Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Aging ; 99: 79-85, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33422896

RESUMO

Accumulation of phosphorylated tau (p-tau) has long been an underappreciated hallmark of Alzheimer's disease. Tau is one of the major components of microtubule networks in neurons, and its abnormal phosphorylation and aggregation are closely related to the impairment of axonal transport. Abnormalities in axonal transport can impede autophagy in neurons, interrupting the autophagic clearance of amyloid beta. The ubiquitin proteasome system (UPS) maintains intracellular proteostasis by degrading abnormal or redundant proteins. Ever-mounting evidence suggests that UPS deficits contribute to p-tau accumulation. And targeting UPS attenuates tau pathology. This review endeavors to exam the potential role of UPS in p-tau aggregation, and how pathogenic tau may inflict other abnormalities such as amyloid beta accumulation in Alzheimer's disease.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Ubiquitina/metabolismo , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Autofagia/fisiologia , Transporte Axonal/fisiologia , Humanos , Fosforilação , Agregação Patológica de Proteínas/enzimologia , Proteostase
2.
Acta Neuropathol Commun ; 8(1): 96, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32611392

RESUMO

Accumulation of misfolded phosphorylated Tau (Tauopathy) can be triggered by mutations or by trauma, and is associated with synapse loss, gliosis, neurodegeneration and memory deficits. Fyn kinase physically associates with Tau and regulates subcellular distribution. Here, we assessed whether pharmacological Fyn inhibition alters Tauopathy. In P301S transgenic mice, chronic Fyn inhibition prevented deficits in spatial memory and passive avoidance learning. The behavioral improvement was coupled with reduced accumulation of phospho-Tau in the hippocampus, with reductions in glial activation and with recovery of presynaptic markers. We extended this analysis to a trauma model in which very mild repetitive closed head injury was paired with chronic variable stress over 2 weeks to produce persistent memory deficits and Tau accumulation. In this model, Fyn inhibition beginning 24 h after the trauma ended rescued memory performance and reduced phospho-Tau accumulation. Thus, inhibition of Fyn kinase may have therapeutic benefit in clinical Tauopathies.


Assuntos
Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Sinapses/patologia , Tauopatias/patologia , Proteínas tau/efeitos dos fármacos , Proteínas tau/metabolismo , Idoso de 80 Anos ou mais , Animais , Benzodioxóis/farmacologia , Concussão Encefálica/complicações , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/patologia , Quinazolinas/farmacologia , Tauopatias/etiologia , Tauopatias/metabolismo
3.
Neurosci Lett ; 678: 131-137, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29775672

RESUMO

Proteasome and lysosome are responsible for the homeostasis of proteins, lipids and carbohydrates in cells. Numerous reports indicate the proteolytic pathways have altered functions during neurodegeneration and aging. Dementia with Lewy bodies (DLB) is one of the leading forms of dementia, and the proteolytic alteration in DLB has not yet been fully investigated. This study shows that the components of proteasome and lysosome had selectively altered gene expression and enzymatic functions. Specifically, PSMB8, an inducible proteasomal ß subunit, had elevated mRNA level and protein level in DLB brain compared with age-matched controls. The proteasomal caspase-like peptidase showed significant decreased activity in DLB brains and the trypsin-like/chemotrypsin-like activities did not reach statistical significance. Lysosomal cathepsin B and D had elevated mRNA levels while only cathepsin B showed elevated enzymatic activity in DLB brains. This data indicate that the alteration of proteolytic pathways is highly selective and comprehensive. Further study to elucidate the correlation between neurodegenerative development and the alteration of proteolytic pathways would be important for therapeutic development.


Assuntos
Encéfalo/enzimologia , Catepsinas/metabolismo , Doença por Corpos de Lewy/enzimologia , Lisossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Expressão Gênica , Humanos , Masculino , Agregação Patológica de Proteínas/enzimologia , Proteólise , Regulação para Cima
4.
Proc Natl Acad Sci U S A ; 115(18): 4661-4665, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29666246

RESUMO

Aberrant accumulation of misfolded Cu, Zn superoxide dismutase (SOD1) is a hallmark of SOD1-associated amyotrophic lateral sclerosis (ALS), an invariably fatal neurodegenerative disease. While recent discovery of nonnative trimeric SOD1-associated neurotoxicity has suggested a potential pathway for motor neuron impairment, it is yet unknown whether large, insoluble aggregates are cytotoxic. Here we designed SOD1 mutations that specifically stabilize either the fibrillar form or the trimeric state of SOD1. The designed mutants display elevated populations of fibrils or trimers correspondingly, as demonstrated by gel filtration chromatography and electron microscopy. The trimer-stabilizing mutant, G147P, promoted cell death, even more potently in comparison with the aggressive ALS-associated mutants A4V and G93A. In contrast, the fibril-stabilizing mutants, N53I and D101I, positively impacted the survival of motor neuron-like cells. Hence, we conclude the SOD1 oligomer and not the mature form of aggregated fibril is critical for the neurotoxic effects in the model of ALS. The formation of large aggregates is in competition with trimer formation, suggesting that aggregation may be a protective mechanism against formation of toxic oligomeric intermediates.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Modelos Biológicos , Agregação Patológica de Proteínas/enzimologia , Superóxido Dismutase-1/metabolismo , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Agregação Patológica de Proteínas/genética , Superóxido Dismutase/genética , Superóxido Dismutase-1/genética
6.
Crit Rev Biochem Mol Biol ; 52(5): 554-582, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28635330

RESUMO

Insulin-degrading enzyme (IDE) is a ubiquitous zinc peptidase of the inverzincin family, which has been initially discovered as the enzyme responsible for insulin catabolism; therefore, its involvement in the onset of diabetes has been largely investigated. However, further studies on IDE unraveled its ability to degrade several other polypeptides, such as ß-amyloid, amylin, and glucagon, envisaging the possible implication of IDE dys-regulation in the "aggregopathies" and, in particular, in neurodegenerative diseases. Over the last decade, a novel scenario on IDE biology has emerged, pointing out a multi-functional role of this enzyme in several basic cellular processes. In particular, latest advances indicate that IDE behaves as a heat shock protein and modulates the ubiquitin-proteasome system, suggesting a major implication in proteins turnover and cell homeostasis. In addition, recent observations have highlighted that the regulation of glucose metabolism by IDE is not merely based on its largely proposed role in the degradation of insulin in vivo. There is increasing evidence that improper IDE function, regulation, or trafficking might contribute to the etiology of metabolic diseases. In addition, the enzymatic activity of IDE is affected by metals levels, thus suggesting a role also in the metal homeostasis (metallostasis), which is thought to be tightly linked to the malfunction of the "quality control" machinery of the cell. Focusing on the physiological role of IDE, we will address a comprehensive vision of the very complex scenario in which IDE takes part, outlining its crucial role in interconnecting several relevant cellular processes.


Assuntos
Insulisina/metabolismo , Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Animais , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Humanos , Insulisina/fisiologia , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/patologia , Conformação Proteica
7.
Acta Neuropathol ; 134(1): 113-127, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28527045

RESUMO

Neuronal loss in numerous neurodegenerative disorders has been linked to protein aggregation and oxidative stress. Emerging data regarding overlapping proteinopathy in traditionally distinct neurodegenerative diseases suggest that disease-modifying treatments targeting these pathological features may exhibit efficacy across multiple disorders. Here, we describe proteinopathy distinct from classic synucleinopathy, predominantly comprised of the anti-oxidant enzyme superoxide dismutase-1 (SOD1), in the Parkinson's disease brain. Significant expression of this pathology closely reflected the regional pattern of neuronal loss. The protein composition and non-amyloid macrostructure of these novel aggregates closely resembles that of neurotoxic SOD1 deposits in SOD1-associated familial amyotrophic lateral sclerosis (fALS). Consistent with the hypothesis that deposition of protein aggregates in neurodegenerative disorders reflects upstream dysfunction, we demonstrated that SOD1 in the Parkinson's disease brain exhibits evidence of misfolding and metal deficiency, similar to that seen in mutant SOD1 in fALS. Our data suggest common mechanisms of toxic SOD1 aggregation in both disorders and a potential role for SOD1 dysfunction in neuronal loss in the Parkinson's disease brain. This shared restricted proteinopathy highlights the potential translation of therapeutic approaches targeting SOD1 toxicity, already in clinical trials for ALS, into disease-modifying treatments for Parkinson's disease.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Encéfalo/patologia , Doença de Parkinson/patologia , Superóxido Dismutase-1/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Esclerose Lateral Amiotrófica/enzimologia , Encéfalo/enzimologia , Contagem de Células , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Corpos de Lewy/enzimologia , Corpos de Lewy/patologia , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , Neurônios/enzimologia , Neurônios/patologia , Doença de Parkinson/enzimologia , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/patologia , Dobramento de Proteína , Medula Espinal/enzimologia , Medula Espinal/patologia
8.
J Clin Invest ; 127(4): 1438-1450, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28263187

RESUMO

Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Doença de Huntington/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Agregação Patológica de Proteínas/enzimologia , Animais , Feminino , Células HEK293 , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Camundongos Transgênicos , Mutação
9.
Methods Enzymol ; 584: 157-183, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28065262

RESUMO

γ-Secretase is a key player in the pathogenesis of Alzheimer's disease (AD). The intramembrane-cleaving enzyme initially cleaves a C-terminal fragment of the amyloid precursor protein (APP) at the ɛ-site within its transmembrane domain to release the APP intracellular domain. Subsequent stepwise carboxy-terminal trimming cleavages eventually release amyloid-ß (Aß) peptides of 37-43 amino acids into the extracellular space. Aß42 as well as the much less abundant Aß43 species are highly aggregation prone and can deposit as plaques in the brains of affected patients, which are widely believed to be causative of AD. Disappointingly, due to lack of efficacy and side effects likely attributable to the inhibition of the crucial substrate Notch, inhibitors of γ-secretase that lower Aß generation failed in clinical trials of AD. There is hope, however, that recently developed potent γ-secretase modulators (GSMs) provide a safer approach for disease modification. These compounds have the unique property of primarily shifting the generation of Aß42 toward that of shorter peptides without affecting the ɛ-site cleavage of Notch and other substrates. In this chapter, we describe methods to investigate how GSMs affect the activity of the enzyme as well as how their molecular targets are identified.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Peptídeos beta-Amiloides/química , Ativadores de Enzimas/química , Biologia Molecular/métodos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/agonistas , Sítios de Ligação , Ativadores de Enzimas/uso terapêutico , Humanos , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/enzimologia
10.
J Biol Chem ; 291(41): 21363-21374, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27551041

RESUMO

Congenital insensitivity to pain with anhidrosis (CIPA) is a rare autosomal recessive disorder characterized by insensitivity to noxious stimuli and variable intellectual disability (ID) due to mutations in the NTRK1 gene encoding the NGF receptor TrkA. To get an insight in the effect of NTRK1 mutations in the cognitive phenotype we biochemically characterized three TrkA mutations identified in children diagnosed of CIPA with variable ID. These mutations are located in different domains of the protein; L213P in the extracellular domain, Δ736 in the kinase domain, and C300stop in the extracellular domain, a new mutation causing CIPA diagnosed in a Spanish teenager. We found that TrkA mutations induce misfolding, retention in the endoplasmic reticulum (ER), and aggregation in a mutation-dependent manner. The distinct mutations are degraded with a different kinetics by different ER quality control mechanisms; although C300stop is rapidly disposed by autophagy, Δ736 degradation is sensitive to the proteasome and to autophagy inhibitors, and L213P is a long-lived protein refractory to degradation. In addition L213P enhances the formation of autophagic vesicles triggering an increase in the autophagic flux with deleterious consequences. Mouse cortical neurons expressing L213P showed the accumulation of LC3-GFP positive puncta and dystrophic neurites. Our data suggest that TrkA misfolding and aggregation induced by some CIPA mutations disrupt the autophagy homeostasis causing neurodegeneration. We propose that distinct disease-causing mutations of TrkA generate different levels of cell toxicity, which may provide an explanation of the variable intellectual disability observed in CIPA patients.


Assuntos
Autofagia , Hipo-Hidrose/enzimologia , Mutação de Sentido Incorreto , Doenças Neurodegenerativas/enzimologia , Insensibilidade Congênita à Dor/enzimologia , Agregação Patológica de Proteínas/enzimologia , Deficiências na Proteostase/enzimologia , Receptor trkA/metabolismo , Adolescente , Substituição de Aminoácidos , Animais , Córtex Cerebral/enzimologia , Feminino , Células HeLa , Humanos , Hipo-Hidrose/genética , Masculino , Camundongos , Camundongos Mutantes , Doenças Neurodegenerativas/genética , Nociceptores/enzimologia , Insensibilidade Congênita à Dor/genética , Agregação Patológica de Proteínas/genética , Deficiências na Proteostase/genética , Receptor trkA/genética
11.
J Biol Chem ; 291(33): 17197-208, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27261461

RESUMO

Protein misfolding is implicated in neurodegenerative diseases such as ALS, where mutations of superoxide dismutase 1 (SOD1) account for about 20% of the inherited mutations. Human SOD1 (hSOD1) contains four cysteines, including Cys(57) and Cys(146), which have been linked to protein stability and folding via forming a disulfide bond, and Cys(6) and Cys(111) as free thiols. But the roles of the cellular oxidation-reduction (redox) environment in SOD1 folding and aggregation are not well understood. Here we explore the effects of cellular redox systems on the aggregation of hSOD1 proteins. We found that the known hSOD1 mutations G93A and A4V increased the capability of the thioredoxin and glutaredoxin systems to reduce hSOD1 compared with wild-type hSOD1. Treatment with inhibitors of these redox systems resulted in an increase of hSOD1 aggregates in the cytoplasm of cells transfected with mutants but not in cells transfected with wild-type hSOD1 or those containing a secondary C111G mutation. This aggregation may be coupled to changes in the redox state of the G93A and A4V mutants upon mild oxidative stress. These results strongly suggest that the thioredoxin and glutaredoxin systems are the key regulators for hSOD1 aggregation and may play critical roles in the pathogenesis of ALS.


Assuntos
Esclerose Lateral Amiotrófica , Estresse Oxidativo , Agregação Patológica de Proteínas , Dobramento de Proteína , Superóxido Dismutase-1 , Substituição de Aminoácidos , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/genética , Animais , Linhagem Celular Tumoral , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Humanos , Mutação de Sentido Incorreto , Oxirredução , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/genética , Ratos , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
12.
Cell Mol Neurobiol ; 36(3): 377-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26908139

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neuromuscular degenerative disorder with a poorly defined etiology. ALS patients experience motor weakness, which starts focally and spreads throughout the nervous system, culminating in paralysis and death within a few years of diagnosis. While the vast majority of clinical ALS is sporadic with no known cause, mutations in human copper-zinc superoxide dismutase 1 (SOD1) cause about 20 % of inherited cases of ALS. ALS with SOD1 mutations is caused by a toxic gain of function associated with the propensity of mutant SOD1 to misfold, presenting a non-native structure. The mechanisms responsible for the progressive spreading of ALS pathology have been the focus of intense study. We have shown that misfolded SOD1 protein can seed misfolding and aggregation of endogenous wild-type SOD1 similar to amyloid-ß and prion protein seeding. Our recent observations demonstrate a transfer of the misfolded SOD1 species from cell to cell, modeling the intercellular transmission of disease through the neuroaxis. We have shown that both mutant and misfolded wild-type SOD1 can traverse cell-to-cell, either as protein aggregates that are released from dying cells and taken up by neighboring cells via macropinocytosis, or in association with vesicles which are released into the extracellular environment. Furthermore, once misfolding of wild-type SOD1 has been initiated in a human cell culture, it can induce misfolding in naïve cell cultures over multiple passages of media transfer long after the initial misfolding template is degraded. Herein we review the data on mechanisms of intercellular transmission of misfolded SOD1.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/patologia , Exossomos/metabolismo , Dobramento de Proteína , Transdução de Sinais , Superóxido Dismutase-1/química , Superóxido Dismutase-1/metabolismo , Animais , Humanos , Agregação Patológica de Proteínas/enzimologia
13.
Antioxid Redox Signal ; 23(6): 550-64, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26094487

RESUMO

AIMS: Loss-of-function mutations in GBA1, which cause the autosomal recessive lysosomal storage disease, Gaucher disease (GD), are also a key genetic risk factor for the α-synucleinopathies, including Parkinson's disease (PD) and dementia with Lewy bodies. GBA1 encodes for the lysosomal hydrolase glucocerebrosidase and reductions in this enzyme result in the accumulation of the glycolipid substrates glucosylceramide and glucosylsphingosine. Deficits in autophagy and lysosomal degradation pathways likely contribute to the pathological accumulation of α-synuclein in PD. In this report we used conduritol-ß-epoxide (CBE), a potent selective irreversible competitive inhibitor of glucocerebrosidase, to model reduced glucocerebrosidase activity in vivo, and tested whether sustained glucocerebrosidase inhibition in mice could induce neuropathological abnormalities including α-synucleinopathy, and neurodegeneration. RESULTS: Our data demonstrate that daily systemic CBE treatment over 28 days caused accumulation of insoluble α-synuclein aggregates in the substantia nigra, and altered levels of proteins involved in the autophagy lysosomal system. These neuropathological changes were paralleled by widespread neuroinflammation, upregulation of complement C1q, abnormalities in synaptic, axonal transport and cytoskeletal proteins, and neurodegeneration. INNOVATION: A reduction in brain GCase activity has been linked to sporadic PD and normal aging, and may contribute to the susceptibility of vulnerable neurons to degeneration. This report demonstrates that systemic reduction of GCase activity using chemical inhibition, leads to neuropathological changes in the brain reminiscent of α-synucleinopathy. CONCLUSIONS: These data reveal a link between reduced glucocerebrosidase and the development of α-synucleinopathy and pathophysiological abnormalities in mice, and support the development of GCase therapeutics to reduce α-synucleinopathy in PD and related disorders.


Assuntos
Complemento C1q/metabolismo , Glucosilceramidase/antagonistas & inibidores , Inositol/análogos & derivados , Microglia/fisiologia , Agregação Patológica de Proteínas/enzimologia , alfa-Sinucleína/metabolismo , Animais , Autofagia , Transporte Axonal , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Ativação do Complemento , Glucosilceramidase/metabolismo , Inositol/farmacologia , Masculino , Camundongos , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/enzimologia , Agregação Patológica de Proteínas/induzido quimicamente , Proteínas/metabolismo , Transmissão Sináptica
14.
Proc Natl Acad Sci U S A ; 111(43): E4568-76, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25316790

RESUMO

Protein framework alterations in heritable Cu, Zn superoxide dismutase (SOD) mutants cause misassembly and aggregation in cells affected by the motor neuron disease ALS. However, the mechanistic relationship between superoxide dismutase 1 (SOD1) mutations and human disease is controversial, with many hypotheses postulated for the propensity of specific SOD mutants to cause ALS. Here, we experimentally identify distinguishing attributes of ALS mutant SOD proteins that correlate with clinical severity by applying solution biophysical techniques to six ALS mutants at human SOD hotspot glycine 93. A small-angle X-ray scattering (SAXS) assay and other structural methods assessed aggregation propensity by defining the size and shape of fibrillar SOD aggregates after mild biochemical perturbations. Inductively coupled plasma MS quantified metal ion binding stoichiometry, and pulsed dipolar ESR spectroscopy evaluated the Cu(2+) binding site and defined cross-dimer copper-copper distance distributions. Importantly, we find that copper deficiency in these mutants promotes aggregation in a manner strikingly consistent with their clinical severities. G93 mutants seem to properly incorporate metal ions under physiological conditions when assisted by the copper chaperone but release copper under destabilizing conditions more readily than the WT enzyme. Altered intradimer flexibility in ALS mutants may cause differential metal retention and promote distinct aggregation trends observed for mutant proteins in vitro and in ALS patients. Combined biophysical and structural results test and link copper retention to the framework destabilization hypothesis as a unifying general mechanism for both SOD aggregation and ALS disease progression, with implications for disease severity and therapeutic intervention strategies.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/patologia , Mutação/genética , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/genética , Superóxido Dismutase/genética , Ácidos/metabolismo , Esclerose Lateral Amiotrófica/genética , Cobre/farmacologia , Cristalografia por Raios X , Ácido Edético/farmacologia , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fenótipo , Substâncias Protetoras/farmacologia , Espalhamento a Baixo Ângulo , Soluções , Superóxido Dismutase/química , Superóxido Dismutase-1
15.
Essays Biochem ; 56: 149-65, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25131593

RESUMO

ALS (amyotrophic lateral sclerosis) is a fatal neurodegenerative syndrome characterized by progressive paralysis and motor neuron death. Although the pathological mechanisms that cause ALS remain unclear, accumulating evidence supports that ALS is a protein misfolding disorder. Mutations in Cu,Zn-SOD1 (copper/zinc superoxide dismutase 1) are a common cause of familial ALS. They have complex effects on different forms of SOD1, but generally destabilize the protein and enhance various modes of misfolding and aggregation. In addition, there is some evidence that destabilized covalently modified wild-type SOD1 may be involved in disease. Among the multitude of misfolded/aggregated species observed for SOD1, multiple species may impair various cellular components at different disease stages. Newly developed antibodies that recognize different structural features of SOD1 represent a powerful tool for further unravelling the roles of different SOD1 structures in disease. Evidence for similar cellular targets of misfolded/aggregated proteins, loss of cellular proteostasis and cell-cell transmission of aggregates point to common pathological mechanisms between ALS and other misfolding diseases, such as Alzheimer's, Parkinson's and prion diseases, as well as serpinopathies. The recent progress in understanding the molecular basis for these devastating diseases provides numerous avenues for developing urgently needed therapeutics.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Estabilidade Enzimática , Humanos , Mutação , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/genética , Dobramento de Proteína , Estrutura Secundária de Proteína , Superóxido Dismutase/genética
16.
Mol Nutr Food Res ; 58(10): 1931-40, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25066301

RESUMO

SCOPE: Dietary polyphenols are suggested to play a role in the prevention of Alzheimer's disease, of which accumulation of aggregated beta amyloid (Aß) is a key histopathological hallmark. We used the transgenic Caenorhabditis elegans strain CL2006, which expresses human Aß1₋42 under control of a muscle-specific promoter and responds to Aß1₋42 aggregation with paralysis, to test effects of the polyphenol quercetin on the phenotype. METHODS AND RESULTS: Quercetin dose-dependently decreased the amount of aggregated proteins in solution and also paralysis in CL2006. The knockdown of key components of unfolded protein response in mitochondria or the endoplasmic reticulum by RNA-interference (RNAi) enhanced paralysis in CL2006 but did not prevent the paralysis reducing activities of quercetin. RNAi for essential members of proteasomal protein degradation or macroautophagy also significantly increased paralysis but prevented quercetin from being effective. Quercetin increased proteasomal activity and, moreover, enhanced the flow of proteins through the macroautophagy pathway as reflected by reduced lysosome staining. CONCLUSION: The proteostasis network, including unfolded protein response, defines the aggregation of Aß1₋42 and the associated paralysis phenotype in a nematode model for Alzheimer's disease. The polyphenol quercetin, by specifically activating macroautophagy and proteasomal degradation pathways, proved able to prevent Aß1₋42 agregation and paralysis.


Assuntos
Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/antagonistas & inibidores , Antioxidantes/uso terapêutico , Caenorhabditis elegans , Modelos Animais de Doenças , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fragmentos de Peptídeos/antagonistas & inibidores , Quercetina/uso terapêutico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Animais Geneticamente Modificados , Antioxidantes/administração & dosagem , Autofagia , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Suplementos Nutricionais , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Músculos/enzimologia , Músculos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Paralisia/induzido quimicamente , Paralisia/prevenção & controle , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Agregação Patológica de Proteínas/enzimologia , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/prevenção & controle , Proteólise , Quercetina/administração & dosagem , Interferência de RNA , Resposta a Proteínas não Dobradas
17.
Neurobiol Aging ; 35(11): 2458-2464, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24973121

RESUMO

The enzyme 5-lipoxygenase (5LO) is upregulated in Alzheimer's disease (AD), and its pharmacologic blockade with zileuton slows down the development of the AD-like phenotype in young AD mice. However, its efficacy after the AD pathology is established is unknown. To this end, starting at 12 months of age triple transgenic mice (3xTg) received zileuton, a selective 5LO inhibitor, or placebo for 3 months, and then the effect of this treatment on behavior, amyloid, and tau pathology assessed. Although mice on placebo showed worsening of their memory, treated mice performed even better than at baseline. Compared with placebo, treated mice had significantly less Aß deposits and tau phosphorylation secondary to reduced γ-secretase and CDK-5 activation, respectively. Our data provide novel insights into the disease-modifying action of pharmacologically inhibiting 5LO as a viable AD therapeutic approach. They represent the successful completion of preclinical studies for the development of this class of drug as clinically applicable therapy for the disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Proteínas Amiloidogênicas/metabolismo , Hidroxiureia/análogos & derivados , Inibidores de Lipoxigenase/uso terapêutico , Memória , Agregados Proteicos , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/enzimologia , Proteínas tau/metabolismo , Doença de Alzheimer/psicologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Araquidonato 5-Lipoxigenase/fisiologia , Quinase 5 Dependente de Ciclina/metabolismo , Hidroxiureia/farmacologia , Hidroxiureia/uso terapêutico , Inibidores de Lipoxigenase/farmacologia , Camundongos Transgênicos , Terapia de Alvo Molecular , Fosforilação , Agregação Patológica de Proteínas/psicologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...