Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Toxicol Appl Pharmacol ; 421: 115534, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33852878

RESUMO

Monomethyl auristatin E (MMAE) is a potent anti-cancer microtubule-targeting agent (MTA) used as a payload in three approved MMAE-containing antibody drug conjugates (ADCs) and multiple ADCs in clinical development to treat different types of cancers. Unfortunately, MMAE-ADCs can induce peripheral neuropathy, a frequent adverse event leading to treatment dose reduction or discontinuation and subsequent clinical termination of many MMAE-ADCs. MMAE-ADC-induced peripheral neuropathy is attributed to non-specific uptake of the ADC in peripheral nerves and release of MMAE, disrupting microtubules (MTs) and causing neurodegeneration. However, molecular mechanisms underlying MMAE and MMAE-ADC effects on MTs remain unclear. Here, we characterized MMAE-tubulin/MT interactions in reconstituted in vitro soluble tubulin or MT systems and evaluated MMAE and vcMMAE-ADCs in cultured human MCF7 cells. MMAE bound to soluble tubulin heterodimers with a maximum stoichiometry of ~1:1, bound abundantly along the length of pre-assembled MTs and with high affinity at MT ends, introduced structural defects, suppressed MT dynamics, and reduced the kinetics and extent of MT assembly while promoting tubulin ring formation. In cells, MMAE and MMAE-ADC (via nonspecific uptake) suppressed proliferation, mitosis and MT dynamics, and disrupted the MT network. Comparing MMAE action to other MTAs supports the hypothesis that peripheral neuropathy severity is determined by the precise mechanism(s) of each individual drug-MT interaction (location of binding, affinity, effects on morphology and dynamics). This work demonstrates that MMAE binds extensively to tubulin and MTs and causes severe MT dysregulation, providing convincing evidence that MMAE-mediated inhibition of MT-dependent axonal transport leads to severe peripheral neuropathy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Microtúbulos/efeitos dos fármacos , Oligopeptídeos/toxicidade , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Sistema Nervoso Periférico/efeitos dos fármacos , Moduladores de Tubulina/toxicidade , Tubulina (Proteína)/metabolismo , Transporte Axonal/efeitos dos fármacos , Sítios de Ligação , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Microtúbulos/patologia , Mitose/efeitos dos fármacos , Oligopeptídeos/metabolismo , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/patologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Ligação Proteica , Medição de Risco , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Fuso Acromático/patologia , Moduladores de Tubulina/metabolismo
2.
J Biol Chem ; 294(33): 12265-12280, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31266806

RESUMO

The microtubule (MT)-associated protein tau regulates the critical growing and shortening behaviors of MTs, and its normal activity is essential for neuronal development and maintenance. Accordingly, aberrant tau action is tightly associated with Alzheimer's disease and is genetically linked to several additional neurodegenerative diseases known as tauopathies. Although tau is known to promote net MT growth and stability, the precise mechanistic details governing its regulation of MT dynamics remain unclear. Here, we have used the slowly-hydrolyzable GTP analog, guanylyl-(α,ß)-methylene-diphosphonate (GMPCPP), to examine the structural effects of tau at MT ends that may otherwise be too transient to observe. The addition of both four-repeat (4R) and three-repeat (3R) tau isoforms to pre-formed GMPCPP MTs resulted in the formation of extended, multiprotofilament-wide projections at MT ends. Furthermore, at temperatures too low for assembly of bona fide MTs, both tau isoforms promoted the formation of long spiral ribbons from GMPCPP tubulin heterodimers. In addition, GMPCPP MTs undergoing cold-induced disassembly in the presence of 4R tau (and to a much lesser extent 3R tau) also formed spirals. Finally, three pathological tau mutations known to cause neurodegeneration and dementia were differentially compromised in their abilities to stabilize MT disassembly intermediates. Taken together, we propose that tau promotes the formation/stabilization of intermediate states in MT assembly and disassembly by promoting both longitudinal and lateral tubulin-tubulin contacts. We hypothesize that these activities represent fundamental aspects of tau action that normally occur at the GTP-rich ends of GTP/GDP MTs and that may be compromised in neurodegeneration-causing tau variants.


Assuntos
Microtúbulos/química , Tubulina (Proteína)/química , Proteínas tau/química , Demência/metabolismo , Humanos , Microtúbulos/genética , Microtúbulos/metabolismo , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(9): 2758-63, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25691742

RESUMO

Intrinsically disordered proteins (IDPs) are a unique class of proteins that have no stable native structure, a feature that allows them to adopt a wide variety of extended and compact conformations that facilitate a large number of vital physiological functions. One of the most well-known IDPs is the microtubule-associated tau protein, which regulates microtubule growth in the nervous system. However, dysfunctions in tau can lead to tau oligomerization, fibril formation, and neurodegenerative disease, including Alzheimer's disease. Using a combination of simulations and experiments, we explore the role of osmolytes in regulating the conformation and aggregation propensities of the R2/wt peptide, a fragment of tau containing the aggregating paired helical filament (PHF6*). We show that the osmolytes urea and trimethylamine N-oxide (TMAO) shift the population of IDP monomer structures, but that no new conformational ensembles emerge. Although urea halts aggregation, TMAO promotes the formation of compact oligomers (including helical oligomers) through a newly proposed mechanism of redistribution of water around the perimeter of the peptide. We put forth a "superposition of ensembles" hypothesis to rationalize the mechanism by which IDP structure and aggregation is regulated in the cell.


Assuntos
Simulação de Dinâmica Molecular , Peptídeos/química , Agregação Patológica de Proteínas , Proteínas tau/química , Humanos , Metilaminas/química , Estrutura Secundária de Proteína , Ureia/química
4.
Int J Mass Spectrom ; 420: 24-34, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-29056865

RESUMO

The early oligomerization of amyloid ß-protein (Aß) is a crucial step in the etiology of Alzheimer's disease (AD), in which soluble and highly neurotoxic oligomers are produced and accumulated inside neurons. In search of therapeutic solutions for AD treatment and prevention, potent inhibitors that remodel Aß assembly and prevent neurotoxic oligomer formation offer a promising approach. In particular, several polyphenolic compounds have shown anti-aggregation properties and good efficacy on inhibiting oligomeric amyloid formation. 1,2,3,4,6-penta-O-galloyl-ß-D-glucopyranose is a large polyphenol that has been shown to be effective at inhibiting aggregation of full-length Aß1-40 and Aß1-42, but has the opposite effect on the C-terminal fragment Aß25-35. Here, we use a combination of ion mobility coupled to mass spectrometry (IMS-MS), transmission electron microscopy (TEM) and molecular dynamics (MD) simulations to elucidate the inhibitory effect of PGG on aggregation of full-length Aß1-40 and Aß1-42. We show that PGG interacts strongly with these two peptides, especially in their N-terminal metal binding regions, and suppresses the formation of Aß1-40 tetramer and Aß1-42 dodecamer. By exploring multiple facets of polyphenol-amyloid interactions, we provide a molecular basis for the opposing effects of PGG on full-length Aß and its C-terminal fragments.

5.
J Am Chem Soc ; 138(2): 549-57, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26700445

RESUMO

In order to evaluate potential therapeutic targets for treatment of amyloidoses such as Alzheimer's disease (AD), it is essential to determine the structures of toxic amyloid oligomers. However, for the amyloid ß-protein peptide (Aß), thought to be the seminal neuropathogenetic agent in AD, its fast aggregation kinetics and the rapid equilibrium dynamics among oligomers of different size pose significant experimental challenges. Here we use ion-mobility mass spectrometry, in combination with electron microscopy, atomic force microscopy, and computational modeling, to test the hypothesis that Aß peptides can form oligomeric structures resembling cylindrins and ß-barrels. These structures are hypothesized to cause neuronal injury and death through perturbation of plasma membrane integrity. We show that hexamers of C-terminal Aß fragments, including Aß(24-34), Aß(25-35) and Aß(26-36), have collision cross sections similar to those of cylindrins. We also show that linking two identical fragments head-to-tail using diglycine increases the proportion of cylindrin-sized oligomers. In addition, we find that larger oligomers of these fragments may adopt ß-barrel structures and that ß-barrels can be formed by folding an out-of-register ß-sheet, a common type of structure found in amyloid proteins.


Assuntos
Peptídeos beta-Amiloides/química , Proteínas Sanguíneas/química , Sequência de Aminoácidos , Microscopia Eletrônica de Transmissão , Conformação Proteica , Espectrometria de Massas por Ionização por Electrospray
6.
J Neurochem ; 137(6): 939-54, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26953146

RESUMO

Despite extensive structure-function analyses, the molecular mechanisms of normal and pathological tau action remain poorly understood. How does the C-terminal microtubule-binding region regulate microtubule dynamics and bundling? In what biophysical form does tau transfer trans-synaptically from one neuron to another, promoting neurodegeneration and dementia? Previous biochemical/biophysical work led to the hypothesis that tau can dimerize via electrostatic interactions between two N-terminal 'projection domains' aligned in an anti-parallel fashion, generating a multivalent complex capable of interacting with multiple tubulin subunits. We sought to test this dimerization model directly. Native gel analyses of full-length tau and deletion constructs demonstrate that the N-terminal region leads to multiple bands, consistent with oligomerization. Ferguson analyses of native gels indicate that an N-terminal fragment (tau(45-230) ) assembles into heptamers/octamers. Ferguson analyses of denaturing gels demonstrates that tau(45-230) can dimerize even in sodium dodecyl sulfate. Atomic force microscopy reveals multiple levels of oligomerization by both full-length tau and tau(45-230) . Finally, ion mobility-mass spectrometric analyses of tau(106-144) , a small peptide containing the core of the hypothesized dimerization region, also demonstrate oligomerization. Thus, multiple independent strategies demonstrate that the N-terminal region of tau can mediate higher order oligomerization, which may have important implications for both normal and pathological tau action. The microtubule-associated protein tau is essential for neuronal development and maintenance, but is also central to Alzheimer's and related dementias. Unfortunately, the molecular mechanisms underlying normal and pathological tau action remain poorly understood. Here, we demonstrate that tau can homo-oligomerize, providing novel mechanistic models for normal tau action (promoting microtubule growth and bundling, suppressing microtubule shortening) and pathological tau action (poisoning of oligomeric complexes).


Assuntos
Microtúbulos/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Dimerização , Humanos , Espectrometria de Massas , Microscopia de Força Atômica , Modelos Biológicos , Peptídeos/química , Ligação Proteica , Proteínas tau/genética
7.
Anal Chem ; 88(1): 868-76, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26632663

RESUMO

Ion-mobility mass spectrometry is utilized to examine the metacluster formation of serine, asparagine, isoleucine, and tryptophan. These amino acids are representative of different classes of noncharged amino acids. We show that they can form relatively large metaclusters in solution that are difficult or impossible to observe by traditional solution techniques. We further demonstrate, as an example, that the formation of Ser metaclusters is not an ESI artifact because large metaclusters can be detected in negative polarity and low concentration with similar cross sections to those measured in positive polarity and higher concentration. The growth trends of tryptophan and isoleucine metaclusters, along with serine, asparagine, and the previously studied phenylalanine, are balanced among various intrinsic properties of individual amino acids (e.g., hydrophobicity, size, and shape). The metacluster cross sections of hydrophilic residues (Ser, Asn, Trp) tend to stay on or fall below the isotropic model trend lines whereas those of hydrophobic amino acids (Ile, Phe) deviate positively from the isotropic trend lines. The growth trends correlate well to the predicted aggregation propensity of individual amino acids. From the metacluster data, we introduce a novel approach to score and predict aggregation propensity of peptides, which can offer a significant improvement over the existing methods in terms of accuracy. Using a set of hexapeptides, we show that the strong negative deviations of Ser metaclusters from the isotropic model leads a prediction of microcrystalline formation for the SFSFSF peptide, whereas the strong positive deviation of Ile leads to prediction or fibril formation for the NININI peptide. Both predictions are confirmed experimentally using ion mobility and TEM measurements. The peptide SISISI is predicted to only weakly aggregate, a prediction confirmed by TEM.


Assuntos
Aminoácidos/análise , Peptídeos/síntese química , Espectrometria de Massas , Peptídeos/química , Agregados Proteicos , Conformação Proteica
8.
Phys Chem Chem Phys ; 15(23): 8916-28, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23515417

RESUMO

The microtubule associated protein tau is essential for the development and maintenance of the nervous system. Tau dysfunction is associated with a class of diseases called tauopathies, in which tau is found in an aggregated form. This paper focuses on a small aggregating fragment of tau, (273)GKVQIINKKLDL(284), encompassing the (PHF6*) region that plays a central role in tau aggregation. Using a combination of simulations and experiments, we probe the self-assembly of this peptide, with an emphasis on characterizing the early steps of aggregation. Ion-mobility mass spectrometry experiments provide a size distribution of early oligomers, TEM studies provide a time course of aggregation, and enhanced sampling molecular dynamics simulations provide atomistically detailed structural information about this intrinsically disordered peptide. Our studies indicate that a point mutation, as well the addition of heparin, lead to a shift in the conformations populated by the earliest oligomers, affecting the kinetics of subsequent fibril formation as well as the morphology of the resulting aggregates. In particular, a mutant associated with a K280 deletion (a mutation that causes a heritable form of neurodegeneration/dementia in the context of full length tau) is seen to aggregate more readily than its wild-type counterpart. Simulations and experiment reveal that the ΔK280 mutant peptide adopts extended conformations to a greater extent than the wild-type peptide, facilitating aggregation through the pre-structuring of the peptide into a fibril-competent structure.


Assuntos
Deleção de Sequência , Tauopatias/genética , Tauopatias/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Sequência de Aminoácidos , Heparina/metabolismo , Humanos , Simulação de Dinâmica Molecular , Mutação Puntual , Conformação Proteica , Multimerização Proteica , Proteínas tau/química , Proteínas tau/ultraestrutura
9.
J Neurosci ; 31(27): 9858-68, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21734277

RESUMO

Aggregated filamentous forms of hyperphosphorylated tau (a microtubule-associated protein) represent pathological hallmarks of Alzheimer's disease (AD) and other tauopathies. While axonal transport dysfunction is thought to represent a primary pathogenic factor in AD and other neurodegenerative diseases, the direct molecular link between pathogenic forms of tau and deficits in axonal transport remain unclear. Recently, we demonstrated that filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Here, we demonstrate that amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway in axoplasms isolated from squid giant axons. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Importantly, immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity, an early marker of pathological tau. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT. Results from these studies reveal a novel role for tau in modulating axonal phosphotransferases and provide a molecular basis for a toxic gain-of-function associated with pathogenic forms of tau.


Assuntos
Transporte Axonal/genética , Axônios/patologia , Encéfalo/patologia , Cinesinas/metabolismo , Fosfotransferases/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Análise de Variância , Animais , Transporte Axonal/efeitos dos fármacos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Decapodiformes , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Técnicas In Vitro , Cinesinas/genética , Modelos Biológicos , Mutagênese/genética , Fragmentos de Peptídeos/metabolismo , Isótopos de Fósforo/farmacocinética , Fosfotransferases/genética , Proteínas Proto-Oncogênicas c-jun/farmacocinética , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais/genética , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
10.
Biochemistry ; 48(51): 12290-7, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-19919107

RESUMO

Alzheimer's disease and other tauopathies are characterized by the intracellular accumulation of insoluble filaments of the microtubule-associated protein tau. The six canonical tau isoforms in the adult brain consist of an N-terminal "projection" domain followed by a proline-rich region, a microtubule-binding repeat region, and a C-terminal tail. However, alternative splicing in exon 6 produces an additional set of tau isoforms, termed 6D and 6P, which contain only the N-terminus and part of the proline-rich region. We have previously shown that constructs representing N-terminal fragments of tau, which resemble the naturally occurring 6P and 6D isoforms, inhibit polymerization of the full-length protein in an in vitro filament formation assay and traced the inhibitory activity to amino acids 18-42. Here we report that 6P and 6D tau isoforms inhibit polymerization of full-length tau (hTau40) in a similar manner, likely by stabilizing full-length tau in a soluble conformation. The absence of exons 2 and 3 decreased the effectiveness of the 6D isoforms but not the 6P variants or the N-terminal tau fragments from our previous study, indicating that the 18-42 region is not the sole determinant of inhibitory ability. Finally, this paper demonstrates that inhibition is blocked by pseudophosphorylation of tyrosines 18 and 29, providing a potential link between tyrosine phosphorylation and disease progression. Taken together, these results indicate that the 6P/6D isoforms are potential endogenous inhibitors of tau filament formation and suggest a mechanism by which this ability may be disrupted in disease.


Assuntos
Dobramento de Proteína , Proteínas tau/química , Processamento Alternativo , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
11.
J Neurosci Res ; 87(2): 440-51, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18798283

RESUMO

The neuropathology of Alzheimer's disease (AD) and other tauopathies is characterized by filamentous deposits of the microtubule-associated protein tau, but the relationship between tau polymerization and neurotoxicity is unknown. Here, we examined effects of filamentous tau on fast axonal transport (FAT) using isolated squid axoplasm. Monomeric and filamentous forms of recombinant human tau were perfused in axoplasm, and their effects on kinesin- and dynein-dependent FAT rates were evaluated by video microscopy. Although perfusion of monomeric tau at physiological concentrations showed no effect, tau filaments at the same concentrations selectively inhibited anterograde (kinesin-dependent) FAT, triggering the release of conventional kinesin from axoplasmic vesicles. Pharmacological experiments indicated that the effect of tau filaments on FAT is mediated by protein phosphatase 1 (PP1) and glycogen synthase kinase-3 (GSK-3) activities. Moreover, deletion analysis suggested that these effects depend on a conserved 18-amino-acid sequence at the amino terminus of tau. Interestingly, monomeric tau isoforms lacking the C-terminal half of the molecule (including the microtubule binding region) recapitulated the effects of full-length filamentous tau. Our results suggest that pathological tau aggregation contributes to neurodegeneration by altering a regulatory pathway for FAT.


Assuntos
Transporte Axonal/fisiologia , Cinesinas/metabolismo , Proteínas tau/metabolismo , Proteínas tau/toxicidade , Animais , Citoesqueleto/metabolismo , Decapodiformes , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Immunoblotting , Microtúbulos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas tau/química
12.
Methods Cell Biol ; 141: 217-228, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28882303

RESUMO

The proper organization and function of the mammalian nervous system relies on neuronal processes or "neurites," extended morphological projections that include axons and dendrites. Tau is a structural microtubule-associated protein that is widely expressed in the nervous system that mediates the establishment of cell polarity, neurite outgrowth, and axonal transport. A useful model for studying the establishment and maintenance of these neuronal structures are rat neuronal PC12 cells, which can be induced to express tau and project neurites by treating the cells with nerve growth factor. Here, we present a simple method for continuously measuring the rate of neurite outgrowth and retraction over time by neurite length and neurite area analyses. This method uses freely available ImageJ software and widely available phase-contrast imaging.


Assuntos
Microscopia de Contraste de Fase/métodos , Neuritos/ultraestrutura , Crescimento Neuronal , Proteínas tau/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Diferenciação Celular , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Neuritos/metabolismo , Células PC12 , Ratos
13.
Cancer Chemother Pharmacol ; 80(1): 151-164, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28567478

RESUMO

Cabazitaxel is a novel taxane approved for treatment of metastatic hormone-refractory prostate cancer in patients pretreated with docetaxel. Cabazitaxel, docetaxel, and paclitaxel bind specifically to tubulin in microtubules, disrupting functions essential to tumor growth. High levels of ßIII-tubulin isotype expression are associated with tumor aggressivity and drug resistance. To understand cabazitaxel's increased efficacy, we examined binding of radio-labeled cabazitaxel and docetaxel to microtubules and the drugs' suppression of microtubule dynamic instability in vitro in microtubules assembled from purified bovine brain tubulin containing or devoid of ßIII-tubulin. We found that cabazitaxel suppresses microtubule dynamic instability significantly more potently in the presence of ßIII-tubulin than in its absence. In contrast, docetaxel showed no ßIII-tubulin-enhanced microtubule stabilization. We also asked if the selective potency of cabazitaxel on ßIII-tubulin-containing purified microtubules in vitro extends to cabazitaxel's effects in human tumor cells. Using MCF7 human breast adenocarcinoma cells, we found that cabazitaxel also suppressed microtubule shortening rates, shortening lengths, and dynamicity significantly more strongly in cells with normal levels of ßIII-tubulin than after 50% reduction of ßIII-tubulin expression by siRNA knockdown. Cabazitaxel also more strongly induced mitotic arrest in MCF7 cells with normal ßIII-tubulin levels than after ßIII-tubulin reduction. In contrast, docetaxel had little or no ßIII-tubulin-dependent selective effect on microtubule dynamics or mitotic arrest. The selective potency of cabazitaxel on purified ßIII-tubulin-containing microtubules and in cells expressing ßIII-tubulin suggests that cabazitaxel may be unusual among microtubule-targeted drugs in its superior anti-tumor efficacy in tumors overexpressing ßIII-tubulin.


Assuntos
Microtúbulos/metabolismo , Taxoides/farmacologia , Moduladores de Tubulina/farmacologia , Tubulina (Proteína)/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Encéfalo/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Bovinos , Docetaxel , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , RNA Interferente Pequeno/administração & dosagem , Tubulina (Proteína)/genética
14.
Methods Cell Biol ; 141: 3-26, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28882309

RESUMO

In this chapter, we describe methods for the purification of both untagged and polyhistidine-tagged tau protein. These protocols utilize a bacterial expression system to produce the tau isoform of interest, followed by heat treatment and column chromatography to separate tau from impurities. These techniques yield a biochemically pure protein with which to pursue any number of questions regarding the mechanisms of tau action.


Assuntos
Cromatografia de Afinidade/métodos , Histidina/metabolismo , Proteínas tau/isolamento & purificação , Proteínas tau/metabolismo , Histidina/química , Histidina/genética , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas tau/genética
15.
ACS Chem Neurosci ; 7(2): 218-26, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26629788

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease characterized by extracellular deposits of amyloid ß protein (Aß) in the brain. The conversion of soluble monomers to amyloid Aß fibrils is a complicated process and involves several transient oligomeric species, which are widely believed to be highly toxic and play a crucial role in the etiology of AD. The development of inhibitors to prevent formation of small and midsized oligomers is a promising strategy for AD treatment. In this work, we employ ion mobility spectrometry (IMS), transmission electron microscopy (TEM), and molecular dynamics (MD) simulations to elucidate the structural modulation promoted by two potential inhibitors of Aß oligomerization, cucurbit[7]uril (CB[7]) and 1,2,3,4,6-penta-O-galloyl-ß-d-glucopyranose (PGG), on early oligomer and fibril formation of the Aß25-35 fragment. One and two CB[7] molecules bind to Aß25-35 monomers and dimers, respectively, and suppress aggregation by remodeling early oligomer structures and inhibiting the formation of higher-order oligomers. On the other hand, nonselective binding was observed between PGG and Aß25-35. The interactions between PGG and Aß25-35, surprisingly, enhanced the formation of Aß aggregates by promoting extended Aß25-35 conformations in both homo- and hetero-oligomers. When both ligands were present, the inhibitory effect of CB[7] overrode the stimulatory effect of PGG on Aß25-35 aggregation, suppressing the formation of large amyloid oligomers and eliminating the structural conversion from isotropic to ß-rich topologies induced by PGG. Our results provide mechanistic insights into CB[7] and PGG action on Aß oligomerization. They also demonstrate the power of the IMS technique to investigate mechanisms of multiple small-molecule agents on the amyloid formation process.


Assuntos
Peptídeos beta-Amiloides/síntese química , Peptídeos beta-Amiloides/metabolismo , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Taninos Hidrolisáveis/farmacologia , Imidazóis/farmacologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Simulação por Computador , Humanos , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Fatores de Tempo
16.
J Phys Chem B ; 119(45): 14421-32, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26484390

RESUMO

A peptide fragment of the human tau protein which stacks to form neat cross ß-sheet fibrils, resembling that found in pathological aggregation, (273)GKVQIINKKLDL(284) (here "R2/WT"), was modified with a spin-label at the N-terminus. With the resulting peptide, R2/G273C-SL, we probed events at time scales spanning seconds to hours after aggregation is initiated using transmission electron microscopy (TEM), thioflavin T (THT) fluorescence, ion mobility mass spectrometry (IMMS), electron paramagnetic resonance (EPR), and Overhauser dynamic nuclear polarization (ODNP) to determine if deliberate changes to its conformational states and population in solution influence downstream propensity to form fibrillar aggregates. We find varying solution conditions by adding the osmolyte urea or TMAO, or simply using different buffers (acetate buffer, phosphate buffer, or water), produces significant differences in early monomer/dimer populations and conformations. Crucially, these characteristics of the peptide in solution state before aggregation is initiated dictate the fibril formation propensity after aggregation. We conclude the driving forces that accelerate aggregation, when heparin is added, do not override the subtle intra- or interprotein interactions induced by the initial solvent conditions. In other words, the balance of protein-protein vs protein-solvent interactions present in the initial solution conditions is a critical driving force for fibril formation.


Assuntos
Proteínas tau/química , Sequência de Aminoácidos , Benzotiazóis , Espectroscopia de Ressonância de Spin Eletrônica , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Soluções , Espectrometria de Massas por Ionização por Electrospray , Tiazóis/química
17.
J Phys Chem B ; 119(13): 4582-93, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25775228

RESUMO

Self-aggregation of the microtubule-binding protein Tau reduces its functionality and is tightly associated with Tau-related diseases, termed tauopathies. Tau aggregation is also strongly associated with two nucleating six-residue segments, namely PHF6 (VQIVYK) and PHF6* (VQIINK). In this paper, using experiments and computational modeling, we study the self-assembly of individual and binary mixtures of Tau fragments containing PHF6* (R2/wt; (273)GKVQIINKKLDL(284)) and PHF6 (R3/wt; (306)VQIVYKPVDLSK(317)) and a mutant R2/ΔK280 associated with a neurodegenerative tauopathy. The initial stage of aggregation is probed by ion-mobility mass spectrometry, the kinetics of aggregation monitored with Thioflavin T assays, and the morphology of aggregates visualized by transmission electron microscopy. Insights into the structure of early aggregates and the factors stabilizing the aggregates are obtained from replica exchange molecular dynamics simulations. Our data suggest that R3/wt has a much stronger aggregation propensity than either R2/wt or R2/ΔK280. Heterodimers containing R3/wt are less stable than R3/wt homodimers but much more stable than homodimers of R2/wt and R2/ΔK280, suggesting a possible role of PHF6*-PHF6 interactions in initiating the aggregation of full-length Tau. Lastly, R2/ΔK280 binds more strongly to R3/wt than R2/wt, suggesting a possible mechanism for a pathological loss of normal Tau function.


Assuntos
Proteínas tau/química , Proteínas tau/genética , Benzotiazóis , Dimerização , Humanos , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Tiazóis/química
18.
J Phys Chem B ; 118(26): 7247-56, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24915112

RESUMO

Five different mutants of [Leu-5] Enkephalin YGGFL peptide have been investigated for fibril formation propensities. The early oligomer structures have been probed with a combination of ion-mobility mass spectrometry and computational modeling. The two peptides YVIFL and YVVFL form oligomers and amyloid-like fibrils. YVVFV shows an early stage oligomer distribution similar to those of the previous two, but amyloid-like aggregates are less abundant. Atomic resolution X-ray structures of YVVFV show two different modes of interactions at the dry interface between steric zippers and pairs of antiparallel ß-sheets, but both are less favorable than the packing motif found in YVVFL. Both YVVFV and YVVFL can form a Class 6 steric zipper. However, in YVVFV, the strands between mating sheets are parallel to each other and in YVVFL they are antiparallel. The overall data highlight the importance of structurally characterizing high order oligomers within oligomerization pathways in studies of nanostructure assembly.


Assuntos
Amiloide/química , Encefalinas/química , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Cristalografia por Raios X , Encefalinas/genética , Encefalinas/metabolismo , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Nanoestruturas/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
19.
J Alzheimers Dis ; 39(2): 301-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24150109

RESUMO

We tested the hypothesis that mutant tau proteins that cause neurodegeneration and dementia differentially alter kinesin translocation along microtubules (MTs) relative to normal tau in vitro. We employed complementary in vitro motility assays using purified recombinant kinesin, purified recombinant tau, and purified bovine brain α:ß tubulin to isolate interactions among these components without any contribution by cellular regulatory mechanisms. We found that kinesin translocates slower along MTs assembled by any of three independent tau mutants (4-repeat P301L tau, 4-repeat ΔN296 tau, and 4-repeat R406W tau) relative to its translocation rate along MTs assembled by normal, 4-repeat wild type (WT) tau. Moreover, the R406W mutation exhibited isoform specific effects; while kinesin translocation along 4-repeat R406W tau assembled MTs is slower than along MTs assembled by 4-repeat WT tau, the R406W mutation had no effect in the 3-repeat tau context. These data provide strong support for the notion that aberrant modulation of kinesin translocation is a component of tau-mediated neuronal cell death and dementia. Finally, we showed that assembling MTs with taxol before coating them with mutant tau obscured effects of the mutant tau that were readily apparent using more physiologically relevant MTs assembled with tau alone, raising important issues regarding the use of taxol as an experimental reagent and novel insights into therapeutic mechanisms of taxol action.


Assuntos
Encéfalo/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Mutação , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Bovinos , Humanos , Microtúbulos/efeitos dos fármacos , Doenças Neurodegenerativas/genética , Paclitaxel/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pontos Quânticos , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacologia
20.
PLoS One ; 8(9): e76409, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086739

RESUMO

Over two dozen mutations in the gene encoding the microtubule associated protein tau cause a variety of neurodegenerative dementias known as tauopathies, including frontotemporal dementia (FTD), PSP, CBD and Pick's disease. The vast majority of these mutations map to the C-terminal region of tau possessing microtubule assembly and microtubule dynamics regulatory activities as well as the ability to promote pathological tau aggregation. Here, we describe a novel and non-conservative tau mutation (G55R) mapping to an alternatively spliced exon encoding part of the N-terminal region of the protein in a patient with the behavioral variant of FTD. Although less well understood than the C-terminal region of tau, the N-terminal region can influence both MT mediated effects as well as tau aggregation. The mutation changes an uncharged glycine to a basic arginine in the midst of a highly conserved and very acidic region. In vitro, 4-repeat G55R tau nucleates microtubule assembly more effectively than wild-type 4-repeat tau; surprisingly, this effect is tau isoform specific and is not observed in a 3-repeat G55R tau versus 3-repeat wild-type tau comparison. In contrast, the G55R mutation has no effect upon the abilities of tau to regulate MT growing and shortening dynamics or to aggregate. Additionally, the mutation has no effect upon kinesin translocation in a microtubule gliding assay. Together, (i) we have identified a novel tau mutation mapping to a mutation deficient region of the protein in a bvFTD patient, and (ii) the G55R mutation affects the ability of tau to nucleate microtubule assembly in vitro in a 4-repeat tau isoform specific manner. This altered capability could markedly affect in vivo microtubule function and neuronal cell biology. We consider G55R to be a candidate mutation for bvFTD since additional criteria required to establish causality are not yet available for assessment.


Assuntos
Demência Frontotemporal/genética , Microtúbulos/metabolismo , Mutação de Sentido Incorreto/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Sequência de Aminoácidos , Feminino , Demência Frontotemporal/patologia , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Polônia , Alinhamento de Sequência , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA