Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 5275, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29588456

RESUMO

Anesthetic agents have been implicated in the causation of neurological and cognitive deficits after surgery, the exacerbation of chronic neurodegenerative disease, and were recently reported to promote the onset of the neurologic respiratory disease Congenital Central Hypoventilation Syndrome (CCHS), related to misfolding of the transcription factor Phox2B. To study how anesthetic agents could affect neuronal function through alterations to protein folding, we created neuronal cell models emulating the graded disease severity of CCHS. We found that the gas anesthetic isoflurane and the opiate morphine potentiated aggregation and mislocalization of Phox2B variants, similar to that seen in CCHS, and observed transcript and protein level changes consistent with activation of the endoplasmic reticulum (ER) unfolded protein response. Attenuation of ER stress pathways did not result in a correction of Phox2B misfolding, indicating a primary effect of isoflurane on protein structure. We also observed that isoflurane hindered the folding and activity of proteins that rely heavily on ER function, like the CFTR channel. Our results show how anesthetic drugs can alter protein folding and induce ER stress, indicating a mechanism by which these agents may affect neuronal function after surgery.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Proteínas de Homeodomínio/metabolismo , Hipoventilação/congênito , Isoflurano/efeitos adversos , Morfina/efeitos adversos , Agregação Patológica de Proteínas/induzido quimicamente , Apneia do Sono Tipo Central/induzido quimicamente , Fatores de Transcrição/metabolismo , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/análise , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Homeodomínio/análise , Humanos , Hipoventilação/induzido quimicamente , Hipoventilação/metabolismo , Hipoventilação/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Dobramento de Proteína/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Apneia do Sono Tipo Central/metabolismo , Apneia do Sono Tipo Central/patologia , Fatores de Transcrição/análise , Resposta a Proteínas não Dobradas/efeitos dos fármacos
2.
Hum Mutat ; 39(2): 219-236, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29098737

RESUMO

Heterozygous mutations in the PHOX2B gene are causative of congenital central hypoventilation syndrome (CCHS), a neurocristopathy characterized by defective autonomic control of breathing due to the impaired differentiation of neural crest cells. Among PHOX2B mutations, polyalanine (polyAla) expansions are almost exclusively associated with isolated CCHS, whereas frameshift variants, although less frequent, are often more severe than polyAla expansions and identified in syndromic CCHS. This article provides a complete review of all the frameshift mutations identified in cases of isolated and syndromic CCHS reported in the literature as well as those identified by us and not yet published. These were considered in terms of both their structure, whether the underlying indels induced frameshifts of either 1 or 2 steps ("frame 2" and "frame 3" mutations respectively), and clinical associations. Furthermore, we evaluated the structural and functional effects of one "frame 3" mutation identified in a patient with isolated CCHS, and one "frame 2" mutation identified in a patient with syndromic CCHS, also affected with Hirschsprung's disease and neuroblastoma. The data thus obtained confirm that the type of translational frame affects the severity of the transcriptional dysfunction and the predisposition to isolated or syndromic CCHS.


Assuntos
Mutação da Fase de Leitura/genética , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Hipoventilação/congênito , Apneia do Sono Tipo Central/genética , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Humanos , Hipoventilação/genética , Hipoventilação/metabolismo , Microscopia de Fluorescência , Mutação , Apneia do Sono Tipo Central/metabolismo
3.
J Biol Chem ; 292(14): 5784-5800, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28246169

RESUMO

Polyalanine (poly(A)) diseases are caused by the expansion of translated GCN triplet nucleotide sequences encoding poly(A) tracts in proteins. To date, nine human disorders have been found to be associated with poly(A) tract expansions, including congenital central hypoventilation syndrome and oculopharyngeal muscular dystrophy. Previous studies have demonstrated that unexpanded wild-type poly(A)-containing proteins localize to the cell nucleus, whereas expanded poly(A)-containing proteins primarily localize to the cytoplasm. Because most of these poly(A) disease proteins are transcription factors, this mislocalization causes cellular transcriptional dysregulation leading to cellular dysfunction. Correcting this faulty localization could potentially point to strategies to treat the aforementioned disorders, so there is a pressing need to identify the mechanisms underlying the mislocalization of expanded poly(A) protein. Here, we performed a glutathione S-transferase pulldown assay followed by mass spectrometry and identified eukaryotic translation elongation factor 1 α1 (eEF1A1) as an interacting partner with expanded poly(A)-containing proteins. Strikingly, knockdown of eEF1A1 expression partially corrected the mislocalization of the expanded poly(A) proteins in the cytoplasm and restored their functions in the nucleus. We further demonstrated that the expanded poly(A) domain itself can serve as a nuclear export signal. Taken together, this study demonstrates that eEF1A1 regulates the subcellular location of expanded poly(A) proteins and is therefore a potential therapeutic target for combating the pathogenesis of poly(A) diseases.


Assuntos
Sinais de Exportação Nuclear , Fator 1 de Elongação de Peptídeos/metabolismo , Peptídeos/metabolismo , Expansão das Repetições de Trinucleotídeos , Células HEK293 , Humanos , Hipoventilação/congênito , Hipoventilação/genética , Hipoventilação/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Transporte Proteico/genética , Apneia do Sono Tipo Central/genética , Apneia do Sono Tipo Central/metabolismo
4.
J Mol Med (Berl) ; 90(9): 1025-35, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22307522

RESUMO

Expansions of a polyalanine (polyA) stretch in the coding region of the PHOX2B gene cause congenital central hypoventilation syndrome (CCHS), a neurocristopathy characterized by the absence of adequate control of autonomic breathing. Expansion of polyA in PHOX2B leads to protein misfolding and accumulation into inclusions. The mechanisms that regulate mutant protein degradation and turnover have been poorly elucidated. Here, we investigate the regulation of degradation of wild-type and polyA-expanded PHOX2B. We show that expanded PHOX2B is targeted for degradation through the ubiquitin-proteasome system, resulting in lowered levels of the mutant protein relative to its wild-type counterpart. Moreover, we show that mutant PHOX2B forms ubiquitin-positive inclusions, which sequester wild-type PHOX2B. This sequestration correlates with reduced transcriptional activity of endogenous wild-type protein in neuroblastoma cells. Finally, we show that the E3 ubiquitin ligase TRIM11 plays a critical role in the clearance of mutant PHOX2B through the proteasome. Importantly, clearance of mutant PHOX2B by TRIM11 correlates with a rescue of PHOX2B transcriptional activity. We propose that CCHS is partially caused by a dominant-negative effect of expanded PHOX2B due to the retention of the wild-type protein in pathogenic aggregates. Our results demonstrate that TRIM11 is a novel modifier of mutant PHOX2B toxicity and represents a potential therapeutic target for CCHS.


Assuntos
Proteínas de Homeodomínio/metabolismo , Hipoventilação/congênito , Peptídeos/metabolismo , Apneia do Sono Tipo Central/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Células HeLa , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/genética , Humanos , Hipoventilação/genética , Hipoventilação/metabolismo , Proteínas Mutantes/análise , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Peptídeos/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Apneia do Sono Tipo Central/genética , Fatores de Transcrição/análise , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas com Motivo Tripartido , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/análise
5.
Neurobiol Dis ; 45(1): 508-18, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21964250

RESUMO

Heterozygous in frame duplications of the PHOX2B gene, leading to polyalanine (polyAla) expansions ranging from +5 to +13 residues of a 20-alanine stretch, have been identified in the vast majority of patients affected with Congenital Central Hypoventilation Syndrome (CCHS), a rare neurocristopathy characterized by absence of adequate autonomic control of respiration with decreased sensitivity to hypoxia and hypercapnia. Ventilatory supports such as tracheostomy, nasal mask or diaphragm pacing represent the only options available for affected. We have already shown that the severity of the CCHS phenotype correlates with the length of polyAla expansions, ultimately leading to formation of toxic intracytoplasmic aggregates and impaired PHOX2B mediated transactivation of target gene promoters, such as DBH. At present, there is no specific treatment to reduce cell aggregates and to ameliorate patients' respiration. In this work, we have undertaken in vitro analyses aimed at assessing the effects of molecules on the cellular response to polyAla PHOX2B aggregates. In particular, we tested 17-AAG, ibuprofen, 4-PBA, curcumin, trehalose, congo red and chrysamine G for their ability to i) recover the nuclear localisation of polyAla expanded PHOX2B, ii) rescue of PHOX2B mediated transactivation of the DBH promoter, and iii) clearance of PHOX2B (+13 Ala) aggregates. Our data have suggested that 17-AAG and curcumin are effective in vitro in both rescuing the nuclear localization and transactivation activity of PHOX2B carrying the largest expansion of polyAla and promoting the clearance of aggregates of these mutant proteins inducing molecular mechanisms such as ubiquitin-proteasome (UPS), autophagy and heat shock protein (HSP) systems.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Hipoventilação/congênito , Peptídeos/genética , Apneia do Sono Tipo Central/genética , Fatores de Transcrição/genética , Animais , Benzoatos/farmacologia , Benzoquinonas/farmacologia , Compostos de Bifenilo/farmacologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Vermelho Congo/farmacologia , Curcumina/farmacologia , Células HeLa , Proteínas de Homeodomínio/metabolismo , Humanos , Hipoventilação/genética , Hipoventilação/metabolismo , Ibuprofeno/farmacologia , Lactamas Macrocíclicas/farmacologia , Peptídeos/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Apneia do Sono Tipo Central/metabolismo , Fatores de Transcrição/metabolismo , Trealose/farmacologia
6.
Nat Neurosci ; 6(10): 1091-100, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14513037

RESUMO

The genetic basis for the development of brainstem neurons that generate respiratory rhythm is unknown. Here we show that mice deficient for the transcription factor MafB die from central apnea at birth and are defective for respiratory rhythmogenesis in vitro. MafB is expressed in a subpopulation of neurons in the preBötzinger complex (preBötC), a putative principal site of rhythmogenesis. Brainstems from Mafb(-/-) mice are insensitive to preBötC electrolytic lesion or stimulation and modulation of rhythmogenesis by hypoxia or peptidergic input. Furthermore, in Mafb(-/-) mice the preBötC, but not major neuromodulatory groups, presents severe anatomical defects with loss of cellularity. Our results show an essential role of MafB in central respiratory control, possibly involving the specification of rhythmogenic preBötC neurons.


Assuntos
Proteínas Aviárias , Proteínas de Ligação a DNA/deficiência , Neurônios/metabolismo , Proteínas Oncogênicas , Respiração/genética , Centro Respiratório/fisiopatologia , Apneia do Sono Tipo Central/genética , Fatores de Transcrição/deficiência , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/embriologia , Vias Aferentes/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Estimulação Elétrica , Feto , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição MafB , Camundongos , Camundongos Knockout , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/embriologia , Rede Nervosa/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Técnicas de Cultura de Órgãos , Periodicidade , Receptores da Neurocinina-1/agonistas , Receptores da Neurocinina-1/metabolismo , Respiração/efeitos dos fármacos , Centro Respiratório/anormalidades , Centro Respiratório/patologia , Apneia do Sono Tipo Central/metabolismo , Apneia do Sono Tipo Central/fisiopatologia , Substância P/metabolismo , Substância P/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA