Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Xenobiotica ; 52(8): 916-927, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36282181

RESUMO

Aggregates of the protein α-synuclein are associated with pathophysiology of Parkinson's disease and are present in Lewy Bodies found in the brains of Parkinson's patients. We previously demonstrated that bifunctional compounds composed of caffeine linked via a six carbon chain to either 1-aminoindan (C8-6-I) or nicotine (C8-6-N) bind α-synuclein and protect yeast cells from α-synuclein mediated toxicity.A critical step in development of positron emission tomography (PET) probes for neurodegenerative diseases is evaluation of their metabolic stability. We determined that C8-6-I, and C8-6-N both undergo phase 1 P450 metabolism in mouse, rat, and human liver microsomes. We utilised this metabolic information to guide the design of fluorinated analogues for use as PET probes and determined that the fluorine in 19F-C8-6-I and 19F-C8-6-N is stable to P450 enzymes.We have developed and validated an analytical HPLC-UV method following FDA and EMA guidelines to measure in vitro phase 1 kinetics of these compounds and determine their Vmax, KM and CLint,u in mouse liver microsomes. We found that C8-6-I and 19F-C8-6-I have a two- to fourfold lower CLint,u than C8-6-N, and 19F-C8-6-N. Our approach shows a simple, specific, and effective system to design and develop compounds as PET probes.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Humanos , Camundongos , Ratos , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Cromatografia Líquida de Alta Pressão , Cinética , Corpos de Lewy/metabolismo , Doença de Parkinson/metabolismo
2.
Transl Neurodegener ; 11(1): 9, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35139916

RESUMO

BACKGROUND: Accumulating α-synuclein (α-syn) aggregates in neurons and glial cells are the staples of many synucleinopathy disorders, such as Parkinson's disease (PD). Since brain adenosine becomes greatly elevated in ageing brains and chronic adenosine A1 receptor (A1R) stimulation leads to neurodegeneration, we determined whether adenosine or A1R receptor ligands mimic the action of known compounds that promote α-syn aggregation (e.g., the amphetamine analogue 2-aminoindan) or inhibit α-syn aggregation (e.g., Rasagiline metabolite 1-aminoindan). In the present study, we determined whether adenosine, A1R receptor agonist N6-Cyclopentyladenosine (CPA) and antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) could directly interact with α-syn to modulate α-syn aggregation and neurodegeneration of dopaminergic neurons in the substantia nigra (SN). METHODS: Nanopore analysis and molecular docking were used to test the binding properties of CPA and DPCPX with α-syn in vitro. Sprague-Dawley rats were administered with 7-day intraperitoneal injections of the A1R ligands and 1- and 2-aminoindan, and levels of α-syn aggregation and neurodegeneration were examined in the SN pars compacta and hippocampal regions using confocal imaging and Western blotting. RESULTS: Using nanopore analysis, we showed that the A1R agonists (CPA and adenosine) interacted with the N-terminus of α-syn, similar to 2-aminoindan, which is expected to promote a "knot" conformation and α-syn misfolding. In contrast, the A1R antagonist DPCPX interacted with the N- and C-termini of α-syn, similar to 1-aminoindan, which is expected to promote a "loop" conformation that prevents α-syn misfolding. Molecular docking studies revealed that adenosine, CPA and 2-aminoindan interacted with the hydrophobic core of α-syn N-terminus, whereas DPCPX and 1-aminoindan showed direct binding to the N- and C-terminal hydrophobic pockets. Confocal imaging and Western blot analyses revealed that chronic treatments with CPA alone or in combination with 2-aminoindan increased α-syn expression/aggregation and neurodegeneration in both SN pars compacta and hippocampus. In contrast, DPCPX and 1-aminoindan attenuated the CPA-induced α-syn expression/aggregation and neurodegeneration in SN and hippocampus. CONCLUSIONS: The results indicate that A1R agonists and drugs promoting a "knot" conformation of α-syn can cause α-synucleinopathy and increase neuronal degeneration, whereas A1R antagonists and drugs promoting a "loop" conformation of α-syn can be harnessed for possible neuroprotective therapies to decrease α-synucleinopathy in PD.


Assuntos
Doença de Parkinson , Sinucleinopatias , Animais , Neurônios Dopaminérgicos/metabolismo , Ligantes , Simulação de Acoplamento Molecular , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , alfa-Sinucleína/metabolismo
3.
Xenobiotica ; 51(8): 885-900, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34187286

RESUMO

A challenge in the development of novel 18F-labelled positron emission tomography (PET) imaging probes is identification of metabolically stable sites to incorporate the 18F radioisotope. Metabolic loss of 18F from PET probes in vivo can lead to misleading biodistribution data as displaced 18F can accumulate in various tissues.In this study we report on in vitro hepatic microsomal metabolism of novel caffeine containing bifunctional compounds (C8-6-I, C8-6-N, C8-6-C8) that can prevent in vitro aggregation of α-synuclein, which is associated with the pathophysiology of Parkinson's disease. The metabolic profile obtained guided us to synthesize stable isotope 19F-labelled analogues in which the fluorine was introduced at the metabolically stable N7 of the caffeine moiety.An in vitro hepatic microsomal metabolism study of the 19F-labelled analogues resulted in similar metabolites to the unlabelled compounds and demonstrated that the fluorine was metabolically stable, suggesting that these analogues are appropriate PET imaging probes. This straightforward in vitro strategy is valuable for avoiding costly stability failures when designing radiolabelled compounds for PET imaging.


Assuntos
Radioisótopos de Flúor , alfa-Sinucleína , Tomografia por Emissão de Pósitrons , Distribuição Tecidual
4.
Electrophoresis ; 38(2): 350-360, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27570136

RESUMO

Seven α-synuclein-drug complexes have been studied by nanopore analysis in which an AC field of 200 mV from 10 MHz to 1 GHz has been superimposed on the standard electrophoretic DC voltage of 100 mV. α-Synuclein has a large dipole moment and in the absence of drug the AC field causes the molecule to oscillate at the entrance to the pore and reduces its ability to translocate through the pore. Thus more bumping events are observed in the current blockade histograms. The binding of drugs to α-synuclein has a large effect on the event profiles depending on the region of α-synuclein to which the drugs bind. Caffeine and (-)-nicotine bind both the N- and C-termini causing the protein to adopt a loop conformation that allows translocation even in the AC field. Metformin, which binds only to the C-terminus also facilitates translocation. For these drugs there is good evidence that the AC field is causing the complex to dissociate as it enters the pore that has not been observed previously. In contrast, complexes with (+)-amphetamine that has an N-terminal binding site and cocaine that binds to the central region of the protein, show only small changes in the event profiles in an AC field.


Assuntos
Eletroforese , Nanoporos , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Cafeína , Eletricidade , Humanos , Metformina , Modelos Químicos , Nicotina , Doença de Parkinson , Preparações Farmacêuticas/análise
5.
ACS Chem Neurosci ; 7(12): 1671-1680, 2016 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-27673434

RESUMO

The misfolding of α-synuclein is a critical event in the death of dopaminergic neurons and the progression of Parkinson's disease. Previously, it was suggested that drugs, which bind to α-synuclein and form a loop structure between the N- and C-termini, tend to be neuroprotective, whereas others, which cause a more compact structure, tend to be neurotoxic. To improve the binding to α-synuclein, eight novel compounds were synthesized from a caffeine scaffold attached to (R,S)-1-aminoindan, (R,S)-nicotine, and metformin, and their binding to α-synuclein determined through nanopore analysis and isothermal titration calorimetry. The ability of the dimers to interact with α-synuclein in a cell system was assayed in a yeast model of PD which expresses an AS-GFP (α-synuclein-Green Fluorescent Protein) construct under the control of a galactose promoter. In 5 mM galactose this yeast strain will not grow and large cytoplasmic foci are observed by fluorescent microscopy. Two of the dimers, C8-6-I and C8-6-N, at a concentration of 0.1 µM allowed the yeast to grow normally in 5 mM galactose and the AS-GFP became localized to the periphery of the cell. Both dimers were superior when compared to the monomeric compounds. The presence of the dimers also caused the disappearance of preformed cytoplasmic foci. Nanopore analysis of C8-6-I and C8-6-N were consistent with simultaneous binding to both the N- and C-terminus of α-synuclein but the binding constants were only 105 M-1.


Assuntos
Antiparkinsonianos/farmacologia , Leveduras/efeitos dos fármacos , Leveduras/crescimento & desenvolvimento , alfa-Sinucleína/metabolismo , Antiparkinsonianos/síntese química , Calorimetria , Dimerização , Avaliação Pré-Clínica de Medicamentos , Galactose/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia de Fluorescência , Doença de Parkinson/prevenção & controle , Ligação Proteica , Leveduras/genética , Leveduras/metabolismo , alfa-Sinucleína/genética
6.
ACS Chem Neurosci ; 6(12): 1930-40, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26378986

RESUMO

The misfolding of α-synuclein is a critical event in the death of dopaminergic neurons and the progression of Parkinson's disease. Drugs that bind to α-synuclein and form a loop structure between the N- and C-terminus tend to be neuroprotective, whereas others that cause a more compact structure tend to be neurotoxic. The binding of several natural products and other drugs that are involved in dopamine metabolism were investigated by nanopore analysis and isothermal titration calorimetry. The antinausea drugs, cinnarizine and metoclopramide, do not bind to α-synuclein, whereas amphetamine and the herbicides, paraquat and rotenone, bind tightly and cause α-synuclein to adopt a more compact conformation. The recreational drug, cocaine, binds to α-synuclein, whereas heroin and methadone do not. Metformin, which is prescribed for diabetes and is neuroprotective, binds well without causing α-synuclein to adopt a more compact conformation. Methylphenidate (ritalin) binds to sites in both the N- and C-terminus and causes α-synuclein to adopt a loop conformation. In contrast, amphetamine only binds to the N-terminus. Except for cinnarizine and metoclopramide, there is a good correlation between the mode of binding to α-synuclein and whether a drug is neuroprotective or neurotoxic.


Assuntos
Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/metabolismo , Animais , Humanos , Conformação Molecular , Doença de Parkinson/metabolismo , Ligação Proteica/efeitos dos fármacos , alfa-Sinucleína/efeitos dos fármacos
7.
Analyst ; 140(14): 4813-9, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25699656

RESUMO

In standard nanopore analysis a constant DC voltage is used to electrophoretically drive small molecules and peptides towards a pore. Superposition of an AC voltage at particular frequencies causes molecules to oscillate as they approach the pore which can alter the event parameters, the blockade current (I) and blockade time (T). Four peptides with similar structures were studied. Alpha-helical peptides A10 (FmocDDA10KK), A14, A18 and retro-inverso A10. It was shown that the ratio of translocations to bumping events could be manipulated by a combination of AC voltages and frequencies. In particular, A10 could be studied without interference from retro-inverso A10. Similarly, a large, intrinsically disordered protein of 140 amino acids, α-synuclein, which translocates the pore readily in a DC field could be prevented from doing so by application of an AC field of 200 mV at 100 MHz.


Assuntos
Eletricidade , Nanoporos , Peptídeos/química
8.
J Biol Chem ; 290(8): 5174-5189, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25561740

RESUMO

Mutations in the human ChlR1 (DDX11) gene are associated with a unique genetic disorder known as Warsaw breakage syndrome characterized by cellular defects in genome maintenance. The DNA triplex helix structures that form by Hoogsteen or reverse Hoogsteen hydrogen bonding are examples of alternate DNA structures that can be a source of genomic instability. In this study, we have examined the ability of human ChlR1 helicase to destabilize DNA triplexes. Biochemical studies demonstrated that ChlR1 efficiently melted both intermolecular and intramolecular DNA triplex substrates in an ATP-dependent manner. Compared with other substrates such as replication fork and G-quadruplex DNA, triplex DNA was a preferred substrate for ChlR1. Also, compared with FANCJ, a helicase of the same family, the triplex resolving activity of ChlR1 is unique. On the other hand, the mutant protein from a Warsaw breakage syndrome patient failed to unwind these triplexes. A previously characterized triplex DNA-specific antibody (Jel 466) bound triplex DNA structures and inhibited ChlR1 unwinding activity. Moreover, cellular assays demonstrated that there were increased triplex DNA content and double-stranded breaks in ChlR1-depleted cells, but not in FANCJ(-/-) cells, when cells were treated with a triplex stabilizing compound benzoquinoquinoxaline, suggesting that ChlR1 melting of triple-helix structures is distinctive and physiologically important to defend genome integrity. On the basis of our results, we conclude that the abundance of ChlR1 known to exist in vivo is likely to be a strong deterrent to the stability of triplexes that can potentially form in the human genome.


Assuntos
RNA Helicases DEAD-box/metabolismo , DNA Helicases/metabolismo , Distúrbios no Reparo do DNA/enzimologia , DNA/metabolismo , Genoma Humano , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , RNA Helicases DEAD-box/genética , DNA/genética , DNA Helicases/genética , Distúrbios no Reparo do DNA/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Células HEK293 , Humanos
9.
ACS Chem Neurosci ; 6(2): 347-55, 2015 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25514361

RESUMO

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.


Assuntos
Indanos/farmacologia , Fármacos Neuroprotetores/farmacologia , alfa-Sinucleína/metabolismo , Calorimetria , Indanos/química , Indanos/metabolismo , Metanfetamina/metabolismo , Inibidores da Monoaminoxidase/química , Inibidores da Monoaminoxidase/farmacologia , Nanoporos , Fármacos Neuroprotetores/química , Ligação Proteica , Selegilina/química , Selegilina/farmacologia
10.
Eur J Med Chem ; 88: 42-54, 2014 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-25081642

RESUMO

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.


Assuntos
Produtos Biológicos/metabolismo , Descoberta de Drogas/métodos , Nanoporos , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/metabolismo , Sítios de Ligação , Produtos Biológicos/química , Produtos Biológicos/isolamento & purificação , Produtos Biológicos/uso terapêutico , Cafeína/química , Cafeína/metabolismo , Calorimetria , Humanos , Conformação Molecular , Nicotina/análogos & derivados , Nicotina/química , Nicotina/metabolismo , Ressonância Magnética Nuclear Biomolecular , Dobramento de Proteína , Relação Estrutura-Atividade , alfa-Sinucleína/antagonistas & inibidores
11.
FEBS J ; 281(12): 2738-53, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24725464

RESUMO

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.


Assuntos
Cobre/metabolismo , Dopamina/metabolismo , alfa-Sinucleína/metabolismo , Sítios de Ligação , Calorimetria , Conformação Proteica , Transporte Proteico , alfa-Sinucleína/química
12.
PLoS One ; 9(2): e88004, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505349

RESUMO

The application of nanopore sensing utilizing the α-hemolysin pore to probe proteins at single-molecule resolution has expanded rapidly. In some studies protein translocation through the α-hemolysin has been reported. However, there is no direct evidence, as yet, that proteins can translocate the α-hemolysin pore. The biggest challenge to obtaining direct evidence is the lack of a highly sensitive assay to detect very low numbers of protein molecules. Furthermore, if an activity based assay is applied then the proteins translocating by unfolding should refold back to an active confirmation for the assay technique to work. To overcome these challenges we selected a model enzyme, ribonuclease A, that readily refolds to an active conformation even after unfolding it with denaturants. In addition we have developed a highly sensitive reverse transcription polymerase chain reaction based activity assay for ribonuclease A. Initially, ribonuclease A, a protein with a positive net charge and dimensions larger than the smallest diameter of the pore, was subjected to nanopore analysis under different experimental conditions. Surprisingly, although the protein was added to the cis chamber (grounded) and a positive potential was applied, the interaction of ribonuclease A with α-hemolysin pore induced small and large blockade events in the presence and the absence of a reducing and/or denaturing agent. Upon measuring the zeta potential, it was found that the protein undergoes a charge reversal under the experimental conditions used for nanopore sensing. From the investigation of the effect of voltage on the interaction of ribonuclease A with the α-hemolysin pore, it was impossible to conclude if the events observed were translocations. However, upon testing for ribonuclease A activity on the trans chamber it was found that ribonuclease A does not translocate the α-hemolysin pore.


Assuntos
Proteínas Hemolisinas/metabolismo , Transporte Proteico/fisiologia , Ribonuclease Pancreático/metabolismo , Nanoporos , Nanoestruturas , Nanotecnologia/métodos
13.
Protein Pept Lett ; 21(3): 247-55, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24370255

RESUMO

In this minireview, the nanopore analysis of peptides and proteins in the presence of divalent metal ions will be surveyed. In all cases the binding of the metal ions causes the peptide or protein to adopt a more compact conformation which can no longer enter the α-hemolysin pore. In the absence of Zn(II) the 30-amino acid Zn-finger peptide can readily translocate the pore; but upon addition of Zn(II) the peptide folds and only bumping events are observed. Similarly, the octapeptide repeat from the N-terminus of the prion protein binds Cu(II), which prevents it from translocating. The full-length prion protein also undergoes conformational changes upon binding Cu(II), which results in an increase in the proportion of bumping events. Myelin basic protein of 170 residues is intrinsically disordered and, perhaps surprisingly, for a basic protein of this size, can translocate against the electric field based on the observation that the event time increases with increasing voltage. It, too, folds into a more compact conformation upon binding Cu(II) and Zn(II), which prevents translocation. Finally even proteins such as maltose binding protein which does not contain a formal binding site for metal ions undergoes conformational changes in the presence of the metal chelator, EDTA. Thus, contamination of proteins with trace metal ions should be considered when studying proteins and peptides by nanopore analysis.


Assuntos
Cobre/metabolismo , Nanoporos , Peptídeos/química , Dobramento de Proteína , Proteínas/química , Zinco/metabolismo , Animais , Humanos , Modelos Moleculares , Nanoporos/ultraestrutura , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Transporte Proteico , Proteínas/metabolismo
14.
Prion ; 7(4): 301-11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23787697

RESUMO

Transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative diseases that are based on the misfolding of a cellular prion protein (PrP(C)) into an infectious, pathological conformation (PrP(Sc)). There is proof-of-principle evidence that a prion vaccine is possible but this is tempered with concerns of the potential dangers associated with induction of immune responses to a widely-expressed self-protein. By targeting epitopes that are specifically exposed upon protein misfolding, our group developed a vaccine that induces PrP(Sc)-specific antibody responses. Here we consider the ability of this polyclonal antibody (SN6b) to bind to a mutant of PrP(C) associated with spontaneous prion disease. Polyclonal antibodies were selected to mimic the vaccination outcome and also explore all possible protein conformations of the recombinant bovine prion protein with mutation T194A [bPrP(T194A)]. This mutant is a homolog of the human T183A mutation of PrP(C) that is associated with early onset of familial dementia. With nanopore analysis, under non-denaturing conditions, we observed binding of the SN6b antibody to bPrP(T194A). This interaction was confirmed through ELISAs as well as immunoprecipitation of the recombinant and cellularly expressed forms of bPrP(T194A). This interaction did not promote formation of a protease resistant conformation of PrP in vitro. Collectively, these findings support the disease-specific approach for immunotherapy of prion diseases but also suggest that the concept of conformation-specific immunotherapy may be complicated in individuals who are genetically predisposed to PrP(C) misfolding.


Assuntos
Anticorpos/química , Mutação de Sentido Incorreto , Proteínas PrPC/química , Proteínas PrPSc/química , Substituição de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/imunologia , Especificidade de Anticorpos , Bovinos , Demência/genética , Demência/imunologia , Demência/terapia , Células HEK293 , Humanos , Imunoterapia , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPSc/genética , Proteínas PrPSc/imunologia , Dobramento de Proteína , Vacinas/química , Vacinas/genética , Vacinas/imunologia
15.
Prion ; 7(6): 511-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24401607

RESUMO

Species, as well as individuals within species, have unique susceptibilities to prion infection that are likely based on sequence differences in cellular prion protein (PrP(C)). Species barriers to transmission also reflect PrP(C) sequence differences. Defining the structure-activity relationship of PrP(C)/PrP(Sc) with respect to infectivity/susceptibility will benefit disease understanding and assessment of transmission risks. Here, nanopore analysis is employed to investigate genotypes of sheep PrP(C) corresponding to differential susceptibilities to scrapie infection. Under non-denaturing conditions scrapie resistant (ARR) and susceptible (VRQ) genotypes display similar, type I (bumping) predominant event profiles, suggesting a conserved folding pattern. Under increasingly denaturing conditions both proteins shift to type II (intercalation/translocation) events but with different sensitivities to unfolding. Specifically, when pre-incubated in 2M Gdn-HCl, the VRQ variant had more of type II events as compared with the ARR protein, suggesting a more flexible unfolding pattern. Addition of PrP(Sc)-specific polyclonal antibody (YML) to the ARR variant, pre-incubated in 2M Gdn-HCl, reduced the number of type II events with no clear intercalation/translocation peak, whereas for VRQ, type II events above blockades of 90 pA bound YML. A second PrP(Sc)-specific antibody (SN6b) to a different cryptic epitope reduced type II events for VRQ but not the ARR variant. Collectively, the event patterns associated with sequential denaturation, as well as interactions with PrP(Sc)-specific antibodies, support unique patterns and/or propensities of misfolding between the genotypes. Overall, nanopore analysis identifies intermediate conformations that occur during the unfolding pathways of ARR and VRQ genotypes and may help to understand the correlation of structural properties that induce protein misfolding.


Assuntos
Nanoporos , Proteínas PrPC/genética , Scrapie/genética , Ovinos/genética , Animais , Genótipo , Nanoporos/ultraestrutura , Proteínas PrPC/química , Conformação Proteica , Desdobramento de Proteína
16.
FEBS Lett ; 586(19): 3222-8, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22771474

RESUMO

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.


Assuntos
Metanfetamina/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Sítios de Ligação , Dicroísmo Circular , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Humanos , Técnicas In Vitro , Corpos de Lewy/efeitos dos fármacos , Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/etiologia , Doença por Corpos de Lewy/metabolismo , Metanfetamina/toxicidade , Modelos Moleculares , Nanoporos , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos
17.
Metallomics ; 4(6): 539-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22544081

RESUMO

Nanopore analysis is a promising technique for studying the conformation of proteins and protein/protein interactions. Two proteins (bacterial thioredoxin and maltose binding protein) were subjected to nanopore analysis with α-hemolysin. Two types of events were observed; bumping events with a blockade current less than -40 pA and intercalation events with blockade currents between -40 pA and -100 pA. In potassium phosphate buffer, pH 7.8, both proteins gave intercalation events but the frequency of these events was significantly reduced in TRIS or HEPES buffers especially in the presence of 0.01 mM divalent metal ions. The frequency of events was restored by the addition of EDTA. For maltose binding protein, the frequency of intercalation events was also decreased in the presence of maltose but not lactose to which it does not bind. It is proposed that the events with large blockade currents represent transient intercalation of a loop or end of the protein into the pore and that divalent metal ions inhibit this process. The results demonstrate that the choice of buffer and the effects of metal ion contamination are important considerations in nanopore analysis.


Assuntos
Ácido Edético/química , Nanoporos , Mapeamento de Interação de Proteínas/métodos , Proteínas/análise , Soluções Tampão , Proteínas Hemolisinas/análise , Proteínas Hemolisinas/química , Proteínas Ligantes de Maltose/análise , Proteínas Ligantes de Maltose/química , Modelos Moleculares , Proteínas/química , Tiorredoxinas/análise , Tiorredoxinas/química
18.
Prion ; 6(2): 116-23, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22421211

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/química , Biotecnologia/métodos , Nanoporos , Príons/química , alfa-Sinucleína/química , Peptídeos beta-Amiloides/metabolismo , Animais , Bovinos , Humanos , Técnicas de Patch-Clamp , Príons/metabolismo , Dobramento de Proteína , alfa-Sinucleína/metabolismo
19.
Chem Commun (Camb) ; 48(13): 1928-30, 2012 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-22228143

RESUMO

The interaction of two peptides with the α-hemolysin pore was studied in the presence of a MHz AC field. For an α-helical peptide the proportion of bumping events increased with increasing AC field whereas for a linear peptide with no dipole moment only small changes in the event profiles were observed.


Assuntos
Eletricidade , Proteínas Hemolisinas/metabolismo , Nanoporos , Peptídeos/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Peptídeos/química , Transporte Proteico
20.
J Pept Sci ; 17(11): 726-34, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21766390

RESUMO

Nanopore analysis is an emerging technique of structural biology which employs nanopores, such as the α-hemolysin pore, as a biosensor. A voltage applied across a membrane containing a nanopore generates a current, which is partially blocked when a molecule interacts with the pore. The magnitude (I) and the duration (T) of the current blockade provide an event signature for that molecule. Two peptides, CY12(+)T1 and CY12(-)T1 with net charges + 2 and - 2, respectively, were analysed using different applied voltages and all four possible orientations of the electrodes and pore. The four orientations were vestibule downstream (VD), vestibule upstream (VU), stem downstream (SD) and stem upstream (SU) where vestibule and stem refer to the side of the pore on which the peptide was placed and downstream and upstream refer to the application of a positive or negative electrophoretic force, respectively. For CY12(+)T1, the effect of voltage on the event duration was consistent with translocation in the VD and SD configurations, but only intercalation events were observed in the VU and SU configurations. For CY12(-)T1, translocations were only observed in the VD and VU configurations. The results are interpreted in terms of two energy barriers on either side of the lumen of the pore. The difference in height of the barriers determines the preferred direction of exit. Electroosmotic flow and current rectification due to the pore as well as the dipole moment and charge of the peptide also play significant roles. Thus, factors other than simple electrophoresis are important for determining the interaction of small peptides with the pore.


Assuntos
Eletricidade , Proteínas Hemolisinas/química , Peptídeos/química , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...