Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Audiol ; 60(4): 239-245, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32985284

RESUMO

OBJECTIVE: Rehabilitation options for conductive and mixed hearing loss are continually expanding, but without standard outcome measures comparison between different treatments is difficult. To meaningfully inform clinicians and patients core outcome sets (COS), determined via a recognised methodology, are needed. Following our previous work that identified hearing, physical, economic and psychosocial as core areas of a future COS, the AURONET group reviewed hearing outcome measures used in existing literature and assigned them into different domains within the hearing core area. DESIGN: Scoping review. STUDY SAMPLE: Literature including hearing outcome measurements for the treatment of conductive and/or mixed hearing loss. RESULTS: The literature search identified 1434 studies, with 278 subsequently selected for inclusion. A total of 837 hearing outcome measures were reported and grouped into nine domains. The largest domain constituted pure-tone threshold measurements accounting for 65% of the total outcome measures extracted, followed by the domains of speech testing (20%) and questionnaires (9%). Studies of hearing implants more commonly included speech tests or hearing questionnaires compared with studies of middle ear surgery. CONCLUSIONS: A wide range of outcome measures are currently used, highlighting the importance of developing a COS to inform individual practice and reporting in trials/research.


Assuntos
Surdez , Perda Auditiva Condutiva-Neurossensorial Mista , Perda Auditiva , Adulto , Audição , Perda Auditiva/diagnóstico , Perda Auditiva Condutiva/diagnóstico , Perda Auditiva Condutiva/terapia , Humanos , Avaliação de Resultados em Cuidados de Saúde , Resultado do Tratamento
2.
Cancer Res ; 77(8): 1997-2007, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28381547

RESUMO

In pancreatic ductal adenocarcinoma (PDAC), mutant KRAS stimulates the translation initiation factor eIF5A and upregulates the focal adhesion kinase PEAK1, which transmits integrin and growth factor signals mediated by the tumor microenvironment. Although eIF5A-PEAK1 signaling contributes to multiple aggressive cancer cell phenotypes, the downstream signaling processes that mediate these responses are uncharacterized. Through proteomics and informatic analyses of PEAK1-depleted PDAC cells, we defined protein translation, cytoskeleton organization, and cell-cycle regulatory pathways as major pathways controlled by PEAK1. Biochemical and functional studies revealed that the transcription factors YAP1 and TAZ are key targets of eIF5A-PEAK1 signaling. YAP1/TAZ coimmunoprecipitated with PEAK1. Interfering with eIF5A-PEAK1 signaling in PDAC cells inhibited YAP/TAZ protein expression, decreasing expression of stem cell-associated transcription factors (STF) including Oct4, Nanog, c-Myc, and TEAD, thereby decreasing three-dimensional (3D) tumor sphere growth. Conversely, amplified eIF5A-PEAK1 signaling increased YAP1/TAZ expression, increasing expression of STF and enhancing 3D tumor sphere growth. Informatic interrogation of mRNA sequence databases revealed upregulation of the eIF5A-PEAK1-YAP1-TEAD signaling module in PDAC patients. Taken together, our findings indicate that eIF5A-PEAK1-YAP signaling contributes to PDAC development by regulating an STF program associated with increased tumorigenicity. Cancer Res; 77(8); 1997-2007. ©2017 AACR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fatores de Iniciação de Peptídeos/biossíntese , Fatores de Iniciação de Peptídeos/genética , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP , Fator de Iniciação de Tradução Eucariótico 5A
3.
J Biol Chem ; 290(50): 29907-19, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26483550

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with an overall survival rate of less than 5%. The poor patient outcome in PDAC is largely due to the high prevalence of systemic metastasis at the time of diagnosis and lack of effective therapeutics that target disseminated cells. The fact that the underlying mechanisms driving PDAC cell migration and dissemination are poorly understood have hindered drug development and compounded the lack of clinical success in this disease. Recent evidence indicates that mutational activation of K-Ras up-regulates eIF5A, a component of the cellular translational machinery that is critical for PDAC progression. However, the role of eIF5A in PDAC cell migration and metastasis has not been investigated. We report here that pharmacological inhibition or genetic knockdown of eIF5A reduces PDAC cell migration, invasion, and metastasis in vitro and in vivo. Proteomic profiling and bioinformatic analyses revealed that eIF5A controls an integrated network of cytoskeleton-regulatory proteins involved in cell migration. Functional interrogation of this network uncovered a critical RhoA/ROCK signaling node that operates downstream of eIF5A in invasive PDAC cells. Importantly, eIF5A mediates PDAC cell migration and invasion by modulating RhoA/ROCK protein expression levels. Together our findings implicate eIF5A as a cytoskeletal rheostat controlling RhoA/ROCK protein expression during PDAC cell migration and metastasis. Our findings also implicate the eIF5A/RhoA/ROCK module as a potential new therapeutic target to treat metastatic PDAC cells.


Assuntos
Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Fatores de Iniciação de Peptídeos/fisiologia , Proteínas de Ligação a RNA/fisiologia , Quinases Associadas a rho/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Fatores de Iniciação de Peptídeos/genética , Proteínas de Ligação a RNA/genética , Fator de Iniciação de Tradução Eucariótico 5A
4.
Australas Phys Eng Sci Med ; 38(2): 325-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25749989

RESUMO

The Australian Radiation Protection and Nuclear Safety Agency (ARPANSA) maintains a (60)Co teletherapy source primarily for the calibration of therapy dosemeters. The source and encapsulating head were replaced in early 2010 with an Eldorado 78 head and new (60)Co source. In this article we present the results of ongoing accuracy and stability measurements since the replacement. A number of formal and informal indirect comparisons have been carried out with laboratories holding primary and secondary standards for (60)Co. ARPANSA chambers have also been calibrated at international primary standard laboratories allowing comparison of calibration coefficients and thus (60)Co absorbed dose standards. (60)Co calibration coefficients supplied by manufacturers of chambers were compared to those measured at the ARPANSA when this calibration was traceable to a primary standard. ARPANSA also participates in an annual international mailed dosimetry audit conducted by the International Atomic Energy Agency. The results thus far demonstrate that the absorbed doses to water delivered by the new ARPANSA (60)Co source are consistent with international doses within the stated uncertainties.


Assuntos
Radioisótopos de Cobalto/análise , Radiometria/métodos , Radiometria/normas , Austrália , Calibragem , Auditoria Clínica , Internacionalidade , Sociedades Científicas
5.
Cancer Res ; 74(22): 6671-81, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25261239

RESUMO

Deregulation of protein synthesis is a hallmark of cancer cell proliferation, survival, and metastatic progression. eIF5A1 and its highly related isoform eIF5A2 are translation initiation factors that have been implicated in a range of human malignancies, but how they control cancer development and disease progression is still poorly understood. Here, we investigated how eIF5A proteins regulate pancreatic ductal adenocarcinoma (PDAC) pathogenesis. eIF5A proteins are the only known proteins regulated by a distinct posttranslational modification termed hypusination, which is catalyzed by two enzymes, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). The highly selective nature of the hypusine modification and its amenability to pharmacologic inhibition make eIF5A proteins attractive therapeutic targets. We found that the expression and hypusination of eIF5A proteins are upregulated in human PDAC tissues and in premalignant pancreatic intraepithelial neoplasia tissues isolated from Pdx-1-Cre: LSL-KRAS(G12D) mice. Knockdown of eIF5A proteins in PDAC cells inhibited their growth in vitro and orthotopic tumor growth in vivo, whereas amplification of eIF5A proteins increased PDAC cell growth and tumor formation in mice. Small-molecule inhibitors of DHPS and DOHH both suppressed eIF5A hypusination, preventing PDAC cell growth. Interestingly, we found that eIF5A proteins regulate PDAC cell growth by modulating the expression of PEAK1, a nonreceptor tyrosine kinase essential for PDAC cell growth and therapy resistance. Our findings suggest that eIF5A proteins utilize PEAK1 as a downstream effector to drive PDAC pathogenesis and that pharmacologic inhibition of the eIF5A-hypusine-PEAK1 axis may provide a novel therapeutic strategy to combat this deadly disease.


Assuntos
Carcinoma Ductal Pancreático/etiologia , Lisina/análogos & derivados , Neoplasias Pancreáticas/etiologia , Fatores de Iniciação de Peptídeos/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas de Ligação a RNA/fisiologia , Animais , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Ciclopirox , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Feminino , Humanos , Lisina/fisiologia , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas p21(ras) , Piridonas/farmacologia , Proteínas ras/fisiologia , Gencitabina , Fator de Iniciação de Tradução Eucariótico 5A
6.
Stem Cell Reports ; 2(4): 427-39, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24749068

RESUMO

Little is known about the extracellular signaling factors that govern mammary stem cell behavior. Here, we identify CRIPTO and its cell-surface receptor GRP78 as regulators of stem cell behavior in isolated fetal and adult mammary epithelial cells. We develop a CRIPTO antagonist that promotes differentiation and reduces self-renewal of mammary stem cell-enriched populations cultured ex vivo. By contrast, CRIPTO treatment maintains the stem cell phenotype in these cultures and yields colonies with enhanced mammary gland reconstitution capacity. Surface expression of GRP78 marks CRIPTO-responsive, stem cell-enriched fetal and adult mammary epithelial cells, and deletion of GRP78 from adult mammary epithelial cells blocks their mammary gland reconstitution potential. Together, these findings identify the CRIPTO/GRP78 pathway as a developmentally conserved regulator of fetal and adult mammary stem cell behavior ex vivo, with implications for the stem-like cells found in many cancers.


Assuntos
Proteínas Ligadas por GPI/metabolismo , Proteínas de Choque Térmico/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glândulas Mamárias Humanas/citologia , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Biomarcadores , Diferenciação Celular , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/genética , Expressão Gênica , Proteínas de Choque Térmico/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Glândulas Mamárias Humanas/fisiologia , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Ligação Proteica , Regeneração , Células-Tronco/citologia
7.
Biomaterials ; 33(29): 7064-70, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22809641

RESUMO

Our current understanding of 3-dimensional (3D) cell migration is primarily based on results from fibrous scaffolds with randomly organized internal architecture. Manipulations that change the stiffness of these 3D scaffolds often alter other matrix parameters that can modulate cell motility independently or synergistically, making observations less predictive of how cells behave when migrating in 3D. In order to decouple microstructural influences and stiffness effects, we have designed and fabricated 3D polyethylene glycol (PEG) scaffolds that permit orthogonal tuning of both elastic moduli and microstructure. Scaffolds with log-pile architectures were used to compare the 3D migration properties of normal breast epithelial cells (HMLE) and Twist-transformed cells (HMLET). Our results indicate that the nature of cell migration is significantly impacted by the ability of cells to migrate in the third dimension. 2D ECM-coated PEG substrates revealed no statistically significant difference in cell migration between HMLE and HMLET cells among substrates of different stiffness. However, when cells were allowed to move along the third dimension, substantial differences were observed for cell displacement, velocity and path straightness parameters. Furthermore, these differences were sensitive to both substrate stiffness and the presence of the Twist oncogene. Importantly, these 3D modes of migration provide insight into the potential for oncogene-transformed cells to migrate within and colonize tissues of varying stiffness.


Assuntos
Neoplasias/metabolismo , Polietilenoglicóis/química , Alicerces Teciduais/química , Biofísica/métodos , Mama/citologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Reagentes de Ligações Cruzadas/farmacologia , Elasticidade , Células Epiteliais/citologia , Desenho de Equipamento , Feminino , Humanos , Microscopia Eletrônica de Varredura/métodos , Metástase Neoplásica , Polímeros/química , Estresse Mecânico
8.
Cancer Res ; 72(10): 2554-64, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22589274

RESUMO

Early biomarkers and effective therapeutic strategies are desperately needed to treat pancreatic ductal adenocarcinoma (PDAC), which has a dismal 5-year patient survival rate. Here, we report that the novel tyrosine kinase PEAK1 is upregulated in human malignancies, including human PDACs and pancreatic intraepithelial neoplasia (PanIN). Oncogenic KRas induced a PEAK1-dependent kinase amplification loop between Src, PEAK1, and ErbB2 to drive PDAC tumor growth and metastasis in vivo. Surprisingly, blockade of ErbB2 expression increased Src-dependent PEAK1 expression, PEAK1-dependent Src activation, and tumor growth in vivo, suggesting a mechanism for the observed resistance of patients with PDACs to therapeutic intervention. Importantly, PEAK1 inactivation sensitized PDAC cells to trastuzumab and gemcitabine therapy. Our findings, therefore, suggest that PEAK1 is a novel biomarker, critical signaling hub, and new therapeutic target in PDACs.


Assuntos
Carcinoma Ductal Pancreático/genética , Genes erbB-2 , Proteína Oncogênica pp60(v-src)/genética , Neoplasias Pancreáticas/genética , Proteínas Tirosina Quinases/genética , Animais , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Genes ras , Humanos , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Metástase Neoplásica/genética , Transplante de Neoplasias , Transdução de Sinais/genética , Ativação Transcricional , Regulação para Cima
9.
Dev Biol ; 356(2): 383-97, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21664901

RESUMO

Heart development requires contributions from, and coordinated signaling interactions between, several cell populations, including splanchnic and pharyngeal mesoderm, postotic neural crest and the proepicardium. Here we report that Fgf3 and Fgf10, which are expressed dynamically in and near these cardiovascular progenitors, have redundant and dosage sensitive requirements in multiple aspects of early murine cardiovascular development. Embryos with Fgf3(-/+);Fgf10(-/-), Fgf3(-/-);Fgf10(-/+) and Fgf3(-/-);Fgf10(-/-) genotypes formed an allelic series of increasing severity with respect to embryonic survival, with double mutants dead by E11.5. Morphologic analysis of embryos with three mutant alleles at E11.5-E13.5 and double mutants at E9.5-E11.0 revealed multiple cardiovascular defects affecting the outflow tract, ventricular septum, atrioventricular cushions, ventricular myocardium, dorsal mesenchymal protrusion, pulmonary arteries, epicardium and fourth pharyngeal arch artery. Assessment of molecular markers in E8.0-E10.5 double mutants revealed abnormalities in each progenitor population, and suggests that Fgf3 and Fgf10 are not required for specification of cardiovascular progenitors, but rather for their normal developmental coordination. These results imply that coding or regulatory mutations in FGF3 or FGF10 could contribute to human congenital heart defects.


Assuntos
Vasos Coronários/fisiologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Fator 3 de Crescimento de Fibroblastos/fisiologia , Coração/embriologia , Neovascularização Fisiológica , Animais , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Camundongos , Crista Neural/anormalidades , Gravidez , Proteínas com Domínio T/genética
10.
J Mol Cell Cardiol ; 51(1): 24-32, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21402077

RESUMO

AMP-activated protein kinase (AMPK) is a stress signaling enzyme that orchestrates the regulation of energy-generating and -consuming pathways. Intrinsic AMPK activation protects the heart against ischemic injury and apoptosis, but whether pharmacologic AMPK stimulation mitigates ischemia-reperfusion damage is unknown. The aims of this study were to determine whether direct stimulation of AMPK using a small molecule activator, A-769662, attenuates myocardial ischemia-reperfusion injury and to examine its cardioprotective mechanisms. Isolated mouse hearts pre-treated with A-769662 had better recovery of left ventricular contractile function (55% vs. 29% of baseline rate-pressure product; p=0.03) and less myocardial necrosis (56% reduction in infarct size; p<0.01) during post-ischemic reperfusion compared to control hearts. Pre-treatment with A-769662 in vivo attenuated infarct size in C57Bl/6 mice undergoing left coronary artery occlusion and reperfusion compared to vehicle (36% vs. 18%, p=0.025). Mouse hearts with genetically inactivated AMPK were not protected by A-769662, indicating the specificity of this compound. Pre-treatment with A-769662 increased the phosphorylation and inactivation of eukaryotic elongation factor 2 (eEF2), preserved energy charge during ischemia, delayed the development of ischemic contracture, and reduced myocardial apoptosis and necrosis. A-769662 also augmented endothelial nitric oxide synthase (eNOS) activation during ischemia, which partially attenuated myocardial stunning, but did not prevent necrosis. AMPK is a therapeutic target that can be stimulated by a direct-acting small molecule in order to prevent injury during ischemia-reperfusion. The use of AMPK activators may represent a novel strategy to protect the heart and other solid organs against ischemia.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiotônicos/farmacologia , Ativadores de Enzimas/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pironas/farmacologia , Tiofenos/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo , Coração/fisiopatologia , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Necrose , Óxido Nítrico Sintase Tipo III/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo
11.
Development ; 130(15): 3379-90, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12810586

RESUMO

The inner ear, which contains the sensory organs specialised for audition and balance, develops from an ectodermal placode adjacent to the developing hindbrain. Tissue grafting and recombination experiments suggest that placodal development is directed by signals arising from the underlying mesoderm and adjacent neurectoderm. In mice, Fgf3 is expressed in the neurectoderm prior to and concomitant with placode induction and otic vesicle formation, but its absence affects only the later stages of otic vesicle morphogenesis. We show here that mouse Fgf10 is expressed in the mesenchyme underlying the prospective otic placode. Embryos lacking both Fgf3 and Fgf10 fail to form otic vesicles and have aberrant patterns of otic marker gene expression, suggesting that FGF signals are required for otic placode induction and that these signals emanate from both the hindbrain and mesenchyme. These signals are likely to act directly on the ectoderm, as double mutant embryos showed normal patterns of gene expression in the hindbrain. Cell proliferation and survival were not markedly affected in double mutant embryos, suggesting that the major role of FGF signals in otic induction is to establish normal patterns of gene expression in the prospective placode. Finally, examination of embryos carrying three out of the four mutant Fgf alleles revealed intermediate phenotypes, suggesting a quantitative requirement for FGF signalling in otic vesicle formation.


Assuntos
Orelha/embriologia , Indução Embrionária/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Fator 10 de Crescimento de Fibroblastos , Fator 3 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/genética , Mesoderma/metabolismo , Camundongos , Mutação , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA