Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
G3 (Bethesda) ; 14(2)2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38035793

RESUMO

Cutaneous melanoma is an aggressive type of skin cancer with a complex genetic landscape caused by the malignant transformation of melanocytes. This study aimed at providing an in silico network model based on the systematic profiling of the melanoma-associated genes considering germline mutations, somatic mutations, and genome-wide association study signals accounting for a total of 232 unique melanoma risk genes. A protein-protein interaction network was constructed using the melanoma risk genes as seeds and evaluated to describe the functional landscape in which the melanoma genes operate within the cellular milieu. Not only were the majority of the melanoma risk genes able to interact with each other at the protein level within the core of the network, but this showed significant enrichment for genes whose expression is altered in human melanoma specimens. Functional annotation showed the melanoma risk network to be significantly associated with processes related to DNA metabolism and telomeres, DNA damage and repair, cellular ageing, and response to radiation. We further explored whether the melanoma risk network could be used as an in silico tool to predict the efficacy of anti-melanoma phytochemicals, that are considered active molecules with potentially less systemic toxicity than classical cytotoxic drugs. A significant portion of the melanoma risk network showed differential expression when SK-MEL-28 human melanoma cells were exposed to the phytochemicals harmine and berberine chloride. This reinforced our hypothesis that the network modeling approach not only provides an alternative way to identify molecular pathways relevant to disease but it may also represent an alternative screening approach to prioritize potentially active compounds.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Melanócitos/metabolismo , Melanócitos/patologia
2.
Neurobiol Dis ; 174: 105858, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36096339

RESUMO

Mutations in SPG11, encoding spatacsin, constitute the major cause of autosomal recessive Hereditary Spastic Paraplegia (HSP) with thinning of the corpus callosum. Previous studies showed that spatacsin orchestrates cellular traffic events through the formation of a coat-like complex and its loss of function results in lysosomal and axonal transport impairments. However, the upstream mechanisms that regulate spatacsin trafficking are unknown. Here, using proteomics and CRISPR/Cas9-mediated tagging of endogenous spatacsin, we identified a subset of 14-3-3 proteins as physiological interactors of spatacsin. The interaction is modulated by Protein Kinase A (PKA)-dependent phosphorylation of spatacsin at Ser1955, which initiates spatacsin trafficking from the plasma membrane to the intracellular space. Our study provides novel insight in understanding spatacsin physio-pathological roles with mechanistic dissection of its associated pathways.


Assuntos
Proteínas 14-3-3 , Paraplegia Espástica Hereditária , Humanos , Proteínas 14-3-3/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Paraplegia Espástica Hereditária/genética , Mutação , Corpo Caloso/patologia , Proteínas/genética
3.
Int J Mol Sci ; 23(13)2022 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-35805938

RESUMO

Mutations in LRRK2 and GBA1 are key contributors to genetic risk of developing Parkinson's disease (PD). To investigate how LRRK2 kinase activity interacts with GBA and contributes to lysosomal dysfunctions associated with the pathology of PD. The activity of the lysosomal enzyme ß-Glucocerebrosidase (GCase) was assessed in a human neuroglioma cell model treated with two selective inhibitors of LRKK2 kinase activity (LRRK2-in-1 and MLi-2) and a GCase irreversible inhibitor, condutirol-beta-epoxide (CBE), under 24 and 72 h experimental conditions. We observed levels of GCase activity comparable to controls in response to 24 and 72 h treatments with LRRK2-in-1 and MLi-2. However, GBA protein levels increased upon 72 h treatment with LRRK2-in-1. Moreover, LC3-II protein levels were increased after both 24 and 72 h treatments with LRRK2-in-1, suggesting an activation of the autophagic pathway. These results highlight a possible regulation of lysosomal function through the LRRK2 kinase domain and suggest an interplay between LRRK2 kinase activity and GBA. Although further investigations are needed, the enhancement of GCase activity might restore the defective protein metabolism seen in PD.


Assuntos
Glucosilceramidase , Doença de Parkinson , Linhagem Celular , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glioma/metabolismo , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Humanos , Inositol/análogos & derivados , Inositol/farmacologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mutação , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
4.
Neurobiol Aging ; 116: 67-79, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35580510

RESUMO

Interactors of protein products of known genes for frontotemporal dementia (FTD) are likely to be involved in the molecular pathways towards disease. We therefore applied protein interaction network (PIN) analysis to prioritize candidate genes for rare variant association analysis. We created an FTD-PIN starting from known FTD genes downloading their physical interactors and performed functional enrichment analyses. We identified overrepresented processes in FTD and selected genes (n = 440) belonging to these processes for rare variant analysis in a Belgian cohort of 228 FTD patients and 345 controls. SKAT-O analysis suggested TNFAIP3 as the top gene (p = 0.7 × 10-3) reaching near test-wide significance (p = 2.5 × 10-4). We then analyzed the TNFAIP3-subnetwork within the FTD-PIN which indicated enrichment of several immune signaling networks, suggesting that disrupted immune signaling may be implicated in TNFAIP3-related FTD. Our study demonstrates that integration of PINs with genetic data is a useful approach to increase the power for rare variant association analysis. Furthermore, we present a computational pipeline for identifying potential novel therapeutic targets and risk-modifying variants.


Assuntos
Demência Frontotemporal , Mapas de Interação de Proteínas , Estudos de Coortes , Demência Frontotemporal/metabolismo , Humanos , Sistema Imunitário , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo
5.
Brain Res ; 1701: 75-84, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30055128

RESUMO

LRRK2, the gene encoding the multidomain kinase Leucine-Rich Repeat Kinase 2 (LRRK2), has been linked to familial and sporadic forms of Parkinson's disease (PD), as well as cancer, leprosy and Crohn's disease, establishing it as a target for discovery therapeutics. LRRK2 has been associated with a range of cellular processes, however its physiological and pathological functions remain unclear. The most prevalent LRRK2 mutations in PD have been shown to affect macroautophagy in various cellular models while a role in autophagy signalling has been recapitulated in vivo. Dysregulation of autophagy has been implicated in PD pathology, and this raises the possibility that differential autophagic activity is relevant to disease progression in PD patients carrying LRRK2 mutations. To examine the relevance of LRRK2 to the regulation of macroautophagy in a disease setting we examined the levels of autophagic markers in the basal ganglia of G2019S LRRK2 PD post-mortem tissue, in comparison to pathology-matched idiopathic PD (iPD), using immunoblotting (IB). Significantly lower levels of p62 and LAMP1 were observed in G2019S LRRK2 PD compared to iPD cases. Similarly, an increase in ULK1 was observed in iPD but was not reflected in G2019S LRRK2 PD cases. Furthermore, examination of p62 by immunohistochemistry (IH) recapitulated a distinct signature for G2019S PD. IH of LAMP1, LC3 and ULK1 broadly correlated with the IB results. Our data from a small but pathologically well-characterized cases highlights a divergence of G2019S PD carriers in terms of autophagic response in alpha-synuclein pathology affected brain regions compared to iPD.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/fisiologia , Corpos de Lewy/metabolismo , Doença de Parkinson/metabolismo , Idoso , Idoso de 80 Anos ou mais , Autofagia/fisiologia , Encéfalo/fisiopatologia , Feminino , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Corpos de Lewy/patologia , Proteínas de Membrana Lisossomal/análise , Proteínas de Membrana Lisossomal/metabolismo , Masculino , Doença de Parkinson/genética , Doença de Parkinson/fisiopatologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/metabolismo , alfa-Sinucleína/metabolismo
6.
Cancer Res ; 78(13): 3731-3739, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29794074

RESUMO

In addition to its well-recognized role in neurodegeneration, tau participates in maintenance of genome stability and chromosome integrity. In particular, peripheral cells from patients affected by frontotemporal lobar degeneration carrying a mutation in tau gene (genetic tauopathies), as well as cells from animal models, show chromosome numerical and structural aberrations, chromatin anomalies, and a propensity toward abnormal recombination. As genome instability is tightly linked to cancer development, we hypothesized that mutated tau may be a susceptibility factor for cancer. Here we conducted a retrospective cohort study comparing cancer incidence in families affected by genetic tauopathies to control families. In addition, we carried out a bioinformatics analysis to highlight pathways associated with the tau protein interactome. We report that the risk of developing cancer is significantly higher in families affected by genetic tauopathies, and a high proportion of tau protein interactors are involved in cellular processes particularly relevant to cancer. These findings disclose a novel role of tau as a risk factor for cancer, providing new insights in the various pathologic roles of mutated tau.Significance: This study reveals a novel role for tau as a risk factor for cancer, providing new insights beyond its role in neurodegeneration. Cancer Res; 78(13); 3731-9. ©2018 AACR.


Assuntos
Degeneração Lobar Frontotemporal/genética , Neoplasias/genética , Proteínas tau/genética , Biologia Computacional , Feminino , Predisposição Genética para Doença , Instabilidade Genômica/genética , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias/epidemiologia , Linhagem , Mapas de Interação de Proteínas/genética , Estudos Retrospectivos , Fatores de Risco
7.
Biosci Rep ; 38(2)2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29563162

RESUMO

Unc-51 Like Kinase 1 (ULK1) is a critical regulator of the biogenesis of autophagosomes, the central component of the catabolic macroautophagy pathway. Regulation of ULK1 activity is dependent upon several phosphorylation events acting to repress or activate the enzymatic function of this protein. Phosphorylation of Ser758 ULK1 has been linked to repression of autophagosome biogenesis and was thought to be exclusively dependent upon mTOR complex 1 kinase activity. In the present study, a novel regulation of Ser758 ULK1 phosphorylation is reported following prolonged inhibition of the Parkinson's disease linked protein leucine rich repeat kinase 2 (LRRK2). Here, modulation of Ser758 ULK1 phosphorylation following LRRK2 inhibition is decoupled from the repression of autophagosome biogenesis and independent of mTOR complex 1 activity.


Assuntos
Autofagossomos/enzimologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Linhagem Celular , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fosforilação/genética , Ratos , Serina/genética , Serina/metabolismo , Serina-Treonina Quinases TOR/genética
8.
J Proteome Res ; 16(2): 999-1013, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-28004582

RESUMO

The genetic analysis of complex disorders has undoubtedly led to the identification of a wealth of associations between genes and specific traits. However, moving from genetics to biochemistry one gene at a time has, to date, rather proved inefficient and under-powered to comprehensively explain the molecular basis of phenotypes. Here we present a novel approach, weighted protein-protein interaction network analysis (W-PPI-NA), to highlight key functional players within relevant biological processes associated with a given trait. This is exemplified in the current study by applying W-PPI-NA to frontotemporal dementia (FTD): We first built the state of the art FTD protein network (FTD-PN) and then analyzed both its topological and functional features. The FTD-PN resulted from the sum of the individual interactomes built around FTD-spectrum genes, leading to a total of 4198 nodes. Twenty nine of 4198 nodes, called inter-interactome hubs (IIHs), represented those interactors able to bridge over 60% of the individual interactomes. Functional annotation analysis not only reiterated and reinforced previous findings from single genes and gene-coexpression analyses but also indicated a number of novel potential disease related mechanisms, including DNA damage response, gene expression regulation, and cell waste disposal and potential biomarkers or therapeutic targets including EP300. These processes and targets likely represent the functional core impacted in FTD, reflecting the underlying genetic architecture contributing to disease. The approach presented in this study can be applied to other complex traits for which risk-causative genes are known as it provides a promising tool for setting the foundations for collating genomics and wet laboratory data in a bidirectional manner. This is and will be critical to accelerate molecular target prioritization and drug discovery.


Assuntos
Demência Frontotemporal/genética , Redes Reguladoras de Genes , Redes e Vias Metabólicas/genética , Mapeamento de Interação de Proteínas/métodos , Biologia de Sistemas , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Demência Frontotemporal/diagnóstico , Demência Frontotemporal/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Fenótipo , Proteína com Valosina
9.
Sci Rep ; 6: 35106, 2016 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-27731364

RESUMO

Leucine rich repeat kinase 2 is a complex enzyme with both kinase and GTPase activities, closely linked to the pathogenesis of several human disorders including Parkinson's disease, Crohn's disease, leprosy and cancer. LRRK2 has been implicated in numerous cellular processes; however its physiological function remains unclear. Recent reports suggest that LRRK2 can act to regulate the cellular catabolic process of macroautophagy, although the precise mechanism whereby this occurs has not been identified. To investigate the signalling events through which LRRK2 acts to influence macroautophagy, the mammalian target of rapamycin (mTOR)/Unc-51-like kinase 1 (ULK1) and Beclin-1/phosphatidylinositol 3-kinase (PI3K) pathways were evaluated in astrocytic cell models in the presence and absence of LRRK2 kinase inhibitors. Chemical inhibition of LRRK2 kinase activity resulted in the stimulation of macroautophagy in a non-canonical fashion, independent of mTOR and ULK1, but dependent upon the activation of Beclin 1-containing class III PI3-kinase.


Assuntos
Autofagia/fisiologia , Proteína Beclina-1/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Autofagia/efeitos dos fármacos , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular , Células Cultivadas , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Inibidores de Proteínas Quinases/farmacologia
10.
PeerJ ; 3: e778, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25737818

RESUMO

LRRK2 was identified in 2004 as the causative protein product of the Parkinson's disease locus designated PARK8. In the decade since then, genetic studies have revealed at least 6 dominant mutations in LRRK2 linked to Parkinson's disease, alongside one associated with cancer. It is now well established that coding changes in LRRK2 are one of the most common causes of Parkinson's. Genome-wide association studies (GWAs) have, more recently, reported single nucleotide polymorphisms (SNPs) around the LRRK2 locus to be associated with risk of developing sporadic Parkinson's disease and inflammatory bowel disorder. The functional research that has followed these genetic breakthroughs has generated an extensive literature regarding LRRK2 pathophysiology; however, there is still no consensus as to the biological function of LRRK2. To provide insight into the aspects of cell biology that are consistently related to LRRK2 activity, we analysed the plethora of candidate LRRK2 interactors available through the BioGRID and IntAct data repositories. We then performed GO terms enrichment for the LRRK2 interactome. We found that, in two different enrichment portals, the LRRK2 interactome was associated with terms referring to transport, cellular organization, vesicles and the cytoskeleton. We also verified that 21 of the LRRK2 interactors are genetically linked to risk for Parkinson's disease or inflammatory bowel disorder. The implications of these findings are discussed, with particular regard to potential novel areas of investigation.

11.
FEBS J ; 281(1): 261-74, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24286120

RESUMO

The human ROCO proteins are a family of multi-domain proteins sharing a conserved ROC-COR supra-domain. The family has four members: leucine-rich repeat kinase 1 (LRRK1), leucine-rich repeat kinase 2 (LRRK2), death-associated protein kinase 1 (DAPK1) and malignant fibrous histiocytoma amplified sequences with leucine-rich tandem repeats 1 (MASL1). Previous studies of LRRK1/2 and DAPK1 have shown that the ROC (Ras of complex proteins) domain can bind and hydrolyse GTP, but the cellular consequences of this activity are still unclear. Here, the first biochemical characterization of MASL1 and the impact of GTP binding on MASL1 complex formation are reported. The results demonstrate that MASL1, similar to other ROCO proteins, can bind guanosine nucleotides via its ROC domain. Furthermore, MASL1 exists in two distinct cellular complexes associated with heat shock protein 60, and the formation of a low molecular weight pool of MASL1 is modulated by GTP binding. Finally, loss of GTP enhances MASL1 toxicity in cells. Taken together, these data point to a central role for the ROC/GTPase domain of MASL1 in the regulation of its cellular function.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Chaperonina 60/metabolismo , Proteínas de Ligação a DNA/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas Oncogênicas/metabolismo , Sobrevivência Celular , Citometria de Fluxo , Células HEK293 , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Biochim Biophys Acta ; 1833(12): 2900-2910, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23916833

RESUMO

Leucine Rich Repeat Kinase 2 (LRRK2) is one of the most important genetic contributors to Parkinson's disease. LRRK2 has been implicated in a number of cellular processes, including macroautophagy. To test whether LRRK2 has a role in regulating autophagy, a specific inhibitor of the kinase activity of LRRK2 was applied to human neuroglioma cells and downstream readouts of autophagy examined. The resulting data demonstrate that inhibition of LRRK2 kinase activity stimulates macroautophagy in the absence of any alteration in the translational targets of mTORC1, suggesting that LRRK2 regulates autophagic vesicle formation independent of canonical mTORC1 signaling. This study represents the first pharmacological dissection of the role LRRK2 plays in the autophagy/lysosomal pathway, emphasizing the importance of this pathway as a marker for LRRK2 physiological function. Moreover it highlights the need to dissect autophagy and lysosomal activities in the context of LRRK2 related pathologies with the final aim of understanding their aetiology and identifying specific targets for disease modifying therapies in patients.


Assuntos
Autofagia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Autofagia/efeitos dos fármacos , Benzodiazepinonas/farmacologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Imuno-Histoquímica , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Proteínas Associadas aos Microtúbulos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Proteína Sequestossoma-1
13.
Biochem Soc Trans ; 40(5): 1147-51, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22988880

RESUMO

LRRK2 (leucine-rich repeat kinase 2) is an enzyme implicated in human disease, containing kinase and GTPase functions within the same multidomain open reading frame. Dominant mutations in the LRRK2 gene are the most common cause of familial PD (Parkinson's disease). Additionally, in genome-wide association studies, the LRRK2 locus has been linked to risk of PD, Crohn's disease and leprosy, and LRRK2 has also been linked with cancer. Despite its association with human disease, very little is known about its pathophysiology. Recent reports suggest a functional association between LRRK2 and autophagy. Implications of this set of data for our understanding of LRRK2's role in physiology and disease are discussed in the present paper.


Assuntos
Autofagia , Doença de Parkinson/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Mutação , Doença de Parkinson/enzimologia , Proteínas Serina-Treonina Quinases/genética
14.
Sci Signal ; 5(207): pe2, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22253261

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is linked to various diseases, including Parkinson's disease, cancer, and leprosy. Data from LRRK2 knockout mice has highlighted a possible role for LRRK2 in regulating signaling pathways that are linked to the pathogenesis of Crohn's disease. Here, we examine how LRRK2's role as a signaling hub in the cell could lead to diverse pathologies.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Doença de Crohn/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Camundongos , Camundongos Knockout , Modelos Biológicos , Modelos Genéticos , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais
15.
PLoS One ; 6(9): e24909, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21966382

RESUMO

Accumulation of ß-sheet-rich peptide (Aß) is strongly associated with Alzheimer's disease, characterized by reduction in synapse density, structural alterations of dendritic spines, modification of synaptic protein expression, loss of long-term potentiation and neuronal cell death. Aß species are potent neurotoxins, however the molecular mechanism responsible for Aß toxicity is still unknown. Numerous mechanisms of toxicity were proposed, although there is no agreement about their relative importance in disease pathogenesis. Here, the toxicity of Aß 1-40 and Aß 1-42 monomers, oligomers or fibrils, was evaluated using the N2a cell line. A structure-function relationship between peptide aggregation state and toxic properties was established. Moreover, we demonstrated that Aß toxic species cross the plasma membrane, accumulate in cells and bind to a variety of internal proteins, especially on the cytoskeleton and in the endoplasmatic reticulum (ER). Based on these data we suggest that numerous proteins act as Aß receptors in N2a cells, triggering a multi factorial toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Catepsina D/metabolismo , Proteínas de Choque Térmico/metabolismo , Vimentina/metabolismo , Peptídeos beta-Amiloides/química , Animais , Far-Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Humanos , Imuno-Histoquímica , Lisossomos/metabolismo , Microscopia de Fluorescência , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Ligação Proteica , Multimerização Proteica
16.
Rev. CEFAC ; 13(1): 102-111, jan.-fev. 2011. ilus, tab
Artigo em Português | LILACS | ID: lil-576214

RESUMO

OBJETIVO: avaliar a adequação de um instrumento, que está em fase de implantação, elaborado pela Secretaria Municipal de Saúde de São Paulo quanto ao serviço de saúde e uso da prótese auditiva. MÉTODOS: instrumento composto por 17 perguntas que avaliam o serviço e a satisfação do usuário. A avaliação do serviço refere ao atendimento de forma geral, o tempo de espera pelo aparelho, e o(s) retorno(s) após recebimento do aparelho. A satisfação do usuário avalia o tempo de uso do(s) aparelho(s) durante o dia e em quais momentos na vida do paciente o seu uso trouxe melhora. Foi aplicado em 17 sujeitos, com idades entre 21 a 91 anos, média de 61,8 anos. Onze eram do sexo masculino. RESULTADOS: no indicador para avaliação no serviço sugere valorar sobre testagem com mais de um aparelho. No indicador da satisfação há necessidade de modificar a pontuação quanto ao tempo de uso da prótese e quanto à melhora em determinados locais, há ainda necessidade de acréscimo de local de uso do aparelho. CONCLUSÃO: faz-se necessária a reformulação e reavaliação do instrumento apresentado. Apesar da limitação devido ao pequeno número de indivíduos pesquisados, verificou-se que os respondentes estão relativamente satisfeitos, porém há queixas quanto à adaptação da prótese auditiva.


PURPOSE: assess the adequacy of a questionnaire, which is in the implementation phase, elaborated by the Municipal Department of Health of São Paulo regarding the hearing health service and the use of hearing aids. METHODS: the questionnaire consists of 17 questions that assess the service and user's satisfaction. The assessment of the service comprises the medical care in general, waiting time for the hearing aid, and the return(s) after receiving it. User's satisfaction assesses the period of time when the hearing aid is used during the day and at what times in the patient's life its use has brought improvement. It was applied for 17 subjects, ages 21 to 91 years, mean age: 61.8 years. Eleven were male. RESULTS: the indicator for service assessment suggests pondering over testing with more than one device. The indicator for satisfaction suggests the need to modify the score regarding the period of time the device is used and, regarding the improvement in certain places, it is necessary to add the place where the device is being used. CONCLUSION: it is necessary to reformulate and reevaluate the submitted questionnaire. Despite its limitation due to the small number o studied f individuals, it was found that the respondents were relatively satisfied, although there are complaints as for the adaptability to the hearing aid.

17.
J Neurochem ; 110(5): 1445-56, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19558452

RESUMO

Human sirtuins are a family of seven conserved proteins (SIRT1-7). The most investigated is the silent mating type information regulation-2 homolog (SIRT1, NM_012238), which was associated with neuroprotection in models of polyglutamine toxicity or Alzheimer's disease (AD) and whose activation by the phytocompound resveratrol (RES) has been described. We have examined the neuroprotective role of RES in a cellular model of oxidative stress, a common feature of neurodegeneration. RES prevented toxicity triggered by hydrogen peroxide or 6-hydroxydopamine (6-OHDA). This action was likely mediated by SIRT1 activation, as the protection was lost in the presence of the SIRT1 inhibitor sirtinol and when SIRT1 expression was down-regulated by siRNA approach. RES was also able to protect SK-N-BE from the toxicity arising from two aggregation-prone proteins, the AD-involved amyloid-beta (1-42) peptide (Abeta42) and the familiar Parkinson's disease linked alpha-synuclein(A30P) [alpha-syn(A30P)]. Alpha-syn(A30P) toxicity was restored by sirtinol addition, while a partial RES protective effect against Abeta42 was found even in presence of sirtinol, thus suggesting a direct RES effect on Abeta42 fibrils. We conclude that SIRT1 activation by RES can prevent in our neuroblastoma model the deleterious effects triggered by oxidative stress or alpha-syn(A30P) aggregation, while RES displayed a SIRT1-independent protective action against Abeta42.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Sirtuínas/metabolismo , Estilbenos/farmacologia , alfa-Sinucleína/toxicidade , Sequência de Aminoácidos , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Dados de Sequência Molecular , Estresse Oxidativo/fisiologia , Resveratrol , Sirtuína 1 , Sirtuínas/agonistas
18.
Amyloid ; 16(2): 71-80, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20536398

RESUMO

Investigations of amyloidogenic diseases use synthetic peptides for cell-free and in vitro studies. However, amyloidogenic peptides often show intrinsic variability that markedly affects the reproducibility of experiments. Proof of physicochemical and biological variability with different batches of amyloidogenic peptides have been reported in literature. Here, we show that differences can be observed even within the same batch of Abeta1-42 peptide after storing lyophilised samples at -20 degrees C. This change (referred to as 'peptide aging') was reproduced with Abeta1-40 peptide samples by using a series of lyophilisation cycles, showing that lyophilisation, rather than preserving the physicochemical and biological features of Abeta peptides, introduces wide variability. To counteract synthetic peptide aging, we set up a procedure involving the sequential use of trifluoroacetic acid, formic acid and sodium hydroxide solutions that disaggregate preformed seeds and enriched Abeta peptide solutions into monomers and low-molecular-weight oligomers. This procedure enabled us to obtain reproducible physicochemical and biological features of Abeta peptides, irrespective of their age.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos/química , Sequência de Aminoácidos , Peptídeos beta-Amiloides/ultraestrutura , Western Blotting , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Poliacrilamida , Liofilização , Microscopia Eletrônica , Dados de Sequência Molecular , Peso Molecular
19.
J Mol Biol ; 381(5): 1349-61, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18619462

RESUMO

The existence of several prion strains and their capacity of overcoming species barriers seem to point to a high conformational adaptability of the prion protein. To investigate this structural plasticity, we studied here the aggregation pathways of the human prion peptide PrP82-146, a major component of the Gerstmann-Sträussler-Scheinker amyloid disease. By Fourier transform infrared (FT-IR) spectroscopy, electron microscopy, and atomic force microscopy (AFM), we monitored the time course of PrP82-146 fibril formation. After incubation at 37 degrees C, the unfolded peptide was found to aggregate into oligomers characterized by intermolecular beta-sheet infrared bands. At a critical oligomer concentration, the emergence of a new FT-IR band allowed to detect fibril formation. A different intermolecular beta-sheet interaction of the peptides in oligomers and in fibrils is, therefore, detected by FT-IR spectroscopy, which, in addition, suggests a parallel orientation of the cross beta-sheet structures of PrP82-146 fibrils. By AFM, a wide distribution of PrP82-146 oligomer volumes--the smallest ones containing from 5 to 30 peptides--was observed. Interestingly, the statistical analysis of AFM data enabled us to detect a quantization in the oligomer height values differing by steps of approximately 0.5 nm that could reflect an orientation of oligomer beta-strands parallel with the sample surface. Different morphologies were also detected for fibrils that displayed high heterogeneity in their twisting periodicity and a complex hierarchical assembly. Thermal aggregation of PrP82-146 was also investigated by FT-IR spectroscopy, which indicated for these aggregates an intermolecular beta-sheet interaction different from that observed for oligomers and fibrils. Unexpectedly, random aggregates, induced by solvent evaporation, were found to display a significant alpha-helical structure as well as several beta-sheet components. All these results clearly point to a high plasticity of the PrP82-146 peptide, which was found to be capable of undergoing several aggregation pathways, with end products displaying different secondary structures and intermolecular interactions.


Assuntos
Doença de Gerstmann-Straussler-Scheinker/metabolismo , Peptídeos/química , Príons/química , Amiloide/ultraestrutura , Humanos , Cinética , Microscopia Eletrônica , Microscopia de Fluorescência , Modelos Biológicos , Príons/ultraestrutura , Estrutura Quaternária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier
20.
J Neurosci ; 27(7): 1576-83, 2007 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-17301166

RESUMO

Amyloid fibrils in Gerstmann-Sträussler-Scheinker (GSS) disease are composed of a fragment of the prion protein (PrP), the N and C termini of which correspond to ragged residues 81-90 and 144-153. A synthetic peptide spanning the sequence 82-146 (PrP 82-146) polymerizes into protease-resistant fibrils with the tinctorial properties of amyloid. We investigated the biological activity of PrP 82-146 and of two nonamyloidogenic variants of PrP 82-146 with scrambled amino acid sequence 106-126 or 127-146. Cortical neurons prepared from rat and mouse embryos were chronically exposed to the PrP 82-146 peptides (10-50 microM). PrP 82-146 and the partially scrambled peptides induced neuronal death with a similar dose-response pattern, indicating that neurotoxicity was independent of amyloid fibril formation. Neurotoxicity was significantly reduced by coadministration of an anti-oligomer antibody, suggesting that PrP 82-146 oligomers are primarily responsible for triggering cell death. Neurons from PrP knock-out (Prnp0/0) mice were significantly less sensitive to PrP 82-146 toxicity than neurons expressing PrP. The gliotrophic effect of PrP 82-146 was determined by [methyl-3H]-thymidine incorporation in cultured astrocytes. Treatment with PrP 82-146 stimulated [methyl-3H]-thymidine uptake 3.5-fold. This activity was significantly less when the 106-126 or 127-146 regions were disrupted, indicating that PrP 82-146 amyloid activates the gliotrophic response. Prnp0/0 astrocytes were insensitive to the proliferative stimulus of PrP 82-146. These results underline the role of cerebral accumulation of abnormally folded PrP fragments and indicate that cellular PrP governs the pathogenic process.


Assuntos
Amiloide/química , Astrócitos/efeitos dos fármacos , Doença de Gerstmann-Straussler-Scheinker/metabolismo , Neurônios/efeitos dos fármacos , Proteínas PrPSc/toxicidade , Sequência de Aminoácidos , Amiloide/ultraestrutura , Análise de Variância , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Neuroblastoma , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/toxicidade , Fragmentos de Peptídeos/ultraestrutura , Fosfopiruvato Hidratase/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/ultraestrutura , Ratos , Timidina/metabolismo , Fatores de Tempo , Trítio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA