolismo , Orelha Interna/fisiopatologia , Saco Endolinfático/patologia , Saco Endolinfático/fisiopatologia , Epitélio/metabolismo , Epitélio/patologia , Potenciais Evocados Auditivos , Fatores de Transcrição Forkhead/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , Células Ciliadas Auditivas Externas/ultraestrutura , Perda Auditiva/fisiopatologia , Humanos , Canal de Potássio KCNQ1/metabolismo , Camundongos , Camundongos Knockout , Mutação/genética , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Subunidades Proteicas/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Estria Vascular/metabolismo , Estria Vascular/patologia , Transportadores de Sulfato , ATPases Vacuolares Próton-Translocadoras
7.
Int J Audiol ; 52(1): 23-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23190330

RESUMO

OBJECTIVE: The purpose of this review was to evaluate the current literature on phosphoribosylpyrophosphate synthetase 1 (PRPS1)-related diseases and their consequences on hearing function. DESIGN: A literature search of peer-reviewed, published journal articles was conducted in online bibliographic databases. STUDY SAMPLE: Three databases for medical research were included in this review. RESULTS: Mutations in PRPS1 are associated with a spectrum of non-syndromic to syndromic hearing loss. Hearing loss in male patients with PRPS1 mutations is bilateral, moderate to profound, and can be prelingual or postlingual, progressive or non-progressive. Audiogram shapes associated with PRPS1 deafness are usually residual and flat. Female carriers can have unilateral or bilateral hearing impairment. Gain of function mutations in PRPS1 cause a superactivity of the PRS-I protein whereas the loss-of-function mutations result in X-linked nonsyndromic sensorineural deafness type 2 (DFN2), or in syndromic deafness including Arts syndrome and X-linked Charcot-Marie-Tooth disease-5 (CMTX5). CONCLUSIONS: Lower residual activity in PRS-I leads to a more severe clinical manifestation. Clinical and molecular findings suggest that the four PRPS1 disorders discovered to date belong to the same disease spectrum. Dietary supplementation with S-adenosylmethionine (SAM) appeared to alleviate the symptoms of Arts syndrome patients, suggesting that SAM could compensate for PRS-I deficiency.


Assuntos
Perda Auditiva/genética , Audição/genética , Mutação , Ribose-Fosfato Pirofosfoquinase/genética , Suplementos Nutricionais , Feminino , Predisposição Genética para Doença , Perda Auditiva/diagnóstico , Perda Auditiva/tratamento farmacológico , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Hereditariedade , Humanos , Masculino , Fenótipo , S-Adenosilmetionina/uso terapêutico , Índice de Gravidade de Doença , Fatores Sexuais
8.
Clin Genet ; 82(1): 56-63, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21534946

RESUMO

Mutations in the TMPRSS3 gene are known to cause autosomal recessive non-syndromic hearing impairment (ARNSHI). After undergoing a genome scan, 10 consanguineous Pakistani families with ARNSHI were found to have significant or suggestive evidence of linkage to the TMPRSS3 region. In order to elucidate if the TMPRSS3 gene is responsible for ARNSHI in these families, the gene was sequenced using DNA samples from these families. Six TMPRSS3 variants were found to cosegregate in 10 families. None of these variants were detected in 500 control chromosomes. Four novel variants, three of which are missense [c.310G>A (p.Glu104Lys), c.767C>T (p.Ala256Val) and c.1273T>C (p.Cys425Arg)] and one nonsense [c.310G>T (p.Glu104Stop)], were identified. The pathogenicity of novel missense variants was investigated through bioinformatics analyses. Additionally, the previously reported deletion c.208delC (p.His70ThrfsX19) was identified in one family and the known mutation c.1219T>C (p.Cys407Arg) was found in five families, which makes c.1219T>C (p.Cys407Arg) as the most common TMPRSS3 mutation within the Pakistani population. Identification of these novel variants lends support to the importance of elements within the low-density lipoprotein receptor A (LDLRA) and serine protease domains in structural stability, ligand binding and proteolytic activity for proper TMPRSS3 function within the inner ear.


Assuntos
Orelha Interna/patologia , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Estudos de Casos e Controles , Cromossomos Humanos Par 21 , Consanguinidade , Orelha Interna/enzimologia , Éxons , Feminino , Genes Recessivos , Ligação Genética , Loci Gênicos , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Humanos , Masculino , Modelos Moleculares , Mutação , Linhagem , Fenótipo , Estrutura Terciária de Proteína
9.
Proc Natl Acad Sci U S A ; 107(29): 13051-6, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20616061

RESUMO

A significantly increased risk for dominant sensorineural deafness in patients who have Hirschsprung disease (HSCR) caused by endothelin receptor type B and SOX10 has been reported. Despite the fact that c-RET is the most frequent causal gene of HSCR, it has not been determined whether impairments of c-Ret and c-RET cause congenital deafness in mice and humans. Here, we show that impaired phosphorylation of c-Ret at tyrosine 1062 causes HSCR-linked syndromic congenital deafness in c-Ret knockin (KI) mice. The deafness involves neurodegeneration of spiral ganglion neurons (SGNs) with not only impaired phosphorylation of Akt and NF-kappaB but decreased expression of calbindin D28k in inner ears. The congenital deafness involving neurodegeneration of SGNs in c-Ret KI mice was rescued by introducing constitutively activated RET. Taken together with our results for three patients with congenital deafness with c-RET-mediated severe HSCR, our results indicate that c-Ret and c-RET are a deafness-related molecule in mice and humans.


Assuntos
Perda Auditiva/complicações , Perda Auditiva/enzimologia , Doença de Hirschsprung/complicações , Doença de Hirschsprung/enzimologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Substituição de Aminoácidos/genética , Animais , Surdez/complicações , Surdez/enzimologia , Ativação Enzimática , Técnicas de Introdução de Genes , Perda Auditiva/congênito , Imuno-Histoquímica , Camundongos , Mutação/genética , NF-kappa B/metabolismo , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Neurônios/enzimologia , Neurônios/patologia , Neurônios/ultraestrutura , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Gânglio Espiral da Cóclea/enzimologia , Gânglio Espiral da Cóclea/patologia , Gânglio Espiral da Cóclea/ultraestrutura
10.
J Med Genet ; 47(9): 643-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20472657

RESUMO

BACKGROUND: Moderate to severe prelingual hearing impairment (DFNB84) was observed in an extended consanguineous Palestinian kindred. All affected relatives shared a 12.5 MB homozygous haplotype on chromosome 12q21 with lod score 4.30. This homozygous region harbours the protein tyrosine phosphatase receptor Q gene PTPRQ, which is known to be essential to hearing in mouse. METHODS: Candidate genes in the 12.5 MB homozygous region were characterized genomically and sequenced in deaf and hearing relatives in the family. RESULTS: Sequence of PTPRQ in affected individuals in the extended kindred revealed c.1285C-->T, leading to p.Gln429Stop. This nonsense mutation co-segregated with hearing loss in the family and was homozygous in all affected relatives. The mutation did not appear among 288 Palestinian controls (576 chromosomes), all adults with normal hearing. No homozygous mutations in PTPRQ appeared in any of 218 other probands with hearing loss. CONCLUSION: Identification of the DFNB84 gene represents the first identification of PTPRQ mutation in human hearing loss.


Assuntos
Cílios/genética , Códon sem Sentido/genética , Loci Gênicos/genética , Perda Auditiva/enzimologia , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Análise Mutacional de DNA , Família , Feminino , Humanos , Padrões de Herança/genética , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem
11.
Hear Res ; 257(1-2): 53-62, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19666099

RESUMO

Glycogen synthase kinase-3 (GSK-3) plays an important role in the regulation of apoptosis. However, the role of GSK-3 in the auditory system remains unknown. Here we examined whether the GSK-3-specific inhibitors, SB 216763 and LiCl, could protect against cisplatin-induced cytotoxicity of auditory cells. GSK-3 was activated by cisplatin treatment of HEI-OC1 cells. SB 216763 or LiCl treatments inhibited cisplatin-induced apoptosis in a dose-dependent manner and activated caspase-9, -8 and -3. In rat primary explants of the organ of Corti, SB 216763 or LiCl treatments completely abrogated the cisplatin-induced destruction of outer hair cell arrays. Administration of SB 216763 or LiCl inhibited cochlear destruction and the production of tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and IL-6 in cisplatin-injected mice. Furthermore, administration of SB 216763 or LiCl reduced the thresholds of the auditory brainstem response (ABR) in cisplatin-injected mice. Collectively, these results suggest that cisplatin-induced ototoxicity might be associated with modulation of GSK-3 activation.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Perda Auditiva/prevenção & controle , Indóis/farmacologia , Cloreto de Lítio/farmacologia , Maleimidas/farmacologia , Órgão Espiral/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Estimulação Acústica , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular , Cisplatino/administração & dosagem , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/enzimologia , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Perda Auditiva/fisiopatologia , Indóis/administração & dosagem , Injeções Intraperitoneais , Interleucina-1beta/sangue , Interleucina-6/sangue , Cloreto de Lítio/administração & dosagem , Maleimidas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Órgão Espiral/enzimologia , Órgão Espiral/patologia , Órgão Espiral/fisiopatologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue
12.
Hum Mutat ; 29(1): 130-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17918732

RESUMO

Building on our discovery that mutations in the transmembrane serine protease, TMPRSS3, cause nonsyndromic deafness, we have investigated the contribution of other TMPRSS family members to the auditory function. To identify which of the 16 known TMPRSS genes had a strong likelihood of involvement in hearing function, three types of biological evidence were examined: 1) expression in inner ear tissues; 2) location in a genomic interval that contains a yet unidentified gene for deafness; and 3) evaluation of hearing status of any available Tmprss knockout mouse strains. This analysis demonstrated that, besides TMPRSS3, another TMPRSS gene was essential for hearing and, indeed, mice deficient for Hepsin (Hpn) also known as Tmprss1 exhibited profound hearing loss. In addition, TMPRSS2, TMPRSS5, and CORIN, also named TMPRSS10, showed strong likelihood of involvement based on their inner ear expression and mapping position within deafness loci PKSR7, DFNB24, and DFNB25, respectively. These four TMPRSS genes were then screened for mutations in affected members of the DFNB24 and DFNB25 deafness families, and in a cohort of 362 sporadic deaf cases. This large mutation screen revealed numerous novel sequence variations including three potential pathogenic mutations in the TMPRSS5 gene. The mutant forms of TMPRSS5 showed reduced or absent proteolytic activity. Subsequently, TMPRSS genes with evidence of involvement in deafness were further characterized, and their sites of expression were determined. Tmprss1, 3, and 5 proteins were detected in spiral ganglion neurons. Tmprss3 was also present in the organ of Corti. TMPRSS1 and 3 proteins appeared stably anchored to the endoplasmic reticulum membranes, whereas TMPRSS5 was also detected at the plasma membrane. Collectively, these results provide evidence that TMPRSS1 and TMPRSS3 play and TMPRSS5 may play important and specific roles in hearing.


Assuntos
Perda Auditiva/genética , Proteínas de Membrana/genética , Serina Endopeptidases/genética , Animais , Orelha Interna/metabolismo , Perda Auditiva/enzimologia , Humanos , Imuno-Histoquímica , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/análise , Serina Endopeptidases/metabolismo
13.
Hear Res ; 234(1-2): 10-4, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17933476

RESUMO

Fabry disease (OMIM 301500) is a rare X-linked recessive disorder caused by mutations in the alpha-galactosidase gene (Gla). Loss of Gla activity leads to the abnormal accumulation of glycosphingolipids in lysosomes of predominantly vascular endothelial cells. Clinically the disorder presents with angiokeratomas, clouding of the cornea, and renal, cardiac, and cerebrovascular complications. In addition, there is an increased incidence of sensorineural hearing loss in Fabry patients. In this study, we investigated the loss of alpha-galactosidase A activity on hearing function in Gla-deficient mice (Gla(tm1Kul)). Gla mRNA was readily detected in the cochlea of 2- and 12-month old C57BL/6J and C3HeB/FeJ mice. The targeted allele was introgressed to the normal hearing C3HeB/FeJ strain to eliminate confounding genetic background effects. Auditory brain stem responses (ABR) to click, 8-, 16-, and 32 kHz stimuli measured at regular intervals from animals at the N4 backcross generation and from N4F1 hybrids demonstrated normal hearing in hemizygous and homozygous mutant mice up to 76 weeks of age. By histological criteria, the cyto-architecture of the mutant cochlea showed a normal appearance. The data demonstrate that in the mouse the loss of alpha-galactosidase A activity is genetically or biochemically buffered and not sufficient per se to cause an appreciable degree of hearing impairment.


Assuntos
Cóclea/fisiopatologia , Doença de Fabry/fisiopatologia , Audição , alfa-Galactosidase/metabolismo , Estimulação Acústica , Animais , Cóclea/enzimologia , Cóclea/patologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Doença de Fabry/complicações , Doença de Fabry/enzimologia , Doença de Fabry/genética , Feminino , Perda Auditiva/enzimologia , Perda Auditiva/etiologia , Perda Auditiva/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Fatores de Tempo , alfa-Galactosidase/genética
14.
Mol Endocrinol ; 21(7): 1593-602, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17440044

RESUMO

Dual oxidases generate the hydrogen peroxide needed by thyroid peroxidase for the incorporation of iodine into thyroglobulin, an essential step in thyroid hormone synthesis. Mutations in the human dual oxidase 2 gene, DUOX2, have been shown to underlie several cases of congenital hypothyroidism. We report here the first mouse Duox2 mutation, which provides a new genetic model for studying the specific function of DUOX2 in the thyroid gland and in other organ systems where it is hypothesized to play a role. We mapped the new spontaneous mouse mutation to chromosome 2 and identified it as a T>G base pair change in exon 16 of Duox2. The mutation changes a highly conserved valine to glycine at amino acid position 674 (V674G) and was named "thyroid dyshormonogenesis" (symbol thyd) to signify a defect in thyroid hormone synthesis. Thyroid glands of mutant mice are goitrous and contain few normal follicles, and anterior pituitaries are dysplastic. Serum T(4) in homozygotes is about one-tenth the level of controls and is accompanied by a more than 100-fold increase in TSH. The weight of adult mutant mice is approximately half that of littermate controls, and serum IGF-I is reduced. The cochleae of mutant mice exhibit abnormalities characteristic of hypothyroidism, including a delayed formation of the inner sulcus and tunnel of Corti and an abnormally thickened tectorial membrane. Hearing thresholds of adult mutant mice are on average 50-60 decibels (dB) above those of controls.


Assuntos
Hipotireoidismo Congênito/enzimologia , Hipotireoidismo Congênito/genética , Nanismo/enzimologia , Nanismo/genética , Flavoproteínas/genética , Perda Auditiva/enzimologia , Perda Auditiva/genética , Mutação de Sentido Incorreto , NADPH Oxidases/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Peso Corporal , Cóclea/patologia , Hipotireoidismo Congênito/sangue , Hipotireoidismo Congênito/patologia , Primers do DNA/genética , Modelos Animais de Doenças , Oxidases Duais , Feminino , Flavoproteínas/química , Flavoproteínas/fisiologia , Perda Auditiva/patologia , Perda Auditiva/fisiopatologia , Homozigoto , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , NADPH Oxidases/química , NADPH Oxidases/fisiologia , Fenótipo , Gravidez , Homologia de Sequência de Aminoácidos , Tireotropina/sangue , Tiroxina/sangue
15.
Biochim Biophys Acta ; 1773(8): 1341-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17306896

RESUMO

c-Jun N-terminal kinases (JNKs), also referred to as stress-activated kinases (SAPKs), were initially characterized by their activation in response to cell stress such as UV irradiation. JNK/SAPKs have since been characterized to be involved in proliferation, apoptosis, motility, metabolism and DNA repair. Dysregulated JNK signaling is now believed to contribute to many diseases involving neurodegeneration, chronic inflammation, birth defects, cancer and ischemia/reperfusion injury. In this review, we present our current understanding of JNK regulation and their involvement in homeostasis and dysregulation in human disease.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sequência de Aminoácidos , Animais , Doença/etiologia , Feminino , Perda Auditiva/enzimologia , Humanos , Inflamação/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/química , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fígado/irrigação sanguínea , Fígado/lesões , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Dados de Sequência Molecular , Neoplasias/enzimologia , Defeitos do Tubo Neural/enzimologia , Doenças Neurodegenerativas/enzimologia , Gravidez , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais
16.
Hear Res ; 226(1-2): 168-77, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17098385

RESUMO

Neomycin ototoxicity and electrode insertion trauma both involve activation of the mitogen activated protein kinase (MAPK)/c-Jun-N-terminal kinase (JNK) cell death signal cascade. This article discusses mechanisms of cell death on a cell biology level (e.g. necrosis and apoptosis) and proposes the blocking of JNK signaling as a therapeutic approach for preventing the development of a permanent hearing loss that can be initiated by either neomycin ototoxicity or electrode insertion trauma. Blocking of JNK molecules incorporates the use of a peptide inhibitor (i.e. D-JNKI-1), which is specific for all three isoforms of JNK and has been demonstrated to prevent loss of hearing following either electrode insertion trauma or loss of both hearing and hair cells following exposure to an ototoxic level of neomycin. We present previously unpublished results that control for the effect of perfusate washout of aminoglycoside antibiotic by perfusion of the scala tympani with an inactive form of D-JNKI-1 peptide, i.e. JNKI-1(mut) peptide, which was not presented in the original J. Neurosci. article that tested locally delivered D-JNKI-1 peptide against both noise- and neomycin-induced hearing loss (i.e. Wang, J., Van De Water, T.R., Bonny, C., de Ribaupierre, F., Puel, J.L., Zine, A. 2003a. A peptide inhibitor of c-Jun N-terminal kinase protects against both aminoglycoside and acoustic trauma-induced auditory hair cell death and hearing loss. J. Neurosci. 23, 8596-8607). D-JNKI-1 is a cell permeable peptide that blocks JNK signaling at the level of the three JNK molecular isoforms, which when blocked prevents the increases in hearing thresholds and the loss of auditory hair cells. This unique therapeutic approach may have clinical application for preventing: (1) hearing loss caused by neomycin ototoxicity; and (2) the progressive component of electrode insertion trauma-induced hearing loss.


Assuntos
Perda Auditiva/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Animais , Antibacterianos/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Eletrodos/efeitos adversos , Radicais Livres/metabolismo , Cobaias , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Perda Auditiva/etiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neomicina/toxicidade , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
Cancer Res ; 64(24): 9217-24, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15604295

RESUMO

Cisplatin (CDDP) is a highly effective chemotherapeutic agent but with significant ototoxic side effects. Apoptosis is an important mechanism of cochlear hair cell loss following exposure to an ototoxic level of CDDP. This study examines intracellular pathways involved in hair cell death induced by CDDP exposure in vivo to develop effective therapeutic strategies to protect the auditory receptor from CDDP-initiated hearing loss. Guinea pigs were treated with systemic administration of CDDP. Cochlear hair cells from CDDP-treated animals exhibited classic apoptotic alterations in their morphology. Several important signaling events that regulate the death of CDDP-injured cochlear hair cells were identified. CDDP treatment induced the activation and redistribution of cytosolic Bax and the release of cytochrome c from injured mitochondria. Activation of caspase-9 and caspase-3, but not caspase-8, was detected after treatment with CDDP, and the cleavage of fodrin by activated caspase-3 was observed within damaged hair cells. Intracochlear perfusions with caspase-3 inhibitor (z-DEVD-fmk) and caspase-9 inhibitor (z-LEHD-fmk) prevent hearing loss and loss of sensory cells, but caspase-8 inhibitor (z-IETD-fmk) and cathepsin B inhibitor (z-FA-fmk) do not. Although the stress-activated protein kinase/c-Jun NH(2)-terminal kinase (JNK) signaling pathway is activated in response to CDDP toxicity, intracochlear perfusion of d-JNKI-1, a JNK inhibitor, did not protect against CDDP ototoxicity but instead potentiated the ototoxic effects of CDDP. The results of the present study show that blocking a critical step in apoptosis may be a useful strategy to prevent harmful side effects of CDDP ototoxicity in patients having to undergo chemotherapy.


Assuntos
Inibidores de Caspase , Cisplatino/toxicidade , Inibidores de Cisteína Proteinase/farmacologia , Perda Auditiva/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Animais , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Citocromos c/metabolismo , Feminino , Cobaias , Células Ciliadas Auditivas/efeitos dos fármacos , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Isoenzimas , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas dos Microfilamentos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2
18.
Nat Cell Biol ; 5(5): 422-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12717441

RESUMO

Maintenance of the post-mitotic state in the post-natal mammalian brain is an active process that requires the cyclin-dependent kinase inhibitors (CKIs) p19Ink4d (Ink4d) and p27Kip1 (Kip1). In animals with targeted deletions of both Ink4d and Kip1, terminally differentiated, post-mitotic neurons are observed to re-enter the cell cycle, divide and undergo apoptosis. However, when either Ink4d or Kip1 alone are deleted, the post-mitotic state is maintained, suggesting a redundant role for these genes in mature neurons. In the organ of Corti--the auditory sensory epithelium of mammals--sensory hair cells and supporting cells become post-mitotic during embryogenesis and remain quiescent for the life of the animal. When lost as a result of environmental insult or genetic abnormality, hair cells do not regenerate, and this loss is a common cause of deafness in humans. Here, we report that targeted deletion of Ink4d alone is sufficient to disrupt the maintenance of the post-mitotic state of sensory hair cells in post-natal mice. In Ink4d-/- animals, hair cells are observed to aberrantly re-enter the cell cycle and subsequently undergo apoptosis, resulting in progressive hearing loss. Our results identify a novel mechanism underlying a non-syndromic form of progressive hearing loss in mice.


Assuntos
Apoptose/genética , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Células Ciliadas Auditivas/enzimologia , Perda Auditiva/enzimologia , Perda Auditiva/genética , Regeneração Nervosa/genética , Proteínas Supressoras de Tumor/deficiência , Animais , Caspase 3 , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Divisão Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19 , Inibidor de Quinase Dependente de Ciclina p27 , Dineínas , Feto , Imunofluorescência , Células Ciliadas Auditivas/ultraestrutura , Homeostase/genética , Camundongos , Camundongos Knockout , Miosina VIIa , Miosinas/metabolismo , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA