Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 21584, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34732748

RESUMO

The unfolded protein response (UPR) is a direct consequence of cellular endoplasmic reticulum (ER) stress and a key disease driving mechanism in IPF. The resolution of the UPR is directed by PPP1R15A (GADD34) and leads to the restoration of normal ribosomal activity. While the role of PPP1R15A has been explored in lung epithelial cells, the role of this UPR resolving factor has yet to be explored in lung mesenchymal cells. The objective of the current study was to determine the expression and role of PPP1R15A in IPF fibroblasts and in a bleomycin-induced lung fibrosis model. A survey of IPF lung tissue revealed that PPP1R15A expression was markedly reduced. Targeting PPP1R15A in primary fibroblasts modulated TGF-ß-induced fibroblast to myofibroblast differentiation and exacerbated pulmonary fibrosis in bleomycin-challenged mice. Interestingly, the loss of PPP1R15A appeared to promote lung fibroblast senescence. Taken together, our findings demonstrate the major role of PPP1R15A in the regulation of lung mesenchymal cells, and regulation of PPP1R15A may represent a novel therapeutic strategy in IPF.


Assuntos
Senescência Celular , Fibrose/metabolismo , Proteína Fosfatase 1/genética , Resposta a Proteínas não Dobradas , Idoso , Animais , Bleomicina , Diferenciação Celular , Proliferação de Células , Estresse do Retículo Endoplasmático , Feminino , Fibroblastos/metabolismo , Genótipo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Indóis/farmacologia , Pulmão/metabolismo , Masculino , Mesoderma/citologia , Camundongos , Pessoa de Meia-Idade , Morfolinas/farmacologia , Proteína Fosfatase 1/fisiologia , Análise de Sequência de RNA , Fator de Crescimento Transformador beta/metabolismo
2.
EMBO J ; 40(14): e100715, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34152608

RESUMO

Clearance of mitochondria following damage is critical for neuronal homeostasis. Here, we investigate the role of Miro proteins in mitochondrial turnover by the PINK1/Parkin mitochondrial quality control system in vitro and in vivo. We find that upon mitochondrial damage, Miro is promiscuously ubiquitinated on multiple lysine residues. Genetic deletion of Miro or block of Miro1 ubiquitination and subsequent degradation lead to delayed translocation of the E3 ubiquitin ligase Parkin onto damaged mitochondria and reduced mitochondrial clearance in both fibroblasts and cultured neurons. Disrupted mitophagy in vivo, upon post-natal knockout of Miro1 in hippocampus and cortex, leads to a dramatic increase in mitofusin levels, the appearance of enlarged and hyperfused mitochondria and hyperactivation of the integrated stress response (ISR). Altogether, our results provide new insights into the central role of Miro1 in the regulation of mitochondrial homeostasis and further implicate Miro1 dysfunction in the pathogenesis of human neurodegenerative disease.


Assuntos
Mitocôndrias/metabolismo , Mitofagia/fisiologia , Neurônios/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Doenças Neurodegenerativas/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia
3.
EMBO Mol Med ; 9(10): 1366-1378, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28855301

RESUMO

We have characterised the proteolytic cleavage events responsible for the shedding of triggering receptor expressed on myeloid cells 2 (TREM2) from primary cultures of human macrophages, murine microglia and TREM2-expressing human embryonic kidney (HEK293) cells. In all cell types, a soluble 17 kDa N-terminal cleavage fragment was shed into the conditioned media in a constitutive process that is inhibited by G1254023X and metalloprotease inhibitors and siRNA targeting ADAM10. Inhibitors of serine proteases and matrix metalloproteinases 2/9, and ADAM17 siRNA did not block TREM2 shedding. Peptidomimetic protease inhibitors highlighted a possible cleavage site, and mass spectrometry confirmed that shedding occurred predominantly at the H157-S158 peptide bond for both wild-type and H157Y human TREM2 and for the wild-type murine orthologue. Crucially, we also show that the Alzheimer's disease-associated H157Y TREM2 variant was shed more rapidly than wild type from HEK293 cells, possibly by a novel, batimastat- and ADAM10-siRNA-independent, sheddase activity. These insights offer new therapeutic targets for modulating the innate immune response in Alzheimer's and other neurological diseases.


Assuntos
Doença de Alzheimer/genética , Glicoproteínas de Membrana/metabolismo , Proteólise , Receptores Imunológicos/metabolismo , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Meios de Cultivo Condicionados , Células HEK293 , Humanos , Cetocolesteróis/farmacologia , Macrófagos/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos/genética
4.
Nat Commun ; 7: 13821, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929117

RESUMO

Aberrant protein aggregation is controlled by various chaperones, including CCT (chaperonin containing TCP-1)/TCP-1/TRiC. Mutated CCT4/5 subunits cause sensory neuropathy and CCT5 expression is decreased in Alzheimer's disease. Here, we show that CCT integrity is essential for autophagosome degradation in cells or Drosophila and this phenomenon is orchestrated by the actin cytoskeleton. When autophagic flux is reduced by compromise of individual CCT subunits, various disease-relevant autophagy substrates accumulate and aggregate. The aggregation of proteins like mutant huntingtin, ATXN3 or p62 after CCT2/5/7 depletion is predominantly autophagy dependent, and does not further increase with CCT knockdown in autophagy-defective cells/organisms, implying surprisingly that the effect of loss-of-CCT activity on mutant ATXN3 or huntingtin oligomerization/aggregation is primarily a consequence of autophagy inhibition rather than loss of physiological anti-aggregation activity for these proteins. Thus, our findings reveal an essential partnership between two key components of the proteostasis network and implicate autophagy defects in diseases with compromised CCT complex activity.


Assuntos
Autofagossomos/metabolismo , Autofagia , Chaperonina com TCP-1/metabolismo , Proteína Huntingtina/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Ataxina-3/metabolismo , Drosophila , Feminino , Células HeLa , Humanos , Lisossomos/metabolismo , Masculino , Camundongos Transgênicos , Proteínas de Ligação a RNA/metabolismo
5.
Dis Model Mech ; 8(7): 657-67, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26035384

RESUMO

Metals, including iron, are present at high concentrations in amyloid plaques in individuals with Alzheimer's disease, where they are also thought to be cofactors in generating oxidative stress and modulating amyloid formation. In this study, we present data from several Drosophila models of neurodegenerative proteinopathies indicating that the interaction between iron and amyloid beta peptide (Aß) is specific and is not seen for other aggregation-prone polypeptides. The interaction with iron is likely to be important in the dimerisation of Aß and is mediated by three N-terminal histidines. Transgenic fly lines systematically expressing all combinations of His>Ala substitutions in Aß were generated and used to study the pathological role of these residues. Developmental eye phenotypes, longevity and histological examinations indicate that the N-terminal histidines have distinct position-dependent and -independent mechanisms. The former mediate the toxic effects of metals and Aß aggregation under non-oxidising conditions and the latter are relevant under oxidising conditions. Understanding how Aß mediates neurotoxic effects in vivo will help to better target pathological pathways using aggregation blockers and metal-modifying agents.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Drosophila/metabolismo , Ferro/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Substituição de Aminoácidos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila/genética , Feminino , Ferritinas/metabolismo , Histidina/química , Humanos , Técnicas In Vitro , Oxirredução , Fenótipo , Agregados Proteicos , Agregação Patológica de Proteínas/etiologia , Agregação Patológica de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
Hum Mol Genet ; 24(14): 3929-38, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25954034

RESUMO

Brain-derived neurotrophic factor (BDNF) has a crucial role in learning and memory by promoting neuronal survival and modulating synaptic connectivity. BDNF levels are lower in the brains of individuals with Alzheimer's disease (AD), suggesting a pathogenic involvement. The Drosophila orthologue of BDNF is the highly conserved Neurotrophin 1 (DNT1). BDNF and DNT1 have the same overall protein structure and can be cleaved, resulting in the conversion of a full-length polypeptide into separate pro- and mature-domains. While the BDNF mature-domain is neuroprotective, the role of the pro-domain is less clear. In flies and mammalian cells, we have identified a synergistic toxic interaction between the amyloid-ß peptide (Aß1-42) and the pro-domains of both DNT1 and BDNF. Specifically, we show that DNT1 pro-domain acquires a neurotoxic activity in the presence of Aß1-42. In contrast, DNT1 mature-domain is protective against Aß1-42 toxicity. Likewise, in SH-SY5Y cell culture, BDNF pro-domain is toxic only in the presence of Aß1-42. Western blots indicate that this synergistic interaction likely results from the Aß1-42-induced upregulation of the BDNF pro-domain receptor p75(NTR). The clinical relevance of these findings is underlined by a greater than thirty fold increase in the ratio of BDNF pro- to mature-domains in the brains of individuals with AD. This unbalanced BDNF pro:mature-domain ratio in patients represents a possible biomarker of AD and may offer a target for therapeutic intervention.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Fatores de Crescimento Neural/metabolismo , Fragmentos de Peptídeos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/genética , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas de Drosophila/genética , Feminino , Marcadores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Crescimento Neural/genética , Fragmentos de Peptídeos/genética , Mapas de Interação de Proteínas , Receptor trkB/genética , Receptor trkB/metabolismo , Regulação para Cima
7.
Essays Biochem ; 56: 69-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25131587

RESUMO

The formation of amyloid aggregates is a feature of most, if not all, polypeptide chains. In vivo modelling of this process has been undertaken in the fruitfly Drosophila melanogaster with remarkable success. Models of both neurological and systemic amyloid diseases have been generated and have informed our understanding of disease pathogenesis in two main ways. First, the toxic amyloid species have been at least partially characterized, for example in the case of the Aß (amyloid ß-peptide) associated with Alzheimer's disease. Secondly, the genetic underpinning of model disease-linked phenotypes has been characterized for a number of neurodegenerative disorders. The current challenge is to integrate our understanding of disease-linked processes in the fly with our growing knowledge of human disease, for the benefit of patients.


Assuntos
Amiloidose/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloidose/patologia , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Proteínas tau/metabolismo
8.
Acta Neuropathol Commun ; 2: 43, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24725347

RESUMO

INTRODUCTION: The self-assembly of Aß peptides into a range of conformationally heterogeneous amyloid states represents a fundamental event in Alzheimer's disease. Within these structures oligomeric intermediates are considered to be particularly pathogenic. To test this hypothesis we have used a conformational targeting approach where particular conformational states, such as oligomers or fibrils, are recognized in vivo by state-specific antibody fragments. RESULTS: We show that oligomer targeting with the KW1 antibody fragment, but not fibril targeting with the B10 antibody fragment, affects toxicity in Aß-expressing Drosophila melanogaster. The effect of KW1 is observed to occur selectively with flies expressing Aß(1-40) and not with those expressing Aß(1-42) or the arctic variant of Aß(1-42) This finding is consistent with the binding preference of KW1 for Aß(1-40) oligomers that has been established in vitro. Strikingly, and in contrast to the previously demonstrated in vitro ability of this antibody fragment to block oligomeric toxicity in long-term potentiation measurements, KW1 promotes toxicity in the flies rather than preventing it. This result shows the crucial importance of the environment in determining the influence of antibody binding on the nature and consequences of the protein misfolding and aggregation. CONCLUSIONS: While our data support to the pathological relevance of oligomers, they highlight the issues to be addressed when developing inhibitory strategies that aim to neutralize these states by means of antagonistic binding agents.


Assuntos
Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Anticorpos/uso terapêutico , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Animais Geneticamente Modificados , Anticorpos/química , Anticorpos/genética , Anticorpos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Olho/metabolismo , Olho/ultraestrutura , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Agregação Patológica de Proteínas , Ligação Proteica/efeitos dos fármacos , Conformação Proteica
9.
Biochemistry ; 53(13): 2101-11, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24601543

RESUMO

The aggregation of ß-amyloid (Aß) into toxic oligomers is a hallmark of Alzheimer's disease pathology. Here we present a novel approach for the development of peptides capable of preventing amyloid aggregation based upon the previous selection of natural all-l peptides that bind Aß1-42. Using an intracellular selection system, successful library members were further screened via competition selection to identify the most effective peptides capable of reducing amyloid levels. To circumvent potential issues arising from stability and protease action for these structures, we have replaced all l residues with d residues and inverted the sequence. These retro-inverso (RI) peptide analogues therefore encompass reversed sequences that maintain the overall topological order of the native peptides. Our results demonstrate that efficacy in blocking and reversing amyloid formation is maintained while introducing desirable properties to the peptides. Thioflavin-T assays, circular dichroism, and oblique angle fluorescence microscopy collectively indicate that RI peptides can reduce amyloid load, while 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays demonstrate modest reductions in cell toxicity. These conclusions are reinforced using Drosophila melanogaster studies to monitor pupal hatching rates and fly locomotor activity in the presence of RI peptides delivered via RI-trans-activating transcriptional activator peptide fusions. We demonstrate that the RI-protein fragment complementation assay approach can be used as a generalized method for deriving Aß-interacting peptides. This approach has subsequently led to several peptide candidates being further explored as potential treatments for Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Animais , Dicroísmo Circular , Modelos Animais de Doenças , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Atividade Motora/efeitos dos fármacos , Células PC12 , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/isolamento & purificação , Placa Amiloide/tratamento farmacológico , Estrutura Secundária de Proteína , Ratos
10.
Biochim Biophys Acta ; 1832(12): 2115-26, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23911349

RESUMO

The accumulation of ß-amyloid (Aß) peptide in the brain is one of the pathological hallmarks of Alzheimer's disease and is thought to be of primary aetiological significance. In an unbiased genetic screen, we identified puromycin-sensitive aminopeptidase (PSA) as a potent suppressor of Aß toxicity in a Drosophila model system. We established that coexpression of Drosophila PSA (dPSA) in the flies' brains improved their lifespan, protected against locomotor deficits, and reduced brain Aß levels by clearing the Aß plaque-like deposits. However, confocal microscopy and subcellular fractionation of amyloid-expressing 7PA2 cells demonstrated that PSA localizes to the cytoplasm. Therefore, PSA and Aß are unlikely to be in the same cellular compartment; moreover, when we artificially placed them in the same compartment in flies, we could not detect a direct epistatic interaction. The consequent hypothesis that PSA's suppression of Aß toxicity is indirect was supported by the finding that Aß is not a proteolytic substrate for PSA in vitro. Furthermore, we showed that the enzymatic activity of PSA is not required for rescuing Aß toxicity in neuronal SH-SY5Y cells. We investigated whether the stimulation of autophagy by PSA was responsible for these protective effects. However PSA's promotion of autophagosome fusion with lysosomes required proteolytic activity and so its effect on autophagy is not identical to its protection against Aß toxicity.


Assuntos
Doença de Alzheimer/prevenção & controle , Aminopeptidases/farmacologia , Peptídeos beta-Amiloides/efeitos adversos , Encéfalo/metabolismo , Drosophila melanogaster/metabolismo , Neuroblastoma/prevenção & controle , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Animais Geneticamente Modificados , Autofagia , Western Blotting , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Proteólise , Puromicina/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
11.
J Biol Chem ; 288(11): 7606-7617, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23341460

RESUMO

Cell cycle checkpoints ensure that proliferation occurs only under permissive conditions, but their role in linking nutrient availability to cell division is incompletely understood. Protein folding within the endoplasmic reticulum (ER) is exquisitely sensitive to energy supply and amino acid sources because deficiencies impair luminal protein folding and consequently trigger ER stress signaling. Following ER stress, many cell types arrest within the G(1) phase, although recent studies have identified a novel ER stress G(2) checkpoint. Here, we report that ER stress affects cell cycle progression via two classes of signal: an early inhibition of protein synthesis leading to G(2) delay involving CHK1 and a later induction of G(1) arrest associated both with the induction of p53 target genes and loss of cyclin D(1). We show that substitution of p53/47 for p53 impairs the ER stress G(1) checkpoint, attenuates the recovery of protein translation, and impairs induction of NOXA, a mediator of cell death. We propose that cell cycle regulation in response to ER stress comprises redundant pathways invoked sequentially first to impair G(2) progression prior to ultimate G(1) arrest.


Assuntos
Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Genes p53 , Proteína Supressora de Tumor p53/genética , Animais , Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Separação Celular , Drosophila melanogaster , Citometria de Fluxo , Células HEK293 , Células HeLa , Humanos , Plasmídeos/metabolismo , Biossíntese de Proteínas , Proteína Fosfatase 1/metabolismo , Interferência de RNA , Proteína Supressora de Tumor p53/metabolismo
12.
FASEB J ; 26(1): 192-202, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21965601

RESUMO

We have created a Drosophila model of lysozyme amyloidosis to investigate the in vivo behavior of disease-associated variants. To achieve this objective, wild-type (WT) protein and the amyloidogenic variants F57I and D67H were expressed in Drosophila melanogaster using the UAS-gal4 system and both the ubiquitous and retinal expression drivers Act5C-gal4 and gmr-gal4. The nontransgenic w(1118) Drosophila line was used as a control throughout. We utilized ELISA experiments to probe lysozyme protein levels, scanning electron microscopy for eye phenotype classification, and immunohistochemistry to detect the unfolded protein response (UPR) activation. We observed that expressing the destabilized F57I and D67H lysozymes triggers UPR activation, resulting in degradation of these variants, whereas the WT lysozyme is secreted into the fly hemolymph. Indeed, the level of WT was up to 17 times more abundant than the variant proteins. In addition, the F57I variant gave rise to a significant disruption of the eye development, and this correlated to pronounced UPR activation. These results support the concept that the onset of familial amyloid disease is linked to an inability of the UPR to degrade completely the amyloidogenic lysozymes prior to secretion, resulting in secretion of these destabilized variants, thereby leading to deposition and associated organ damage.


Assuntos
Amiloidose/enzimologia , Anormalidades do Olho/enzimologia , Muramidase/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Amiloidose/patologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Estresse do Retículo Endoplasmático/fisiologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Proteínas de Fluorescência Verde/genética , Hemolinfa/enzimologia , Humanos , Masculino , Metamorfose Biológica/fisiologia , Microscopia Eletrônica de Varredura , Muramidase/genética , Células Fotorreceptoras de Invertebrados/enzimologia , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/ultraestrutura , Solubilidade
13.
Hum Mol Genet ; 21(1): 1-9, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21949354

RESUMO

It is unclear whether mutations in fused in sarcoma (FUS) cause familial amyotrophic lateral sclerosis via a loss-of-function effect due to titrating FUS from the nucleus or a gain-of-function effect from cytoplasmic overabundance. To investigate this question, we generated a series of independent Caenorhabditis elegans lines expressing mutant or wild-type (WT) human FUS. We show that mutant FUS, but not WT-FUS, causes cytoplasmic mislocalization associated with progressive motor dysfunction and reduced lifespan. The severity of the mutant phenotype in C. elegans was directly correlated with the severity of the illness caused by the same mutation in humans, arguing that this model closely replicates key features of the human illness. Importantly, the mutant phenotype could not be rescued by overexpression of WT-FUS, even though WT-FUS had physiological intracellular localization, and was not recruited to the cytoplasmic mutant FUS aggregates. Our data suggest that FUS mutants cause neuronal dysfunction by a dominant gain-of-function effect related either to neurotoxic aggregates of mutant FUS in the cytoplasm or to dysfunction in its RNA-binding functions.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Caenorhabditis elegans/metabolismo , Neurônios Motores/citologia , Mutação , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Morte Celular , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Neurônios Motores/metabolismo
14.
J Biol Chem ; 286(48): 41647-41655, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21998304

RESUMO

Therapies for Alzheimer disease that reduce the production of pathogenic amyloid ß (Aß) peptides have been associated with a range of unwanted effects. For this reason, alternative strategies that promote the clearance of the peptide by preventing its aggregation and deposition in the brain have been favored. In this context we have studied doxycycline, a member of the tetracycline family of antibiotics that has shown neuroprotective effects in a number of models of neurodegenerative disease. We investigated the neuroprotective potential of doxycycline in a Drosophila model of Aß toxicity and sought to correlate any effects with the aggregation state of the peptide. We found that administration of doxycycline to Aß42-expressing flies did not improve their lifespan but was able to slow the progression of their locomotor deficits. We also measured the rough eye phenotype of transgenic flies expressing the E22G variant of Aß42 and showed that doxycycline administration partially rescued the toxicity of Aß in the developing eye. We correlated these in vivo effects with in vitro observations using transmission electron microscopy, dynamic light scattering, and thioflavin T binding. We found that doxycycline prevents Aß fibrillization and favors the generation of smaller, non-amyloid structures that were non-toxic as determined by the lack of caspase 3 activation in a neuroblastoma cell line. Our confirmation that doxycycline can prevent amyloid ß toxicity both in vitro and in vivo supports its therapeutic potential in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Antibacterianos/farmacologia , Doxiciclina/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Animais Geneticamente Modificados , Benzotiazóis , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Drosophila melanogaster , Corantes Fluorescentes/farmacologia , Humanos , Longevidade/efeitos dos fármacos , Tiazóis/farmacologia
15.
J Biol Chem ; 286(6): 4248-56, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21147772

RESUMO

We have previously shown that overexpressing subunits of the iron-binding protein ferritin can rescue the toxicity of the amyloid ß (Aß) peptide in our Drosophila model system. These data point to an important pathogenic role for iron in Alzheimer disease. In this study, we have used an iron-selective chelating compound and RNAi-mediated knockdown of endogenous ferritin to further manipulate iron in the brain. We confirm that chelation of iron protects the fly from the harmful effects of Aß. To understand the pathogenic mechanisms, we have used biophysical techniques to see how iron affects Aß aggregation. We find that iron slows the progression of the Aß peptide from an unstructured conformation to the ordered cross-ß fibrils that are characteristic of amyloid. Finally, using mammalian cell culture systems, we have shown that iron specifically enhances Aß toxicity but only if the metal is present throughout the aggregation process. These data support the hypothesis that iron delays the formation of well ordered aggregates of Aß and so promotes its toxicity in Alzheimer disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Ferritinas/metabolismo , Ferro/metabolismo , Doença de Alzheimer/genética , Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular Tumoral , Drosophila melanogaster , Ferritinas/genética , Humanos
16.
J Cell Sci ; 123(Pt 17): 2892-900, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20682638

RESUMO

The integrated stress response (ISR) protects cells from numerous forms of stress and is involved in the growth of solid tumours; however, it is unclear how the ISR acts on cellular proliferation. We have developed a model of ISR signalling with which to study its effects on tissue growth. Overexpression of the ISR kinase PERK resulted in a striking atrophic eye phenotype in Drosophila melanogaster that could be rescued by co-expressing the eIF2alpha phosphatase GADD34. A genetic screen of 3000 transposon insertions identified grapes, the gene that encodes the Drosophila orthologue of checkpoint kinase 1 (CHK1). Knockdown of grapes by RNAi rescued eye development despite ongoing PERK activation. In mammalian cells, CHK1 was activated by agents that induce ER stress, which resulted in a G2 cell cycle delay. PERK was both necessary and sufficient for CHK1 activation. These findings indicate that non-genotoxic misfolded protein stress accesses DNA-damage-induced cell cycle checkpoints to couple the ISR to cell cycle arrest.


Assuntos
Proteínas Quinases/fisiologia , Estresse Fisiológico/fisiologia , Animais , Ciclo Celular/fisiologia , Proliferação de Células , Quinase 1 do Ponto de Checagem , Dano ao DNA , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/fisiologia , Ativação Enzimática , Olho/crescimento & desenvolvimento , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Fenótipo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Estresse Fisiológico/genética , Fosfatases cdc25/metabolismo , eIF-2 Quinase/biossíntese , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
17.
Front Biosci (Landmark Ed) ; 15(1): 373-96, 2010 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-20036826

RESUMO

Protein misfolding and aggregation are implicated in a wide range of increasingly prevalent human diseases ranging from dementia to diabetes. In this review we discuss the current experimental strategies that are being employed in the investigation of the pathogenesis of three important protein misfolding disorders. The first, Alzheimer's disease (AD), is the most prevalent neurodegenerative disease and is thought to be initiated by the aggregation of a natively unstructured peptide called amyloid beta (Abeta). We discuss methods for the characterization of the aggregation properties of Abeta in vitro and how the results of such experiments can be correlated with data from animal models of disease. We then consider another form of amyloidosis, where a systemic distribution of amyloid deposit is caused by aggregation and deposition of mutational variants of lysozyme. We describe how experiments in vitro, and more recently in vivo, have provided insights into the origins of this disease. Finally we outline the varied paradigms that have been employed in the study of the serpinopathies, and in particular, a dementia caused by neuroserpin polymerization.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Amiloidose/metabolismo , Muramidase/química , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Amiloidose/patologia , Animais , Dicroísmo Circular , Humanos , Microscopia Eletrônica de Transmissão , Muramidase/metabolismo , Conformação Proteica , Dobramento de Proteína
18.
J Biol Chem ; 284(34): 22793-802, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19549782

RESUMO

The serpinopathies are a family of diseases characterized by the accumulation of ordered polymers of mutant protein within the endoplasmic reticulum. They are a diverse group including alpha(1)-antitrypsin deficiency and the inherited dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. We have used transient transfection of COS7 cells and mouse embryonic fibroblasts, PC12 cell lines that conditionally express wild type and mutant neuroserpin and fly models of FENIB to assess the cellular handling of wild type and mutant serpins. By using a polymer-specific monoclonal antibody, we show that mutant neuroserpin forms polymers after a delay of at least 30 min and that polymers can be cleared in PC12 cell lines and from the brain in a fly model of FENIB. At steady state, the fractions of intracellular polymerogenic G392E mutant neuroserpin in the monomeric and polymeric states are comparable. Inhibition of the proteasome with MG132 reveals that both mutant neuroserpin and alpha(1)-antitrypsin are degraded predominantly by endoplasmic reticulum-associated degradation (ERAD). Pharmacological and genetic inhibitions demonstrate that autophagy is responsible for bulk turnover of wild type and mutant serpins, but can be stimulated by rapamycin to compensate for proteasome inhibition. The significance of these findings to the treatment of serpinopathies is discussed.


Assuntos
Autofagia/fisiologia , Retículo Endoplasmático/metabolismo , Mutação/genética , Neuropeptídeos/metabolismo , Serpinas/metabolismo , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Drosophila , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Imunoprecipitação , Leupeptinas/farmacologia , Macrolídeos/farmacologia , Camundongos , Neuropeptídeos/genética , Inibidores de Fosfoinositídeo-3 Quinase , Complexo de Endopeptidases do Proteassoma/metabolismo , Serpinas/genética , Neuroserpina
19.
Curr Opin Chem Biol ; 12(1): 25-31, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18295611

RESUMO

Protein misfolding is the underlying cause of many highly debilitating disorders ranging from Alzheimer's Disease to Cystic Fibrosis. Great strides have been made recently in understanding what causes proteins to misfold, primarily through the use of biophysical and computational techniques that enable systematic and quantitative analysis of the effects of a range of different perturbations in proteins. Correlation of the results of such analyses with observations made in animal models of disease has however been limited by their seemingly irreconcilable differences in methodology and scope. Several recent studies have however begun to overcome this limitation by combining the two approaches. This strategy has made it possible to investigate many of the consequences of protein misfolding in vivo, ranging from disease pathogenesis to epigenetic regulation, in the context of the fundamental physico-chemical principles derived from extensive and highly detailed studies undertaken in vitro.


Assuntos
Modelos Animais de Doenças , Doença , Dobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Animais , Humanos , Proteínas/genética
20.
Hum Mol Genet ; 17(11): 1527-39, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18267959

RESUMO

Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is an autosomal dominant dementia that is characterized by the retention of polymers of neuroserpin as inclusions within the endoplasmic reticulum (ER) of neurons. We have developed monoclonal antibodies that detect polymerized neuroserpin and have used COS-7 cells, stably transfected PC12 cell lines and transgenic Drosophila melanogaster to characterize the cellular handling of all four mutant forms of neuroserpin that cause FENIB. We show a direct correlation between the severity of the disease-causing mutation and the accumulation of neuroserpin polymers in cell and fly models of the disease. Moreover, mutant neuroserpin causes locomotor deficits in the fly allowing us to demonstrate a direct link between polymer accumulation and neuronal toxicity.


Assuntos
Demência/diagnóstico , Demência/metabolismo , Neuropeptídeos/análise , Neuropeptídeos/metabolismo , Serpinas/análise , Serpinas/metabolismo , Animais , Animais Geneticamente Modificados , Anticorpos Monoclonais/imunologia , Células COS , Chlorocebus aethiops , Demência/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Humanos , Neurônios/metabolismo , Neuropeptídeos/genética , Células PC12 , Polímeros/análise , Polímeros/metabolismo , Ratos , Serpinas/genética , Transfecção , Neuroserpina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA