Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Biol ; 20(3): e3001578, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35263320

RESUMEN

Neurodegenerative disorders refer to a group of diseases commonly associated with abnormal protein accumulation and aggregation in the central nervous system. However, the exact role of protein aggregation in the pathophysiology of these disorders remains unclear. This gap in knowledge is due to the lack of experimental models that allow for the spatiotemporal control of protein aggregation, and the investigation of early dynamic events associated with inclusion formation. Here, we report on the development of a light-inducible protein aggregation (LIPA) system that enables spatiotemporal control of α-synuclein (α-syn) aggregation into insoluble deposits called Lewy bodies (LBs), the pathological hallmark of Parkinson disease (PD) and other proteinopathies. We demonstrate that LIPA-α-syn inclusions mimic key biochemical, biophysical, and ultrastructural features of authentic LBs observed in PD-diseased brains. In vivo, LIPA-α-syn aggregates compromise nigrostriatal transmission, induce neurodegeneration and PD-like motor impairments. Collectively, our findings provide a new tool for the generation, visualization, and dissection of the role of α-syn aggregation in PD.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Análisis por Conglomerados , Humanos , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Enfermedad de Parkinson/metabolismo , Agregado de Proteínas , alfa-Sinucleína/metabolismo
2.
ACS Pharmacol Transl Sci ; 4(4): 1463-1467, 2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34423277

RESUMEN

The delivery of hydrophobic therapeutic agents to tumors is a challenge in the treatment of cancers. Here, we review recent advances in coiled-coil protein origami and discuss a proposed programmable protein origami structure, switchable by a protein kinase A/phosphatase switch, as an example of functionalization for designing future protein nanorobots.

3.
ACS Chem Neurosci ; 12(11): 1833-1834, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33979124

RESUMEN

Recent findings showed that preformed fibrils (PFFs) of misfolded proteins, including α-synuclein (α-syn) and amyloid-ß (Aß), activate EGFR in cell cultures and animal models of amyloid propagation. Comparing these supramolecular structures to normal EGFR ligands such as EGF and HB-EGF suggests that these PFFs might trigger the formation of high order clustering of EGFR that stimulates the aggregation of EGFR tyrosine kinase domain (EGFR-TKD) which is known to form fibrils. Subsequently, self-assembled fibril of EGFR-TKDs itself can serve as a seed to induce aggregation of monomeric forms of misfolded proteins in cytoplasm or endosomes. In this model, EGFR serves as an amyloidogenic receptor to facilitate (1) cellular uptake of exogenous PFFs and (2) seeding of endogenous misfolded proteins.


Asunto(s)
Encéfalo , alfa-Sinucleína , Amiloide , Péptidos beta-Amiloides , Proteínas Amiloidogénicas , Animales , Encéfalo/metabolismo , Receptores ErbB , alfa-Sinucleína/metabolismo
4.
Neurotherapeutics ; 18(2): 979-997, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33713002

RESUMEN

Aggregation and deposition of α-synuclein (α-syn) in Lewy bodies within dopamine neurons of substantia nigra (SN) is the pathological hallmark of Parkinson's disease (PD). These toxic α-syn aggregates are believed to propagate from neuron-to-neuron and spread the α-syn pathology throughout the brain beyond dopamine neurons in a prion-like manner. Targeting propagation of such α-syn aggregates is of high interest but requires identifying pathways involving in this process. Evidence from previous Alzheimer's disease reports suggests that EGFR may be involved in the prion-like propagation and seeding of amyloid-ß. We show here that EGFR regulates the uptake of exogenous α-syn-PFFs and the levels of endogenous α-syn in cell cultures and a mouse model of α-syn propagation, respectively. Thus, we tested the therapeutic potentials of AZD3759, a highly selective BBB-penetrating EGFR inhibitor, in a preclinical mouse model of α-syn propagation. AZD3759 decreases activated EGFR levels in the brain and reduces phosphorylated α-synuclein (pSyn) pathology in brain sections, including striatum and SN. As AZD3759 is already in the clinic, this paper's results suggest a possible repositioning of AZD3759 as a disease-modifying approach for PD.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Piperazinas/farmacología , Quinazolinas/farmacología , Sinucleinopatías/prevención & control , alfa-Sinucleína/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Receptores ErbB/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/metabolismo , Quinazolinas/metabolismo , ARN Interferente Pequeño/farmacología , Sinucleinopatías/inducido químicamente , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidad
5.
FEBS J ; 288(2): 452-470, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32365408

RESUMEN

The pathological hallmark of Parkinson's disease (PD) is Lewy bodies that form within the brain from aggregated forms of α-synuclein (α-syn). These toxic α-syn aggregates are transferred from cell to cell by release of fibrils from dying neurons into the extracellular environment, followed by their subsequent uptake by neighboring cells. This process leads to spreading of the pathology throughout the brain in a prion-like manner. Identifying new pathways that hinder the internalization of such α-syn fibrils is of high interest for their downstream potential exploitation as a way to create disease-modifying therapeutics for PD. Here, we show that Thiamet-G, a highly selective pharmacological agent that inhibits the glycoside hydrolase O-GlcNAcase (OGA), blunts the cellular uptake of α-syn fibrils. This effect correlates with increased nucleocytoplasmic levels of O-linked N-acetylglucosamine (O-GlcNAc)-modified proteins, and genetic knockdown of OGA expression closely phenocopies both these effects. These reductions in the uptake of α-syn fibrils caused by inhibition of OGA are both concentration- and time-dependent and are observed in multiple cell lines including mouse primary cortical neurons. Moreover, treatment of cells with the OGT inhibitor, 5SGlcNHex, increases the level of uptake of α-syn PFFs, further supporting O-GlcNAcylation of proteins driving these effects. Notably, this effect is mediated through an unknown mechanism that does not involve well-characterized endocytotic pathways. These data suggest one mechanism by which OGA inhibitors might exert their protective effects in prion-like neuropathologies and support exploration of OGA inhibitors as a potential disease-modifying approach to treat PD.


Asunto(s)
Proteínas Amiloidogénicas/química , Antígenos de Neoplasias/genética , Antiparkinsonianos/farmacología , Inhibidores de Glicósido Hidrolasas/farmacología , Histona Acetiltransferasas/genética , Hialuronoglucosaminidasa/genética , Piranos/farmacología , Tiazoles/farmacología , alfa-Sinucleína/química , Amiloide/antagonistas & inhibidores , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogénicas/antagonistas & inhibidores , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Línea Celular , Regulación de la Expresión Génica , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Hialuronoglucosaminidasa/antagonistas & inhibidores , Hialuronoglucosaminidasa/metabolismo , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Agregado de Proteínas/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
6.
ACS Chem Neurosci ; 11(22): 3704-3706, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33147014

RESUMEN

Postinfection complications of coronavirus disease 2019 (COVID-19) are still unknown, and one of the long-term concerns in infected people are brain pathologies. The question is that severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection may be an environmental factor in accelerating the sporadic neurodegeneration in the infected population. In this regard, induction of protein aggregation in the brain by SARS-CoV-2 intact structure or a peptide derived from spike protein subunits needs to be considered in futures studies. In this paper, we discuss these possibilities using pieces of evidence from other viruses.


Asunto(s)
Betacoronavirus/metabolismo , Encéfalo/metabolismo , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/metabolismo , Neumonía Viral/complicaciones , Neumonía Viral/metabolismo , Agregado de Proteínas/fisiología , Encéfalo/patología , Encéfalo/virología , COVID-19 , Infecciones por Coronavirus/patología , Humanos , Pandemias , Neumonía Viral/patología , SARS-CoV-2 , Factores de Tiempo
7.
Mol Pharmacol ; 98(5): 612-619, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32913137

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are cell surface receptors that are involved in the cellular uptake of pathologic amyloid proteins and viruses, including the novel coronavirus; severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Heparin and heparan sulfate antagonize the binding of these pathogens to HSPGs and stop their cellular internalization, but the anticoagulant effect of these agents has been limiting their use in the treatment of viral infections. Heparin-binding peptides (HBPs) are suitable nonanticoagulant agents that are capable of antagonizing binding of heparin-binding pathogens to HSPGs. Here, we review and discuss the use of HBPs as viral uptake inhibitors and will address their benefits and limitations to treat viral infections. Furthermore, we will discuss a variant of these peptides that is in the clinic and can be considered as a novel therapy in coronavirus disease 2019 (COVID-19) infection. SIGNIFICANCE STATEMENT: The need to discover treatment modalities for COVID-19 is a necessity, and therapeutic interventions such as heparin-binding peptides (HBPs), which are used for other cases, can be beneficial based on their mechanisms of actions. In this paper, we have discussed the application of HBPs as viral uptake inhibitors in COVID-19 and explained possible mechanisms of actions and the therapeutic effects.


Asunto(s)
Antivirales/metabolismo , Betacoronavirus/fisiología , Infecciones por Coronavirus/tratamiento farmacológico , Proteoglicanos de Heparán Sulfato/metabolismo , Péptidos/metabolismo , Neumonía Viral/tratamiento farmacológico , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19 , Proteoglicanos de Heparán Sulfato/química , Humanos , Pandemias , Péptidos/química , Péptidos/farmacología , Péptidos/uso terapéutico , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química
8.
Mol Pharmacol ; 98(1): 13-22, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32350120

RESUMEN

Several reports have been published recently demonstrating a beneficial effect of epidermal growth factor receptor (EGFR) inhibitors in improving pathologic and behavioral conditions in neurodegenerative diseases (NDDs) such as Alzheimer's disease and Amyotrophic Lateral Sclerosis (ALS) as well as the brain and spinal cord injuries (SCI). Despite successful therapeutic effects of EGFR inhibition in these pathologic conditions, there is still no report of proof-of-concept studies in well-characterized animal models using recently developed blood-brain barrier (BBB)-penetrating EGFR inhibitors, which is due to previous conflicting reports concerning the level of EGFR or activated EGFR in normal and pathologic conditions that caused target engagement to be a concern in any future EGFR inhibition therapy. In this review, the level of EGFR expression and activation in the developing central nervous system (CNS) compared with the adult CNS will be explained as well as how neuronal injury or pathologic conditions, especially inflammation and amyloid fibrils, induce reactive astrocytes leading to an increase in the expression and activation of EGFR and, finally, neurodegeneration. Furthermore, in this review, we will discuss two main molecular mechanisms that can be proposed as the neuroprotective effects of EGFR inhibition in these pathologic conditions. We will also review the recent advances in the development of BBB-penetrating EGFR inhibitors in cancer therapy, which may eventually be repositioned for NDDs and SCI therapy in the future. SIGNIFICANCE STATEMENT: Based on the lessons from the applications of EGFR inhibitors in oncology, it is concluded that EGFR inhibitors can be beneficial in treatment of neurodegenerative diseases and spinal cord injuries. They carry their therapeutic potentials through induction of autophagy and attenuation of reactive astrocytes.


Asunto(s)
Lesiones Encefálicas/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Enfermedades Neurodegenerativas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Adulto , Animales , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Sistema Nervioso Central/metabolismo , Niño , Reposicionamiento de Medicamentos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
9.
Mov Disord ; 34(4): 526-535, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30788890

RESUMEN

BACKGROUND: SMPD1 (acid-sphingomyelinase) variants have been associated with Parkinson's disease in recent studies. The objective of this study was to further investigate the role of SMPD1 mutations in PD. METHODS: SMPD1 was sequenced in 3 cohorts (Israel Ashkenazi Jewish cohort, Montreal/Montpellier, and New York), including 1592 PD patients and 975 controls. Additional data were available for 10,709 Ashkenazi Jewish controls. Acid-sphingomyelinase activity was measured by a mass spectrometry-based assay in the New York cohort. α-Synuclein levels were measured in vitro following CRISPR/Cas9-mediated knockout and siRNA knockdown of SMPD1 in HeLa and BE(2)-M17 cells. Lysosomal localization of acid-sphingomyelinase with different mutations was studied, and in silico analysis of their effect on acid-sphingomyelinase structure was performed. RESULTS: SMPD1 mutations were associated with PD in the Ashkenazi Jewish cohort, as 1.4% of PD patients carried the p.L302P or p.fsP330 mutation, compared with 0.37% in 10,709 Ashkenazi Jewish controls (OR, 3.7; 95%CI, 1.6-8.2; P = 0.0025). In the Montreal/Montpellier cohort, the p.A487V variant was nominally associated with PD (1.5% versus 0.14%; P = 0.0065, not significant after correction for multiple comparisons). Among PD patients, reduced acid-sphingomyelinase activity was associated with a 3.5- to 5.8-year earlier onset of PD in the lowest quartile versus the highest quartile of acid-sphingomyelinase activity (P = 0.01-0.001). We further demonstrated that SMPD1 knockout and knockdown resulted in increased α-synuclein levels in HeLa and BE(2)-M17 dopaminergic cells and that the p.L302P and p.fsP330 mutations impair the traffic of acid-sphingomyelinase to the lysosome. CONCLUSIONS: Our results support an association between SMPD1 variants, acid-sphingomyelinase activity, and PD. Furthermore, they suggest that reduced acid-sphingomyelinase activity may lead to α-synuclein accumulation. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Encéfalo/metabolismo , Predisposición Genética a la Enfermedad , Enfermedad de Parkinson/genética , Esfingomielina Fosfodiesterasa/genética , alfa-Sinucleína/metabolismo , Anciano , Encéfalo/patología , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Judíos/genética , Masculino , Persona de Mediana Edad , Mutación , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
10.
J Mol Biol ; 430(20): 3847-3862, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30098337

RESUMEN

Quinolinic acid (QA), a downstream neurometabolite in the kynurenine pathway, the biosynthetic pathway of tryptophan, is associated with neurodegenerative diseases pathology. Mutations in genes encoding kynurenine pathway enzymes, which control the level of QA production, are linked with elevated risk of developing Parkinson's disease. Recent findings have revealed the accumulation and deposition of QA in post-mortem samples, as well as in cellular models of Alzheimer's disease and related disorders. Furthermore, intrastriatal inoculation of mice with QA results in increased levels of phosphorylated α-synuclein and neurodegenerative pathological and behavioral characteristics. However, the cellular and molecular mechanisms underlying the involvement of QA accumulation in protein aggregation and neurodegeneration remain elusive. We recently established that self-assembled ordered structures are formed by various metabolites and hypothesized that these "metabolite amyloids" may seed amyloidogenic proteins. Here we demonstrate the formation of QA amyloid-like fibrillar assemblies and seeding of α-synuclein aggregation by these nanostructures both in vitro and in cell culture. Notably, α-synuclein aggregation kinetics was accelerated by an order of magnitude. Additional amyloid-like properties of QA assemblies were demonstrated using thioflavin T assay, powder X-ray diffraction and cell apoptosis analysis. Moreover, fluorescently labeled QA assemblies were internalized by neuronal cells and co-localized with α-synuclein aggregates. In addition, we observed cell-to-cell propagation of fluorescently labeled QA assemblies in a co-culture of treated and untreated cells. Our findings suggest that excess QA levels, due to mutations in the kynurenine pathway, for example, may lead to the formation of metabolite assemblies that seed α-synuclein aggregation, resulting in neuronal toxicity and induction of Parkinson's disease.


Asunto(s)
Amiloide/química , Ácido Quinolínico/química , alfa-Sinucleína/química , Enfermedad de Alzheimer , Amiloide/metabolismo , Amiloide/ultraestructura , Agregado de Proteínas , Agregación Patológica de Proteínas , Conformación Proteica , Análisis Espectral , Relación Estructura-Actividad , alfa-Sinucleína/metabolismo
11.
Open Biol ; 8(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29367352

RESUMEN

The accumulation of various metabolites appears to be associated with diverse human diseases. However, the aetiological link between metabolic alteration and the observed diseases is still elusive. This includes the correlation between the abnormally high levels of homocysteine and quinolinic acid in Alzheimer's disease, as well as the accumulation of oncometabolites in malignant processes. Here, we suggest and discuss a possible mechanistic insight into metabolite accumulation in conditions such as neurodegenerative diseases and cancer. Our hypothesis is based on the demonstrated ability of metabolites to form amyloid-like structures in inborn error of metabolism disorders and the potential of such metabolite amyloids to promote protein aggregation. This notion can provide a new paradigm for neurodegeneration and cancer, as both conditions were linked to loss of function due to protein aggregation. Similar to the well-established observation of amyloid formation in many degenerative disorders, the formation of amyloids by tumour-suppressor proteins, including p53, was demonstrated in malignant states. Moreover, this new paradigm could fill the gap in understanding the high occurrence of specific types of cancer among genetic error of metabolism patients. This hypothesis offers a fresh view on the aetiology of some of the most abundant human maladies and may redirect the efforts towards new therapeutic developments.


Asunto(s)
Amiloide/metabolismo , Enfermedades Metabólicas/metabolismo , Metaboloma , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Amiloide/química , Animales , Humanos , Enfermedades Metabólicas/epidemiología , Neoplasias/epidemiología , Enfermedades Neurodegenerativas/epidemiología
12.
ACS Chem Neurosci ; 6(2): 347-55, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25514361

RESUMEN

Rasagiline (N-propargyl-1-R-aminoindan) and selegiline (1-deprenyl) are MAO-B inhibitors which are used in the treatment of Parkinson's disease. The binding of rasagiline, selegiline, and their metabolites including 1-aminoindan, 2-aminoindan, and methamphetamine to α-synuclein was investigated by nanopore analysis and isothermal titration calorimetry. Blockade current histograms of α-synuclein alone give a peak at -86 pA which is due to translocation of the protein through the pore. In the presence of rasagiline and R-1-aminoindan, this peak shifts to about -80 pA. In the presence of selegiline and R-methamphetamine, the number of events at -86 pA is reduced and there is a higher proportion of bumping events at about -25 pA which are due to a more compact conformation. Rasagiline can also bind to sites in both the N- and C-terminal regions of α-synuclein. The binding constants of rasagiline and selegiline were estimated by isothermal titration calorimetry to be about 5 × 10(5) and <10(4) M(-1), respectively. A model is presented in which both rasagiline and R-1-aminoindan bind to α-synuclein, forming a loop structure which is less likely to aggregate or form fibrils. In contrast, selegiline binds and forms a more compact structure similar to that formed by methamphetamine.


Asunto(s)
Indanos/farmacología , Fármacos Neuroprotectores/farmacología , alfa-Sinucleína/metabolismo , Calorimetría , Indanos/química , Indanos/metabolismo , Metanfetamina/metabolismo , Inhibidores de la Monoaminooxidasa/química , Inhibidores de la Monoaminooxidasa/farmacología , Nanoporos , Fármacos Neuroprotectores/química , Unión Proteica , Selegilina/química , Selegilina/farmacología
13.
Eur J Med Chem ; 88: 42-54, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25081642

RESUMEN

A major feature of Parkinson's disease is the formation of Lewy bodies in dopaminergic neurons which consist of misfolded α-synuclein. The binding of natural products to α-synuclein was evaluated by nanopore analysis and caffeine, curcumin, and nicotine all caused large conformational changes which may be related to their known neuroprotective effect in Parkinson's disease. The binding of the stereoisomers of nicotine were also studied by ITC, CD and NMR. It is proposed that (-)-nicotine causes the folding of α-synuclein into a loop with interaction between the N- and C-termini. For (+)-nicotine the binding is weaker and mainly involves residues in the N-terminus. Caffeine and nicotine can bind to α-synuclein simultaneously and may provide lead structures for the development of other compounds for the treatment of PD.


Asunto(s)
Productos Biológicos/metabolismo , Descubrimiento de Drogas/métodos , Nanoporos , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Sitios de Unión , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Productos Biológicos/uso terapéutico , Cafeína/química , Cafeína/metabolismo , Calorimetría , Humanos , Conformación Molecular , Nicotina/análogos & derivados , Nicotina/química , Nicotina/metabolismo , Resonancia Magnética Nuclear Biomolecular , Pliegue de Proteína , Relación Estructura-Actividad , alfa-Sinucleína/antagonistas & inhibidores
14.
FEBS J ; 281(12): 2738-53, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24725464

RESUMEN

α-Synuclein (AS) is an intrinsically disordered protein that can misfold and aggregate to form Lewy bodies in dopaminergic neurons, a classic hallmark of Parkinson's disease. The binding of Cu(II) and dopamine to AS was evaluated by nanopore analysis with α-hemolysin. In the absence of Cu(II), wild-type AS (1 µM) readily translocated through the pore with a blockade current of--85 pA, but mostly bumping events were observed in the presence of 25 µM Cu(II). A binding site in the N-terminus was confirmed, because Cu(II) had no effect on the event profile of a peptide consisting of the C-terminal 96-140 residues. In the presence of dopamine (25 µM), the translocation events at--85 pA shifted to--80 pA, which also represents translocation events, because the event time decreases with increasing voltage. Events at--80 pA were also observed for the mutant A30P AS in the presence of dopamine. Event profiles for an N-terminal 1-60-residue peptide and a C-terminal 96-140-residue peptide were both altered in the presence of 25 µM dopamine. In contrast, dopamine had little effect on the CD spectrum of AS, and a single binding site with a Ka of 3.5 × 10(3) m(-1) was estimated by isothermal titration calorimetry. Thus, dopamine can interact with both the N-terminus and the C-terminus. Two-dimensional NMR spectroscopy of AS in the presence of dopamine showed that there were significant changes in the spectra in all regions of the protein. According to these findings, a model is presented in which dopamine induces folding between the N-terminus and C-terminus of AS. Partially folding conformations such as this may represent important intermediates in the misfolding of AS that leads to fibrillization.


Asunto(s)
Cobre/metabolismo , Dopamina/metabolismo , alfa-Sinucleína/metabolismo , Sitios de Unión , Calorimetría , Conformación Proteica , Transporte de Proteínas , alfa-Sinucleína/química
15.
FEBS Lett ; 586(19): 3222-8, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22771474

RESUMEN

α-Synuclein is an intrinsically disordered protein of 140 amino acids which is abundant in dopaminergic neurons. Misfolding and aggregation of α-synuclein leads to the formation of Lewy bodies inside the neurons which is the hallmark of Parkinson's disease and related dementias. Here we show by nanopore analysis that the recreational drug, methamphetamine, binds to the N-terminus of α-synuclein and causes a conformational change which cannot be detected by circular dichroism spectroscopy. The results suggest a mechanism for the psychoactivity of methamphetamine as well as an increased incidence of Parkinson's disease amongst users of the drug.


Asunto(s)
Metanfetamina/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Sitios de Unión , Dicroismo Circular , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Técnicas In Vitro , Cuerpos de Lewy/efectos de los fármacos , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/etiología , Enfermedad por Cuerpos de Lewy/metabolismo , Metanfetamina/toxicidad , Modelos Moleculares , Nanoporos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Unión Proteica , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos
16.
Prion ; 6(2): 116-23, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22421211

RESUMEN

Nanopore analysis is an emerging technique that enables the investigation of the conformation of a single peptide or protein molecule. Briefly, a pore is inserted into a membrane under voltage clamp conditions. When a molecule interacts with the pore there is a change in the current, I, for a time, T. Small unfolded molecules can translocate the pore whereas folded or large molecules tend to simply bump into the pore and then diffuse away. Therefore, the parameters, I and T, are dependent on the conformation of the molecule at the instant at which it encounters the pore. Thus, multiple conformations can be detected simultaneously in a single sample. As well, the analysis can be performed under dilute conditions so that folding or dimerization of a peptide can be followed in real time, which is generally difficult to study for proteins that are prone to aggregate. In this report, we describe our initial analysis of (1) Aß peptides, which are deposited as amyloid plaques in Alzheimer disease, (2) α-synuclein, which is implicated in Parkinson disease and (3) prion proteins whose misfolding is evident in transmissable spongiform encephalopathies. In each case conformational information can be obtained which may help in understanding the early steps in the misfolding pathways.


Asunto(s)
Péptidos beta-Amiloides/química , Biotecnología/métodos , Nanoporos , Priones/química , alfa-Sinucleína/química , Péptidos beta-Amiloides/metabolismo , Animales , Bovinos , Humanos , Técnicas de Placa-Clamp , Priones/metabolismo , Pliegue de Proteína , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...