Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Biochemistry ; 61(14): 1517-1530, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35759798

RESUMO

Expansion of a polyglutamine (polyQ) domain within the first exon of the huntingtin (htt) protein is the underlying cause of Huntington's disease, a genetic neurodegenerative disorder. PolyQ expansion triggers htt aggregation into oligomers, fibrils, and inclusions. The 17 N-terminal amino acids (Nt17) of htt-exon1, which directly precede the polyQ domain enhances polyQ fibrillization and functions as a lipid-binding domain. A variety of post-translational modifications occur within Nt17, including oxidation of two methionine residues. Here, the impact of oxidation within Nt17 on htt aggregation both in the presence and absence of lipid membranes was investigated. Treatment with hydrogen peroxide (H2O2) reduced fibril formation in a dose-dependent manner, resulting in shorter fibrils and an increased oligomer population. With excessive H2O2 treatments, fibrils developed a unique morphological feature around their periphery. In the presence of total brain lipid vesicles, H2O2 impacted fibrillization in a similar manner. That is, oligomerization was promoted at the expense of fibril elongation. The interaction of unoxidized and oxidized htt with supported lipid bilayers was directly observed using in situ atomic force microscopy. Without oxidation, granular htt aggregates developed on the bilayer surface. However, in the presence of H2O2, distinct plateau-like regions initially developed on the bilayer surface that gave way to rougher patches containing granular aggregates. Collectively, these observations suggest that oxidation of methionine residues within Nt17 plays a crucial role in both the aggregation of htt and its ability to interact with lipid surfaces.


Assuntos
Doença de Huntington , Peróxido de Hidrogênio , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Bicamadas Lipídicas/química , Metionina , Proteínas do Tecido Nervoso/metabolismo , Agregados Proteicos
2.
J Chem Inf Model ; 62(17): 4066-4082, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-35881533

RESUMO

The graphite-water interface provides a unique environment for polypeptides that generally favors ordered structures more than in solution. Therefore, systems consisting of designed peptides and graphitic carbon might serve as a convenient medium for controlled self-assembly of functional materials. Here, we computationally designed cyclic peptides that spontaneously fold into a ß-sheet-like conformation at the graphite-water interface and self-assemble, and we subsequently observed evidence of such assembly by atomic force microscopy. Using a novel protocol, we screened nearly 2000 sequences, optimizing for formation of a unique folded conformation while discouraging unfolded or misfolded conformations. A head-to-tail cyclic peptide with the sequence GTGSGTGGPGGGCGTGTGSGPG showed the greatest apparent propensity to fold spontaneously, and this optimized sequence was selected for larger scale molecular dynamics simulations, rigorous free-energy calculations, and experimental validation. In simulations ranging from hundreds of nanoseconds to a few microseconds, we observed spontaneous folding of this peptide at the graphite-water interface under many different conditions, including multiple temperatures (295 and 370 K), with different initial orientations relative to the graphite surface, and using different molecular dynamics force fields (CHARMM and Amber). The thermodynamic stability of the folded conformation on graphite over a range of temperatures was verified by replica-exchange simulations and free-energy calculations. On the other hand, in free solution, the folded conformation was found to be unstable, unfolding in tens of picoseconds. Intermolecular hydrogen bonds promoted self-assembly of the folded peptides into linear arrangements where the peptide backbone exhibited a tendency to align along one of the six zigzag directions of the graphite basal plane. For the optimized peptide, atomic force microscopy revealed growth of single-molecule-thick linear patterns of 6-fold symmetry, consistent with the simulations, while no such patterns were observed for a control peptide with the same amino acid composition but a scrambled sequence.


Assuntos
Grafite , Grafite/química , Simulação de Dinâmica Molecular , Peptídeos/química , Termodinâmica , Água/química
3.
Biochemistry ; 59(49): 4681-4693, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33256402

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by the abnormal expansion of a polyglutamine (polyQ) tract in the first exon of the htt protein (htt). PolyQ expansion triggers the aggregation of htt into a variety of structures, including oligomers and fibrils. This aggregation is impacted by the first 17 N-terminal amino acids (Nt17) of htt that directly precedes the polyQ domain. Beyond impacting aggregation, Nt17 associates with lipid membranes by forming an amphipathic α-helix. Post-translational modifications within Nt17 are known to modify HD pathology, and in particular, phosphorylation at T3, S13, and/or S16 retards fibrillization and ameliorates the phenotype in HD models. Due to Nt17's propensity to interact with lipid membranes, the impact of introducing phosphomimetic mutations (T3D, S13D, and S16D) into htt-exon1 on aggregation in the presence of a variety of model lipid membranes (total brain lipid extract, 1-palmitoyl-2-oleoyl-glycero-3-phosphatidylcholine, and 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-1'-rac-glycerol) was investigated. Phosphomimetic mutations altered htt's interaction with and aggregation in the presence of lipids; however, this was dependent on the lipid system.


Assuntos
Proteína Huntingtina/química , Proteína Huntingtina/genética , Mutação , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Materiais Biomiméticos/química , Fenômenos Biofísicos , Éxons , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Técnicas In Vitro , Lipídeos/química , Microscopia de Força Atômica , Mimetismo Molecular/genética , Peptídeos/química , Fosforilação , Agregados Proteicos/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
4.
Biochemistry ; 59(4): 436-449, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31814404

RESUMO

Huntington's disease is a genetic neurodegenerative disorder characterized by the formation of amyloid fibrils of the huntingtin protein (htt). The 17-residue N-terminal region of htt (Nt17) has been implicated in the formation of early phase oligomeric species, which may be neurotoxic. Because tertiary interactions with a downstream (C-terminal) polyproline (polyP) region of htt may disrupt the formation of oligomers, which are precursors to fibrillar species, the effect of co-incubation of a region of htt with a 10-residue polyP peptide on oligomerization and fibrillization has been examined by atomic force microscopy. From multiple, time-course experiments, morphological changes in oligomeric species are observed for the protein/peptide mixture and compared with the protein alone. Additionally, an overall decrease in fibril formation is observed for the heterogeneous mixture. To consider potential sites of interaction between the Nt17 region and polyP, mixtures containing Nt17 and polyP peptides have been examined by ion mobility spectrometry and gas-phase hydrogen-deuterium exchange coupled with mass spectrometry. These data combined with molecular dynamics simulations suggest that the C-terminal region of Nt17 may be a primary point of contact. One interpretation of the results is that polyP may possibly regulate Nt17 by inducing a random coil region in the C-terminal portion of Nt17, thus decreasing the propensity to form the reactive amphipathic α-helix. A separate interpretation is that the residues important for helix-helix interactions are blocked by polyP association.


Assuntos
Proteína Huntingtina/química , Doença de Huntington/metabolismo , Sequência de Aminoácidos , Amiloide/química , Amiloide/metabolismo , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Cinética , Microscopia de Força Atômica/métodos , Simulação de Dinâmica Molecular , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/química , Conformação Proteica em alfa-Hélice , Estrutura Secundária de Proteína
5.
Anal Biochem ; 609: 113864, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32846147

RESUMO

Mixed polydiacetylene (PDA) lipid vesicles mimic cell membranes and exhibit a colorimetric response induced by mechanical stress, which can be used to determine the affinity of proteins or molecules for lipid membranes. Due to a simple spectroscopic readout, PDA assays are amenable to high-throughput screens; however, these assays exhibit batch-to-batch variability. Sensitivity of the assay is also influenced by physicochemical properties associated with different lipids. Here, a method of normalizing PDA assays to reduce variability and enable direct comparison across lipid systems is described.


Assuntos
Colorimetria/métodos , Bicamadas Lipídicas/química , Polímero Poliacetilênico/análise , Peptídeos beta-Amiloides/análise , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química
6.
Biochemistry ; 58(43): 4361-4373, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31608620

RESUMO

Several diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease (HD), are associated with specific proteins aggregating and depositing within tissues and/or cellular compartments. The aggregation of these proteins is characterized by the formation of extended, ß-sheet rich fibrils, termed amyloid. In addition, a variety of other aggregate species also form, including oligomers and protofibrils. Specifically, HD is caused by the aggregation of the huntingtin (htt) protein that contains an expanded polyglutamine domain. Due to the link between protein aggregation and disease, small molecule aggregation inhibitors have been pursued as potential therapeutic agents. Two such small molecules are epigallocatechin 3-gallate (EGCG) and curcumin, both of which inhibit the fibril formation of several amyloid-forming proteins. However, amyloid formation is a complex process that is strongly influenced by the protein's environment, leading to distinct aggregation pathways. Thus, changes in the protein's environment may alter the effectiveness of aggregation inhibitors. A well-known modulator of amyloid formation is lipid membranes. Here, we investigated if the presence of lipid vesicles altered the ability of EGCG or curcumin to modulate htt aggregation and influence the interaction of htt with lipid membranes. The presence of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine or total brain lipid extract vesicles prevented the curcumin from inhibiting htt fibril formation. In contrast, EGCG's inhibition of htt fibril formation persisted in the presence of lipids. Collectively, these results highlight the complexity of htt aggregation and demonstrate that the presence of lipid membranes is a key modifier of the ability of small molecules to inhibit htt fibril formation.


Assuntos
Proteínas Amiloidogênicas/metabolismo , Catequina/análogos & derivados , Curcumina/química , Proteína Huntingtina/metabolismo , Lipossomos/química , Multimerização Proteica/efeitos dos fármacos , Catequina/química , Humanos , Fosfatidilcolinas/química
7.
Biochemistry ; 58(26): 2893-2905, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31187978

RESUMO

Alzheimer's disease (AD) is pathologically characterized by the formation of extracellular senile plaques, predominately comprised of aggregated ß-amyloid (Aß), deposited in the brain. Aß aggregation can result in a myriad of distinct aggregate species, from soluble oligomers to insoluble fibrils. Aß strongly interacts with membranes, which can be linked to a variety of potential toxic mechanisms associated with AD. Oxidative damage accompanies the formation of Aß aggregates, with a 10-50% proportion of Aß aggregates being oxidized in vivo. Hydrogen peroxide (H2O2) is a reactive oxygen species implicated in a number of neurodegenerative diseases. Recent evidence has demonstrated that the H2O2 concentration fluctuates rapidly in the brain, resulting in large concentration spikes, especially in the synaptic cleft. Here, the impact of environmental H2O2 on Aß aggregation in the presence and absence of lipid membranes is investigated. Aß40 was exposed to H2O2, resulting in the selective oxidation of methionine 35 (Met35) to produce Aß40Met35[O]. While oxidation mildly reduced the rate of Aß aggregation and produced a distinct fibril morphology at high H2O2 concentrations, H2O2 had a much more pronounced impact on Aß aggregation in the presence of total brain lipid extract vesicles. The impact of H2O2 on Aß aggregation in the presence of lipids was associated with a reduced affinity of Aß for the vesicle surface. However, this reduced vesicle affinity was predominately associated with lipid peroxidation rather than Aß oxidation.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peróxido de Hidrogênio/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Humanos , Metabolismo dos Lipídeos , Peroxidação de Lipídeos , Modelos Moleculares , Oxirredução , Agregados Proteicos
8.
Biochemistry ; 56(9): 1199-1217, 2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-28170216

RESUMO

Several hereditary neurological and neuromuscular diseases are caused by an abnormal expansion of trinucleotide repeats. To date, there have been 10 of these trinucleotide repeat disorders associated with an expansion of the codon CAG encoding glutamine (Q). For these polyglutamine (polyQ) diseases, there is a critical threshold length of the CAG repeat required for disease, and further expansion beyond this threshold is correlated with age of onset and symptom severity. PolyQ expansion in the translated proteins promotes their self-assembly into a variety of oligomeric and fibrillar aggregate species that accumulate into the hallmark proteinaceous inclusion bodies associated with each disease. Here, we review aggregation mechanisms of proteins with expanded polyQ-tracts, structural consequences of expanded polyQ ranging from monomers to fibrillar aggregates, the impact of protein context and post-translational modifications on aggregation, and a potential role for lipid membranes in aggregation. As the pathogenic mechanisms that underlie these disorders are often classified as either a gain of toxic function or loss of normal protein function, some toxic mechanisms associated with mutant polyQ tracts will also be discussed.


Assuntos
Doenças Neurodegenerativas/metabolismo , Peptídeos/química , Proteínas/química , Proteínas/metabolismo , Animais , Humanos , Processamento de Proteína Pós-Traducional
9.
Biophys J ; 111(2): 349-362, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27463137

RESUMO

Huntington's disease (HD) is a genetic neurodegenerative disorder caused by an expanded polyglutamine (polyQ) domain near the N-terminus of the huntingtin (htt) protein. Expanded polyQ leads to htt aggregation. The first 17 amino acids (Nt(17)) in htt comprise a lipid-binding domain that undergoes a number of posttranslational modifications that can modulate htt toxicity and subcellular localization. As there are three lysines within Nt(17), we evaluated the impact of lysine acetylation on htt aggregation in solution and on model lipid bilayers. Acetylation of htt-exon1(51Q) and synthetic truncated htt-exon 1 mimicking peptides (Nt(17)-Q35-P10-KK) was achieved using a selective covalent label, sulfo-N-hydroxysuccinimide (NHSA). With this treatment, all three lysine residues (K6, K9, and K15) in Nt(17) were significantly acetylated. N-terminal htt acetylation retarded fibril formation in solution and promoted the formation of larger globular aggregates. Acetylated htt also bound lipid membranes and disrupted the lipid bilayer morphology less aggressively compared with the wild-type. Computational studies provided mechanistic insights into how acetylation alters the interaction of Nt(17) with lipid membranes. Our results highlight that N-terminal acetylation influences the aggregation of htt and its interaction with lipid bilayers.


Assuntos
Éxons , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Bicamadas Lipídicas/metabolismo , Agregados Proteicos , Acetilação , Sequência de Aminoácidos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Proteína Huntingtina/genética , Camundongos , Peptídeos/metabolismo
10.
Biochemistry ; 55(1): 92-102, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26652744

RESUMO

Huntington's disease (HD) is an inherited neurodegenerative disease caused by abnormally long CAG-repeats in the huntingtin gene that encode an expanded polyglutamine (polyQ) domain near the N-terminus of the huntingtin (htt) protein. Expanded polyQ domains are directly correlated to disease-related htt aggregation. Htt is found highly associated with a variety of cellular and subcellular membranes that are predominantly comprised of lipids. Since cholesterol homeostasis is altered in HD, we investigated how varying cholesterol content modifies the interactions between htt and lipid membranes. A combination of Langmuir trough monolayer techniques, vesicle permeability and binding assays, and in situ atomic force microscopy were used to directly monitor the interaction of a model, synthetic htt peptide and a full-length htt-exon1 recombinant protein with model membranes comprised of total brain lipid extract (TBLE) and varying amounts of exogenously added cholesterol. As the cholesterol content of the membrane increased, the extent of htt insertion decreased. Vesicles containing extra cholesterol were resistant to htt-induced permeabilization. Morphological and mechanical changes in the bilayer associated with exposure to htt were also drastically altered by the presence of cholesterol. Disrupted regions of pure TBLE bilayers were grainy in appearance and associated with a large number of globular aggregates. In contrast, morphological changes induced by htt in bilayers enriched in cholesterol were plateau-like with a smooth appearance. Collectively, these observations suggest that the presence and amount of cholesterol in lipid membranes play a critical role in htt binding and aggregation on lipid membranes.


Assuntos
Colesterol/metabolismo , Doença de Huntington/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Lipossomas Unilamelares/metabolismo , Humanos , Proteína Huntingtina , Proteínas do Tecido Nervoso/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Permeabilidade , Agregação Patológica de Proteínas/metabolismo , Ligação Proteica
11.
Biochemistry ; 54(28): 4285-96, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26098795

RESUMO

Early stage oligomer formation of the huntingtin protein may be driven by self-association of the 17-residue amphipathic α-helix at the protein's N-terminus (Nt17). Oligomeric structures have been implicated in neuronal toxicity and may represent important neurotoxic species in Huntington's disease. Therefore, a residue-specific structural characterization of Nt17 is crucial to understanding and potentially inhibiting oligomer formation. Native electrospray ion mobility spectrometry-mass spectrometry (IMS-MS) techniques and molecular dynamics simulations (MDS) have been applied to study coexisting monomer and multimer conformations of Nt17, independent of the remainder of huntingtin exon 1. MDS suggests gas-phase monomer ion structures comprise a helix-turn-coil configuration and a helix-extended-coil region. Elongated dimer species comprise partially helical monomers arranged in an antiparallel geometry. This stacked helical bundle may represent the earliest stages of Nt17-driven oligomer formation. Nt17 monomers and multimers have been further probed using diethylpyrocarbonate (DEPC). An N-terminal site (N-terminus of Threonine-3) and Lysine-6 are modified at higher DEPC concentrations, which led to the formation of an intermediate monomer structure. These modifications resulted in decreased extended monomer ion conformers, as well as a reduction in multimer formation. From the MDS experiments for the dimer ions, Lys6 residues in both monomer constituents interact with Ser16 and Glu12 residues on adjacent peptides; therefore, the decrease in multimer formation could result from disruption of these or similar interactions. This work provides a structurally selective model from which to study Nt17 self-association and provides critical insight toward Nt17 multimerization and, possibly, the early stages of huntingtin exon 1 aggregation.


Assuntos
Proteínas do Tecido Nervoso/química , Dietil Pirocarbonato/química , Humanos , Proteína Huntingtina , Lisina/química , Espectrometria de Massas , Simulação de Dinâmica Molecular , Peptídeos/química , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Treonina/química
12.
Biochemistry ; 53(45): 7038-50, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25349919

RESUMO

The appearance of neuritic amyloid plaques comprised of ß-amyloid peptide (Aß) in the brain is a predominant feature in Alzheimer's disease (AD). In the aggregation process, Aß samples a variety of potentially toxic aggregate species, ranging from small oligomers to fibrils. Aß has the ability to form a variety of morphologically distinct and stable amyloid fibrils. Commonly termed polymorphs, such distinct aggregate species may play a role in variations of AD pathology. It has been well documented that polymorphic aggregates of Aß can be produced by changes in the chemical environment and peptide preparations. As Aß and several of its aggregated forms are known to interact directly with lipid membranes and this interaction may play a role in a variety of potential toxic mechanisms associated with AD, we determine how different Aß(1-40) preparation protocols that lead to distinct polymorphic fibril aggregates influence the interaction of Aß(1-40) with model lipid membranes. Using three distinct protocols for preparing Aß(1-40), the aggregate species formed in the absence and presence of a lipid bilayers were investigated using a variety of scanning probe microscopy techniques. The three preparations of Aß(1-40) promoted distinct oligomeric and fibrillar aggregates in the absence of bilayers that formed at different rates. Despite these differences in aggregation properties, all Aß(1-40) preparations were able to disrupt supported total brain lipid extract bilayers, altering the bilayer's morphological and mechanical properties.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Bicamadas Lipídicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos/fisiologia , Peptídeos beta-Amiloides/química , Bicamadas Lipídicas/química , Fragmentos de Peptídeos/química , Ligação Proteica/fisiologia
13.
Biochemistry ; 53(14): 2355-65, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24670006

RESUMO

Huntington disease (HD) is a genetic neurodegenerative disease caused by an expanded polyglutamine (polyQ) domain in the first exon of the huntingtin (Htt) protein, facilitating its aggregation. Htt interacts with a variety of membraneous structures within the cell, and the first 17 amino acids (Nt17) of Htt directly flanking the polyQ domain comprise an amphiphathic α-helix (AH) lipid-binding domain. AHs are also known to detect membrane curvature. To determine if Htt exon 1 preferentially binds curved membranes, in situ atomic force microscopy (AFM) studies were performed. Supported lipid bilayers are commonly used as model membranes for AFM studies of protein aggregation. However, these supported bilayers usually lack curvature. By forming a bilayer on top of silica nanobeads (50 ± 10 nm) deposited on a silicon substrate, model supported lipid bilayers with flat and curved regions were developed for AFM studies. The presence of the bilayer over the beads was validated by continual imaging of the formation of the bilayer, height measurements, and spatially resolved mechanical measurements of the resulting bilayer using scanning probe acceleration microscopy. Interpretation of this data was facilitated by numerical simulations of the entire imaging process. The curved supported bilayers associated with the beads were found to be more compliant than flat supported bilayers, consistent with the altered packing density of lipids caused by the induced curvature. This model bilayer system was exposed to a synthetic truncated Htt exon 1 peptide (Nt17Q35P10KK), and this peptide preferentially accumulated on curved membranes, consistent with the ability of AHs to sense membrane curvature.


Assuntos
Bicamadas Lipídicas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Huntingtina , Microscopia de Força Atômica , Nanopartículas , Proteínas do Tecido Nervoso/química , Ligação Proteica
14.
J Biol Chem ; 288(21): 14993-5005, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23572526

RESUMO

Huntington disease (HD) is caused by an expanded polyglutamine (poly(Q)) repeat near the N terminus of the huntingtin (htt) protein. Expanded poly(Q) facilitates formation of htt aggregates, eventually leading to deposition of cytoplasmic and intranuclear inclusion bodies containing htt. Flanking sequences directly adjacent to the poly(Q) domain, such as the first 17 amino acids on the N terminus (Nt17) and the polyproline (poly(P)) domain on the C-terminal side of the poly(Q) domain, heavily influence aggregation. Additionally, htt interacts with a variety of membraneous structures within the cell, and Nt17 is implicated in lipid binding. To investigate the interaction between htt exon1 and lipid membranes, a combination of in situ atomic force microscopy, Langmuir trough techniques, and vesicle permeability assays were used to directly monitor the interaction of a variety of synthetic poly(Q) peptides with different combinations of flanking sequences (KK-Q35-KK, KK-Q35-P10-KK, Nt17-Q35-KK, and Nt17-Q35-P10-KK) on model membranes and surfaces. Each peptide aggregated on mica, predominately forming extended, fibrillar aggregates. In contrast, poly(Q) peptides that lacked the Nt17 domain did not appreciably aggregate on or insert into lipid membranes. Nt17 facilitated the interaction of peptides with lipid surfaces, whereas the poly(P) region enhanced this interaction. The aggregation of Nt17-Q35-P10-KK on the lipid bilayer closely resembled that of a htt exon1 construct containing 35 repeat glutamines. Collectively, this data suggests that the Nt17 domain plays a critical role in htt binding and aggregation on lipid membranes, and this lipid/htt interaction can be further modulated by the presence of the poly(P) domain.


Assuntos
Bicamadas Lipídicas/química , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Peptídeos/química , Animais , Éxons , Proteína Huntingtina , Bicamadas Lipídicas/metabolismo , Camundongos , Microscopia de Força Atômica , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
15.
Biochim Biophys Acta ; 1828(8): 1953-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23643759

RESUMO

Huntington's Disease (HD) is a neurodegenerative disorder that is defined by the accumulation of nanoscale aggregates comprised of the huntingtin (htt) protein. Aggregation is directly caused by an expanded polyglutamine (polyQ) domain in htt, leading to a diverse population of aggregate species, such as oligomers, fibrils, and annular aggregates. Furthermore, the length of this polyQ domain is directly related to onset and severity of disease. The first 17 N-terminal amino acids of htt have been shown to further modulate aggregation. Additionally, these 17 amino acids appear to have lipid binding properties as htt interacts with a variety of membrane-containing structures present in cells, such as organelles, and interactions with these membrane surfaces may further modulate htt aggregation. To investigate the interaction between htt exon1 and lipid bilayers, in situ atomic force microscopy (AFM) was used to directly monitor the aggregation of htt exon1 constructs with varying Q-lengths (35Q, 46Q, 51Q, and myc-53Q) on supported lipid membranes comprised of total brain lipid extract. The exon1 fragments accumulated on the lipid membranes, causing disruption of the membrane, in a polyQ dependent manner. Furthermore, the addition of an N-terminal myc-tag to the htt exon1 fragments impeded the interaction of htt with the bilayer.


Assuntos
Bicamadas Lipídicas/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Éxons/genética , Humanos , Proteína Huntingtina , Bicamadas Lipídicas/química , Lipídeos de Membrana/química , Microscopia de Força Atômica , Modelos Moleculares , Proteínas do Tecido Nervoso/genética , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
16.
Biochim Biophys Acta Biomembr ; 1866(6): 184339, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38763270

RESUMO

Huntington's Disease (HD) is caused by an abnormal expansion of the polyglutamine (polyQ) domain within the first exon of the huntingtin protein (htt). This expansion promotes disease-related htt aggregation into amyloid fibrils and the formation of proteinaceous inclusion bodies within neurons. Fibril formation is a complex heterogenous process involving an array of aggregate species such as oligomers, protofibrils, and fibrils. In HD, structural abnormalities of membranes of several organelles develop. In particular, the accumulation of htt fibrils near the endoplasmic reticulum (ER) impinges upon the membrane, resulting in ER damage, altered dynamics, and leakage of Ca2+. Here, the aggregation of htt at a bilayer interface assembled from ER-derived liposomes was investigated, and fibril formation directly on these membranes was enhanced. Based on these observations, simplified model systems were used to investigate mechanisms associated with htt aggregation on ER membranes. As the ER-derived liposome fractions contained residual Ca2+, the role of divalent cations was also investigated. In the absence of lipids, divalent cations had minimal impact on htt structure and aggregation. However, the presence of Ca2+ or Mg2+ played a key role in promoting fibril formation on lipid membranes despite reduced htt insertion into and association with lipid interfaces, suggesting that the ability of divalent cations to promote fibril formation on membranes is mediated by induced changes to the lipid membrane physicochemical properties. With enhanced concentrations of intracellular calcium being a hallmark of HD, the ability of divalent cations to influence htt aggregation at lipid membranes may play a role in aggregation events that lead to organelle abnormalities associated with disease.


Assuntos
Amiloide , Cálcio , Cátions Bivalentes , Retículo Endoplasmático , Proteína Huntingtina , Doença de Huntington , Lipossomos , Retículo Endoplasmático/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/química , Humanos , Cátions Bivalentes/metabolismo , Cálcio/metabolismo , Amiloide/metabolismo , Amiloide/química , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Doença de Huntington/genética , Lipossomos/química , Lipossomos/metabolismo , Magnésio/metabolismo , Magnésio/química , Peptídeos
17.
PLoS One ; 19(3): e0298323, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38483973

RESUMO

Huntington's Disease (HD) is a fatal, neurodegenerative disease caused by aggregation of the huntingtin protein (htt) with an expanded polyglutamine (polyQ) domain into amyloid fibrils. Htt aggregation is modified by flanking sequences surrounding the polyQ domain as well as the binding of htt to lipid membranes. Upon fibrillization, htt fibrils are able to template the aggregation of monomers into fibrils in a phenomenon known as seeding, and this process appears to play a critical role in cell-to-cell spread of HD. Here, exposure of C. elegans expressing a nonpathogenic N-terminal htt fragment (15-repeat glutamine residues) to preformed htt-exon1 fibrils induced inclusion formation and resulted in decreased viability in a dose dependent manner, demonstrating that seeding can induce toxic aggregation of nonpathogenic forms of htt. To better understand this seeding process, the impact of flanking sequences adjacent to the polyQ stretch, polyQ length, and the presence of model lipid membranes on htt seeding was investigated. Htt seeding readily occurred across polyQ lengths and was independent of flanking sequence, suggesting that the structured polyQ domain within fibrils is the key contributor to the seeding phenomenon. However, the addition of lipid vesicles modified seeding efficiency in a manner suggesting that seeding primarily occurs in bulk solution and not at the membrane interface. In addition, fibrils formed in the presence of lipid membranes displayed similar seeding efficiencies. Collectively, this suggests that the polyQ domain that forms the amyloid fibril core is the main driver of seeding in htt aggregation.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Peptídeos , Animais , Humanos , Proteína Huntingtina/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Amiloide/metabolismo , Lipídeos
18.
Biochemistry ; 52(5): 808-17, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23331195

RESUMO

A diverse number of diseases, including Alzheimer's disease, Huntington's disease, and type 2 diabetes, are characterized by the formation of fibrillar protein aggregates termed amyloids. The precise mechanism by which aggregates are toxic remains unclear; however, these proteins have been shown to interact strongly with lipid membranes. We investigated morphological and mechanical changes in model lipid bilayers exposed to amyloid-forming proteins by reconstructing the tapping forces associated with atomic force microscopy (AFM) imaging in solution. Tip/sample tapping forces contain information regarding mechanical properties of surfaces. Interpretation of the mechanical changes in the bilayers was aided by numerical simulations of the entire AFM experiment. Amyloid-forming proteins disrupted distinct regions of the bilayer morphology, and these regions were associated with decreased Young's modulus and adhesive properties. These changes in bilayer mechanical properties upon exposure to amyloid-forming proteins may represent a common mechanism leading to membrane dysfunction in amyloid diseases.


Assuntos
Proteínas Amiloidogênicas/metabolismo , Bicamadas Lipídicas/metabolismo , Peptídeos beta-Amiloides/metabolismo , Módulo de Elasticidade , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Bicamadas Lipídicas/química , Microscopia de Força Atômica , Fragmentos de Peptídeos/metabolismo
19.
J Biol Chem ; 287(19): 16017-28, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22433867

RESUMO

Huntington disease is a genetic neurodegenerative disorder that arises from an expanded polyglutamine region in the N terminus of the HD gene product, huntingtin. Protein inclusions comprised of N-terminal fragments of mutant huntingtin are a characteristic feature of disease, though are likely to play a protective role rather than a causative one in neurodegeneration. Soluble oligomeric assemblies of huntingtin formed early in the aggregation process are candidate toxic species in HD. In the present study, we established an in vitro system to generate recombinant huntingtin in mammalian cells. Using both denaturing and native gel analysis, we have identified novel oligomeric forms of mammalian-derived expanded huntingtin exon-1 N-terminal fragment. These species are transient and were not previously detected using bacterially expressed exon-1 protein. Importantly, these species are recognized by 3B5H10, an antibody that recognizes a two-stranded hairpin conformation of expanded polyglutamine believed to be associated with a toxic form of huntingtin. Interestingly, comparable oligomeric species were not observed for expanded huntingtin shortstop, a 117-amino acid fragment of huntingtin shown previously in mammalian cell lines and transgenic mice, and here in primary cortical neurons, to be non-toxic. Further, we demonstrate that expanded huntingtin shortstop has a reduced ability to form amyloid-like fibrils characteristic of the aggregation pathway for toxic expanded polyglutamine proteins. Taken together, these data provide a possible candidate toxic species in HD. In addition, these studies demonstrate the fundamental differences in early aggregation events between mutant huntingtin exon-1 and shortstop proteins that may underlie the differences in toxicity.


Assuntos
Éxons/genética , Proteínas do Tecido Nervoso/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Western Blotting , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Microscopia de Força Atômica , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Peptídeos/genética , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
20.
Nat Chem Biol ; 7(12): 925-34, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037470

RESUMO

Polyglutamine (polyQ) stretches exceeding a threshold length confer a toxic function to proteins that contain them and cause at least nine neurological disorders. The basis for this toxicity threshold is unclear. Although polyQ expansions render proteins prone to aggregate into inclusion bodies, this may be a neuronal coping response to more toxic forms of polyQ. The exact structure of these more toxic forms is unknown. Here we show that the monoclonal antibody 3B5H10 recognizes a species of polyQ protein in situ that strongly predicts neuronal death. The epitope selectively appears among some of the many low-molecular-weight conformational states assumed by expanded polyQ and disappears in higher-molecular-weight aggregated forms, such as inclusion bodies. These results suggest that protein monomers and possibly small oligomers containing expanded polyQ stretches can adopt a conformation that is recognized by 3B5H10 and is toxic or closely related to a toxic species.


Assuntos
Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Peptídeos/química , Peptídeos/toxicidade , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Epitopos/química , Epitopos/imunologia , Epitopos/toxicidade , Células HEK293 , Humanos , Corpos de Inclusão/química , Peso Molecular , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Peptídeos/imunologia , Relação Estrutura-Atividade , Expansão das Repetições de Trinucleotídeos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa