Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Neurobiol Dis ; 159: 105517, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34563643

RESUMEN

Huntington's disease (HD) is a genetically inherited neurodegenerative disorder caused by expansion of a polyglutamine (polyQ) repeat in the exon-1 of huntingtin protein (HTT). The expanded polyQ enhances the amyloidogenic propensity of HTT exon 1 (HTTex1), which forms a heterogeneous mixture of assemblies with a broad neurotoxicity spectrum. While predominantly intracellular, monomeric and aggregated mutant HTT species are also present in the cerebrospinal fluids of HD patients, however, their biological properties are not well understood. To explore the role of extracellular mutant HTT in aggregation and toxicity, we investigated the uptake and amplification of recombinant HTTex1 assemblies in cell culture models. We find that small HTTex1 fibrils preferentially enter human neurons and trigger the amplification of neurotoxic assemblies; astrocytes or epithelial cells are not permissive. The amplification of HTTex1 in neurons depletes endogenous HTT protein with non-pathogenic polyQ repeat, activates apoptotic caspase-3 pathway and induces nuclear fragmentation. Using a panel of novel monoclonal antibodies and genetic mutation, we identified epitopes within the N-terminal 17 amino acids and proline-rich domain of HTTex1 to be critical in neural uptake and amplification. Synaptosome preparations from the brain homogenates of HD mice also contain mutant HTT species, which enter neurons and behave similar to small recombinant HTTex1 fibrils. These studies suggest that amyloidogenic extracellular mutant HTTex1 assemblies may preferentially enter neurons, propagate and promote neurodegeneration.


Asunto(s)
Astrocitos/metabolismo , Células Epiteliales/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Animales , Apoptosis , Caspasa 3 , Exones , Técnicas de Sustitución del Gen , Humanos , Proteína Huntingtina/genética , Ratones , Ratones Transgénicos , Mutación , Péptidos/genética , Agregación Patológica de Proteínas/genética , Sinaptosomas
2.
Hum Mol Genet ; 27(13): 2330-2343, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29912367

RESUMEN

The N-terminal fragments of mutant huntingtin (mHTT) misfold and assemble into oligomers, which ultimately bundle into insoluble fibrils. Conformations unique to various assemblies of mHTT remain unknown. Knowledge on the half-life of various multimeric structures of mHTT is also scarce. Using a panel of four new antibodies named PHP1-4, we have identified new conformations in monomers and assembled structures of mHTT. PHP1 and PHP2 bind to epitopes within the proline-rich domain (PRD), whereas PHP3 and PHP4 interact with motifs formed at the junction of polyglutamine (polyQ) and polyproline (polyP) repeats of HTT. The PHP1- and PHP2-reactive epitopes are exposed in fibrils of mHTT exon1 (mHTTx1) generated from recombinant proteins and mHTT assemblies, which progressively accumulate in the nuclei, cell bodies and neuropils in the brains of HD mouse models. Notably, electron microscopic examination of brain sections of HD mice revealed that PHP1- and PHP2-reactive mHTT assemblies are present in myelin sheath and in vesicle-like structures. Moreover, PHP1 and PHP2 antibodies block seeding and subsequent fibril assembly of mHTTx1 in vitro and in a cell culture model of HD. PHP3 and PHP4 bind to epitopes in full-length and N-terminal fragments of monomeric mHTT and binding diminishes as the mHTTx1 assembles into fibrils. Interestingly, PHP3 and PHP4 also prevent the aggregation of mHTTx1 in vitro highlighting a regulatory function for the polyQ-polyP motifs. These newly detected conformations may affect fibril assembly, stability and intercellular transport of mHTT.


Asunto(s)
Proteína Huntingtina , Secuencias de Aminoácidos , Animales , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Ratones , Ratones Transgénicos , Agregado de Proteínas , Dominios Proteicos
3.
Proc Natl Acad Sci U S A ; 114(22): 5629-5634, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28228524

RESUMEN

Eps15 (epidermal growth factor receptor pathway substrate 15)-homology domain containing proteins (EHDs) comprise a family of dynamin-related mechano-chemical ATPases involved in cellular membrane trafficking. Previous studies have revealed the structure of the EHD2 dimer, but the molecular mechanisms of membrane recruitment and assembly have remained obscure. Here, we determined the crystal structure of an amino-terminally truncated EHD4 dimer. Compared with the EHD2 structure, the helical domains are 50° rotated relative to the GTPase domain. Using electron paramagnetic spin resonance (EPR), we show that this rotation aligns the two membrane-binding regions in the helical domain toward the lipid bilayer, allowing membrane interaction. A loop rearrangement in GTPase domain creates a new interface for oligomer formation. Our results suggest that the EHD4 structure represents the active EHD conformation, whereas the EHD2 structure is autoinhibited, and reveal a complex series of domain rearrangements accompanying activation. A comparison with other peripheral membrane proteins elucidates common and specific features of this activation mechanism.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos/genética , Línea Celular Tumoral , Cristalografía por Rayos X , Activación Enzimática/fisiología , Células HeLa , Humanos , Unión Proteica , Dominios Proteicos/fisiología , Multimerización de Proteína , Transporte de Proteínas/fisiología
4.
J Biol Chem ; 293(7): 2597-2605, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29282287

RESUMEN

Aggregation of huntingtin protein arising from expanded polyglutamine (polyQ) sequences in the exon-1 region of mutant huntingtin plays a central role in the pathogenesis of Huntington's disease. The huntingtin aggregation pathways are of therapeutic and diagnostic interest, but obtaining critical information from the physiologically relevant htt exon-1 (Httex1) protein has been challenging. Using biophysical techniques and an expression and purification protocol that generates clean, monomeric Httex1, we identified and mapped three distinct aggregation pathways: 1) unseeded in solution; 2) seeded in solution; and 3) membrane-mediated. In solution, aggregation proceeded in a highly stepwise manner, in which the individual domains (N terminus containing 17 amino acids (N17), polyQ, and proline-rich domain (PRD)) become ordered at very different rates. The aggregation was initiated by an early oligomer requiring a pathogenic, expanded Gln length and N17 α-helix formation. In the second phase, ß-sheet forms in the polyQ. The slowest step is the final structural maturation of the PRD. This stepwise mechanism could be bypassed by seeding, which potently accelerated aggregation and was a prerequisite for prion-like spreading in vivo Remarkably, membranes could catalyze aggregation even more potently than seeds, in a process that caused significant membrane damage. The N17 governed membrane-mediated aggregation by anchoring Httex1 to the membrane, enhancing local concentration and promoting collision via two-dimensional diffusion. Considering its central roles in solution and in membrane-mediated aggregation, the N17 represents an attractive target for inhibiting multiple pathways. Our approach should help evaluate such inhibitors and identify diagnostic markers for the misfolded forms identified here.


Asunto(s)
Membrana Celular/metabolismo , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Secuencias de Aminoácidos , Membrana Celular/química , Membrana Celular/genética , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Cinética , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Agregado de Proteínas , Conformación Proteica en Hélice alfa , Dominios Proteicos
5.
Biochemistry ; 56(28): 3579-3586, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28621522

RESUMEN

The fact that the heritable neurodegenerative disorder Huntington's disease (HD) is autosomal dominant means that there is one wild type and one mutant allele in most HD patients. The CAG repeat expansion in the exon 1 of the protein huntingtin (HTTex1) that causes the disease leads to the formation of HTT fibrils in vitro and vivo. An important question for understanding the molecular mechanism of HD is which role wild type HTT plays for the formation, propagation, and structure of these HTT fibrils. Here we report that fibrils of mutant HTTex1 are able to seed the aggregation of wild type HTTex1 into amyloid fibrils, which in turn can seed the fibril formation of mutant HTTex1. Solid-state NMR and electron paramagnetic resonance data showed that wild type HTTex1 fibrils closely resemble the structure of mutant fibrils, with small differences indicating a less extended fibril core. These data suggest that wild type fibrils can faithfully perpetuate the structure of mutant fibrils in HD. However, wild type HTTex1 monomers have a much higher equilibrium solubility compared to mutant HTTex1, and only a small fraction incorporates into fibrils.


Asunto(s)
Amiloide/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Amiloide/química , Amiloide/ultraestructura , Exones , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/ultraestructura , Enfermedad de Huntington/metabolismo , Mutación , Resonancia Magnética Nuclear Biomolecular , Agregado de Proteínas , Solubilidad
6.
J Am Chem Soc ; 138(36): 11526-35, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27548572

RESUMEN

Water dynamics in the hydration shell of the peripheral membrane protein annexin B12 were studied using MD simulations and Overhauser DNP-enhanced NMR. We show that retardation of water motions near phospholipid bilayers is extended by the presence of a membrane-bound protein, up to around 10 Å above that protein. Near the membrane surface, electrostatic interactions with the lipid head groups strongly slow down water dynamics, whereas protein-induced water retardation is weaker and dominates only at distances beyond 10 Å from the membrane surface. The results can be understood from a simple model based on additive contributions from the membrane and the protein to the activation free energy barriers of water diffusion next to the biomolecular surfaces. Furthermore, analysis of the intermolecular vibrations of the water network reveals that retarded water motions near the membrane shift the vibrational modes to higher frequencies, which we used to identify an entropy gradient from the membrane surface toward the bulk water. Our results have implications for processes that take place at lipid membrane surfaces, including molecular recognition, binding, and protein-protein interactions.


Asunto(s)
Proteínas de la Membrana/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Difusión , Entropía , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Proteínas de la Membrana/química , Simulación de Dinámica Molecular , Propiedades de Superficie , Agua/metabolismo
7.
Biochemistry ; 54(25): 3942-9, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26020223

RESUMEN

Amyloid-like fibrils formed by huntingtin exon-1 (htt_ex1) are a hallmark of Huntington's disease (HD). The structure of these fibrils is unknown, and determining their structure is an important step toward understanding the misfolding processes that cause HD. In HD, a polyglutamine (polyQ) domain in htt_ex1 is expanded to a degree that it gains the ability to form aggregates comprising the core of the resulting fibrils. Despite the simplicity of this polyQ sequence, the structure of htt_ex1 fibrils has been difficult to determine. This study provides a detailed structural investigation of fibrils formed by htt_ex1 using solid-state nuclear magnetic resonance (NMR) spectroscopy. We show that the polyQ domain of htt_ex1 forms the static amyloid core similar to polyQ model peptides. The Gln residues of this domain exist in two distinct conformations that are found in separate domains or monomers but are relatively close in space. The rest of htt_ex1 is relatively dynamic on an NMR time scale, especially the proline-rich C-terminus, which we found to be in a polyproline II helical and random coil conformation. We observed a similar dynamic C-terminus in a soluble form of htt_ex1, indicating that the conformation of this part of htt_ex1 is not changed upon its aggregation into an amyloid fibril. From these data, we propose a bottlebrush model for the fibrils formed by htt_ex1. In this model, the polyQ domains form the center and the proline-rich domains the bristles of the bottlebrush.


Asunto(s)
Amiloide/química , Exones , Proteínas del Tejido Nervioso/química , Amiloide/genética , Amiloide/metabolismo , Humanos , Proteína Huntingtina , Espectroscopía de Resonancia Magnética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Agregado de Proteínas , Pliegue de Proteína , Estructura Terciaria de Proteína
8.
J Biol Chem ; 287(8): 5235-41, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22187437

RESUMEN

Misfolding and amyloid fibril formation by human islet amyloid polypeptide (hIAPP) are thought to be important in the pathogenesis of type 2 diabetes, but the structures of the misfolded forms remain poorly understood. Here we developed an approach that combines site-directed spin labeling with continuous wave and pulsed EPR to investigate local secondary structure and to determine the relative orientation of the secondary structure elements with respect to each other. These data indicated that individual hIAPP molecules take up a hairpin fold within the fibril. This fold contains two ß-strands that are much farther apart than expected from previous models. Atomistic structural models were obtained using computational refinement with EPR data as constraints. The resulting family of structures exhibited a left-handed helical twist, in agreement with the twisted morphology observed by electron microscopy. The fibril protofilaments contain stacked hIAPP monomers that form opposing ß-sheets that twist around each other. The two ß-strands of the monomer adopt out-of-plane positions and are staggered by about three peptide layers (∼15 Å). These results provide a mechanism for hIAPP fibril formation and could explain the remarkable stability of the fibrils. Thus, the structural model serves as a starting point for understanding and preventing hIAPP misfolding.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Multimerización de Proteína , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Microscopía Electrónica , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Estabilidad Proteica , Estructura Secundaria de Proteína , alfa-Sinucleína/metabolismo
9.
J Biol Chem ; 287(38): 31739-46, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22801429

RESUMEN

Misfolding and aggregation of huntingtin is one of the hallmarks of Huntington disease, but the overall structure of these aggregates and the mechanisms by which huntingtin misfolds remain poorly understood. Here we used site-directed spin labeling and electron paramagnetic resonance (EPR) spectroscopy to study the structural features of huntingtin exon 1 (HDx1) containing 46 glutamine residues in its polyglutamine (polyQ) region. Despite some residual structuring in the N terminus, we find that soluble HDx1 is highly dynamic. Upon aggregation, the polyQ domain becomes strongly immobilized indicating significant tertiary or quaternary packing interactions. Analysis of spin-spin interactions does not show the close contact between same residues that is characteristic of the parallel, in-register structure commonly found in amyloids. Nevertheless, the same residues are still within 20 Å of each other, suggesting that polyQ domains from different molecules come into proximity in the fibrils. The N terminus has previously been found to take up a helical structure in fibrils. We find that this domain not only becomes structured, but that it also engages in tertiary or quaternary packing interactions. The existence of spin-spin interactions in this region suggests that such contacts could be made between N-terminal domains from different molecules. In contrast, the C-terminal domain is dynamic, contains polyproline II structure, and lacks pronounced packing interactions. This region must be facing away from the core of the fibrils. Collectively, these data provide new constraints for building structural models of HDx1 fibrils.


Asunto(s)
Proteínas del Tejido Nervioso/química , Bioquímica/métodos , Dicroismo Circular , Espectroscopía de Resonancia por Spin del Electrón , Exones , Humanos , Proteína Huntingtina , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Péptidos/química , Conformación Proteica , Desnaturalización Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Tiorredoxinas/química
10.
J Biol Chem ; 285(9): 6071-9, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20018889

RESUMEN

Soluble amyloid oligomers are potent neurotoxins that are involved in a wide range of human degenerative diseases, including Alzheimer disease. In Alzheimer disease, amyloid beta (Abeta) oligomers bind to neuronal synapses, inhibit long term potentiation, and induce cell death. Recent evidence indicates that several immunologically distinct structural variants exist as follows: prefibrillar oligomers (PFOs), fibrillar oligomers (FOs), and annular protofibrils. Despite widespread interest, amyloid oligomers are poorly characterized in terms of structural differences and pathological significance. FOs are immunologically related to fibrils because they react with OC, a conformation-dependent, fibril-specific antibody and do not react with antibodies specific for other types of oligomers. However, fibrillar oligomers are much smaller than fibrils. FOs are soluble at 100,000 x g, rich in beta-sheet structures, but yet bind weakly to thioflavin T. EPR spectroscopy indicates that FOs display significantly more spin-spin interaction at multiple labeled sites than PFOs and are more structurally similar to fibrils. Atomic force microscopy indicates that FOs are approximately one-half to one-third the height of mature fibrils. We found that Abeta FOs do not seed the formation of thioflavin T-positive fibrils from Abeta monomers but instead seed the formation of FOs from Abeta monomers that are positive for the OC anti-fibril antibody. These results indicate that the lattice of FOs is distinct from the fibril lattice even though the polypeptide chains are organized in an immunologically identical conformation. The FOs resulting from seeded reactions have the same dimensions and morphology as the initial seeds, suggesting that the seeds replicate by growing to a limiting size and then splitting, indicating that their lattice is less stable than fibrils. We suggest that FOs may represent small pieces of single fibril protofilament and that the addition of monomers to the ends of FOs is kinetically more favorable than the assembly of the oligomers into fibrils via sheet stacking interaction. These studies provide novel structural insight into the relationship between fibrils and FOs and suggest that the increased toxicity of FOs may be due to their ability to replicate and the exposure of hydrophobic sheet surfaces that are otherwise obscured by sheet-sheet interactions between protofilaments in a fibril.


Asunto(s)
Péptidos beta-Amiloides/química , Fragmentos de Péptidos/química , Multimerización de Proteína , Amiloide , Cristalización , Humanos , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/toxicidad , Estructura Secundaria de Proteína
11.
Sci Rep ; 10(1): 5301, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210350

RESUMEN

Membrane protein oligomerization mediates a wide range of biological events including signal transduction, viral infection and membrane curvature induction. However, the relative contributions of protein-protein and protein-membrane interactions to protein oligomerization remain poorly understood. Here, we used the Ca2+-dependent membrane-binding protein ANXB12 as a model system to determine the relative contributions of protein-protein and protein-membrane interactions toward trimer formation. Using an EPR-based detection method, we find that some protein-protein interactions are essential for trimer formation. Surprisingly, these interactions are largely hydrophobic, and they do not include the previously identified salt bridges, which are less important. Interfering with membrane interaction by mutating selected Ca2+-ligands or by introducing Lys residues in the membrane-binding loops had variable, strongly position-dependent effects on trimer formation. The strongest effect was observed for the E226Q/E105Q mutant, which almost fully abolished trimer formation without preventing membrane interaction. These results indicate that lipids engage in specific, trimer-stabilizing interactions that go beyond simply providing a concentration-enhancing surface. The finding that protein-membrane interactions are just as important as protein-protein interactions in ANXB12 trimer formation raises the possibility that the formation of specific lipid contacts could be a more widely used driving force for membrane-mediated oligomerization of proteins in general.


Asunto(s)
Anexinas/química , Anexinas/metabolismo , Membrana Celular/metabolismo , Membrana Dobles de Lípidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Anexinas/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/química , Lisina/genética , Lisina/metabolismo , Mutación , Conformación Proteica , Multimerización de Proteína
12.
J Phys Chem B ; 122(41): 9507-9515, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30252478

RESUMEN

Intrinsically disordered protein domains not only are found in soluble proteins but also can be part of large protein complexes or protein aggregates. For example, several amyloid fibrils have intrinsically disordered domains framing a rigid ß-sheet-rich core. These disordered domains can often be observed using solution NMR methods in combination with modest magic angle spinning and without perdeuteration. But how can these regions be detected using solution NMR methods when they are part of a fibril that is not tumbling isotropically in solution? Here we addressed this question by investigating the dynamic C-terminus of huntingtin exon-1 (HTTex1) fibrils that are important in Huntington's disease. We assigned the most dynamic regions of the C-terminus of three HTTex1 variants. On the basis of this assignment, we measured site-specific secondary chemical shifts, peak intensities, and R1, R'2, and R1ρ 15N relaxation rates. In addition, we determined the residual 1H-15N dipolar couplings of this region. Our results show that the dipolar couplings are averaged to a very high degree, resulting in an order parameter that is essentially zero. Together, our data show that the C-terminus of HTTex1 is intrinsically disordered and undergoes motions in the high picosecond to low nanosecond range.


Asunto(s)
Amiloide/química , Proteína Huntingtina/química , Amiloide/genética , Exones , Proteína Huntingtina/genética , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Mutación , Isótopos de Nitrógeno , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Dominios Proteicos , Multimerización de Proteína , Espectroscopía de Protones por Resonancia Magnética
13.
Sci Rep ; 7(1): 7802, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798389

RESUMEN

Mitochondrial-derived peptides (MDPs) and their analogs have emerged as wide-spectrum, stress response factors protective in amyloid disease models. MDP cytoprotective functions are generally attributed to anti-apoptotic activity, however, little is known about their capacity to facilitate the cell's unfolded protein response via direct interactions with amyloidogenic proteins. Here, we explored the effects of the MDP-analog, humaninS14G (HNG), and the MDP, small humanin-like peptide 2 (SHLP2), on the misfolding of islet amyloid polypeptide (IAPP), a critical pathogenic step in type 2 diabetes mellitus (T2DM). Our thioflavin T fluorescence studies show that HNG inhibits IAPP misfolding at highly substoichiometric concentrations. Seeded fluorescence and co-sedimentation studies demonstrate MDPs block amyloid seeding and directly bind misfolded, seeding-capable IAPP species. Furthermore, our electron paramagnetic resonance spectroscopy and circular dichroism data indicate MDPs do not act by binding IAPP monomers. Taken together our results reveal a novel chaperone-like activity wherein these MDPs specifically target misfolded amyloid seeds to inhibit IAPP misfolding which, along with direct anti-apoptotic activity and beneficial metabolic effects, make HNG and SHLP2 exciting prospects as T2DM therapeutics. These data also suggest that other mitochondrial stress response factors within the MDP family may be amenable to development into therapeutics for protein-misfolding diseases.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/química , Mitocondrias/química , Dicroismo Circular , Diabetes Mellitus Tipo 2/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Microscopía Electrónica de Transmisión , Unión Proteica , Pliegue de Proteína/efectos de los fármacos
14.
Sci Rep ; 6: 31094, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27531121

RESUMEN

The current diabetes epidemic is associated with a diverse set of risk factors including obesity and exposure to plastics. Notably, significant elevations of negatively charged amphiphilic molecules are observed in obesity (e.g. free fatty acids and phosphatidic acid) and plastics exposure (monophthalate esters). It remains unclear whether these factors share pathogenic mechanisms and whether links exist with islet amyloid polypeptide (IAPP) misfolding, a process central to ß-cell dysfunction and death. Using a combination of fluorescence, circular dichroism and electron microscopy, we show that phosphatidic acid, oleic acid, and the phthalate metabolite MBzP partition into neutral membranes and enhance IAPP misfolding. The elevation of negative charge density caused by the presence of the risk factor molecules stabilizes a common membrane-bound α-helical intermediate that, in turn, facilitates IAPP misfolding. This shared mechanism points to a critical role for the membrane-bound intermediate in disease pathogenesis, making it a potential target for therapeutic intervention.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Membranas/metabolismo , Obesidad/complicaciones , Pliegue de Proteína , Dicroismo Circular , Humanos , Membranas/química , Microscopía Electrónica , Ácido Oléico/metabolismo , Imagen Óptica , Ácidos Fosfatidicos/metabolismo , Ácidos Ftálicos/metabolismo
15.
Structure ; 23(5): 873-881, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25865245

RESUMEN

BAR proteins are involved in a variety of membrane remodeling events but how they can mold membranes into different shapes remains poorly understood. Using electron paramagnetic resonance, we find that vesicle binding of the N-BAR protein amphiphysin is predominantly mediated by the shallow insertion of amphipathic N-terminal helices. In contrast, the interaction with tubes involves deeply inserted N-terminal helices together with the concave surface of the BAR domain, which acts as a scaffold. Combined with the observed concentration dependence of tubulation and BAR domain scaffolding, the data indicate that initial membrane deformations and vesicle binding are mediated by insertion of amphipathic helical wedges, while tubulation requires high protein densities at which oligomeric BAR domain scaffolds form. In addition, we identify a pocket of residues on the concave surface of the BAR domain that insert deeply into tube membrane. Interestingly, this pocket harbors a number of disease mutants in the homologous amphiphysin 2.


Asunto(s)
Drosophila/metabolismo , Proteínas Ligadas a Lípidos/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Animales , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Cristalografía por Rayos X , Drosophila/química , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Modelos Biológicos , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína
16.
PLoS One ; 9(12): e112262, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25464275

RESUMEN

BACKGROUND: In Huntington's disease, expansion of a CAG triplet repeat occurs in exon 1 of the huntingtin gene (HTT), resulting in a protein bearing>35 polyglutamine residues whose N-terminal fragments display a high propensity to misfold and aggregate. Recent data demonstrate that polyglutamine expansion results in conformational changes in the huntingtin protein (HTT), which likely influence its biological and biophysical properties. Developing assays to characterize and measure these conformational changes in isolated proteins and biological samples would advance the testing of novel therapeutic approaches aimed at correcting mutant HTT misfolding. Time-resolved Förster energy transfer (TR-FRET)-based assays represent high-throughput, homogeneous, sensitive immunoassays widely employed for the quantification of proteins of interest. TR-FRET is extremely sensitive to small distances and can therefore provide conformational information based on detection of exposure and relative position of epitopes present on the target protein as recognized by selective antibodies. We have previously reported TR-FRET assays to quantify HTT proteins based on the use of antibodies specific for different amino-terminal HTT epitopes. Here, we investigate the possibility of interrogating HTT protein conformation using these assays. METHODOLOGY/PRINCIPAL FINDINGS: By performing TR-FRET measurements on the same samples (purified recombinant proteins or lysates from cells expressing HTT fragments or full length protein) at different temperatures, we have discovered a temperature-dependent, reversible, polyglutamine-dependent conformational change of wild type and expanded mutant HTT proteins. Circular dichroism spectroscopy confirms the temperature and polyglutamine-dependent change in HTT structure, revealing an effect of polyglutamine length and of temperature on the alpha-helical content of the protein. CONCLUSIONS/SIGNIFICANCE: The temperature- and polyglutamine-dependent effects observed with TR-FRET on HTT proteins represent a simple, scalable, quantitative and sensitive assay to identify genetic and pharmacological modulators of mutant HTT conformation, and potentially to assess the relevance of conformational changes during onset and progression of Huntington's disease.


Asunto(s)
Proteínas Mutantes/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Péptidos/química , Aminoácidos/química , Dicroismo Circular , Progresión de la Enfermedad , Epítopos/química , Exones , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Proteína Huntingtina , Inmunoensayo , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Temperatura , Tiorredoxinas/química
17.
PLoS One ; 7(8): e43519, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22927980

RESUMEN

Mucosotropic, high-risk human papillomaviruses (HPV) are sexually transmitted viruses that are causally associated with the development of cervical cancer. The most common high-risk genotype, HPV16, is an obligatory intracellular virus that must gain entry into host epithelial cells and deliver its double stranded DNA to the nucleus. HPV capsid proteins play a vital role in these steps. Despite the critical nature of these capsid protein-host cell interactions, the precise cellular components necessary for HPV16 infection of epithelial cells remains unknown. Several neutralizing epitopes have been identified for the HPV16 L2 minor capsid protein that can inhibit infection after initial attachment of the virus to the cell surface, which suggests an L2-specific secondary receptor or cofactor is required for infection, but so far no specific L2-receptor has been identified. Here, we demonstrate that the annexin A2 heterotetramer (A2t) contributes to HPV16 infection and co-immunoprecipitates with HPV16 particles on the surface of epithelial cells in an L2-dependent manner. Inhibiting A2t with an endogenous annexin A2 ligand, secretory leukocyte protease inhibitor (SLPI), or with an annexin A2 antibody significantly reduces HPV16 infection. With electron paramagnetic resonance, we demonstrate that a previously identified neutralizing epitope of L2 (aa 108-120) specifically interacts with the S100A10 subunit of A2t. Additionally, mutation of this L2 region significantly reduces binding to A2t and HPV16 pseudovirus infection. Furthermore, downregulation of A2t with shRNA significantly decreases capsid internalization and infection by HPV16. Taken together, these findings indicate that A2t contributes to HPV16 internalization and infection of epithelial cells and this interaction is dependent on the presence of the L2 minor capsid protein.


Asunto(s)
Anexina A2/química , Anexina A2/metabolismo , Proteínas de la Cápside/metabolismo , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/metabolismo , Multimerización de Proteína , Proteínas S100/química , Proteínas S100/metabolismo , Secuencia de Aminoácidos , Anexina A2/genética , Anexina A2/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Células Epiteliales/virología , Epítopos/química , Epítopos/inmunología , Técnicas de Silenciamiento del Gen , Células HeLa , Papillomavirus Humano 16/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Estructura Cuaternaria de Proteína , ARN Interferente Pequeño/genética , Receptores de Superficie Celular , Proteínas S100/genética , Proteínas S100/inmunología , Especificidad por Sustrato
18.
Invest Ophthalmol Vis Sci ; 51(3): 1304-10, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19892876

RESUMEN

PURPOSE: Drusen are a hallmark of eyes affected by age-related macular degeneration. In previous study, a conformational-specific antibody showed drusen to contain nonfibrillar amyloid structures. The current study was undertaken to assess the presence of additional amyloid structures in drusen. METHODS: Sections from human donor eyes were reacted with M204, a monoclonal antibody that recognizes nonfibrillar oligomers; OC, a polyclonal antibody that recognizes amyloid fibrils of various molecular weights; and WO1 and WO2, monoclonal antibodies that are specifically reactive to mature amyloid fibrils. Electron microscopy was used as an independent means of investigating the presence of amyloid fibrils in drusen. RESULTS: The presence of nonfibrillar oligomers was verified using the M204 antibody. OC and WO antibodies stained a wide spectrum of vesicular structures. OC reactivity showed extensive overlap with Abeta immunoreactivity, whereas a partial overlap was seen between Abeta reactivity and that of the WO antibodies. The presence of amyloid fibrils was also visualized by electron microscopy. CONCLUSIONS: These data reveal the presence of a wide spectrum of amyloid structures in drusen. The results are significant, given that specific conformational forms of amyloid are known to be pathogenic in a variety of neurodegenerative diseases. Deposition of these structures may lead to local toxicity of the retinal pigmented epithelium or induction of local inflammatory events that contribute to drusen biogenesis and the pathogenesis of AMD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Drusas Retinianas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/ultraestructura , Anticuerpos Monoclonales , Humanos , Degeneración Macular/patología , Microscopía Confocal , Microscopía Inmunoelectrónica , Epitelio Pigmentado Ocular/ultraestructura , Priones/metabolismo , Drusas Retinianas/patología , Donantes de Tejidos , alfa-Sinucleína/metabolismo
19.
Mol Neurodegener ; 3: 16, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18939994

RESUMEN

BACKGROUND: The multifunctional protein vitronectin is present within the deposits associated with Alzheimer disease (AD), age-related macular degeneration (AMD), atherosclerosis, systemic amyloidoses, and glomerulonephritis. The extent to which vitronectin contributes to amyloid formation within these plaques, which contain misfolded, amyloidogenic proteins, and the role of vitronectin in the pathophysiology of the aforementioned diseases is currently unknown. The investigation of vitronectin aggregation is significant since the formation of oligomeric and fibrillar structures are common features of amyloid proteins. RESULTS: We observed vitronectin immunoreactivity in senile plaques of AD brain, which exhibited overlap with the amyloid fibril-specific OC antibody, suggesting that vitronectin is deposited at sites of amyloid formation. Of particular interest is the growing body of evidence indicating that soluble nonfibrillar oligomers may be responsible for the development and progression of amyloid diseases. In this study we demonstrate that both plasma-purified and recombinant human vitronectin readily form spherical oligomers and typical amyloid fibrils. Vitronectin oligomers are toxic to cultured neuroblastoma and retinal pigment epithelium (RPE) cells, possibly via a membrane-dependent mechanism, as they cause leakage of synthetic vesicles. Oligomer toxicity was attenuated in RPE cells by the anti-oligomer A11 antibody. Vitronectin fibrils contain a C-terminal protease-resistant fragment, which may approximate the core region of residues essential to amyloid formation. CONCLUSION: These data reveal the propensity of vitronectin to behave as an amyloid protein and put forth the possibilities that accumulation of misfolded vitronectin may contribute to aggregate formation seen in age-related amyloid diseases.

20.
Biochemistry ; 45(3): 934-42, 2006 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-16411769

RESUMEN

Annexins are soluble proteins that can interact with membranes in a Ca2+-dependent manner. Recent studies have shown that they can also undergo Ca2+-independent membrane interactions that are modulated by pH and phospholipid composition. Here, we investigated the structural changes that occurred during Ca2+-independent interaction of annexin B12 with phospholipid vesicles as a function of pH. Electron paramagnetic resonance analysis of a helical hairpin encompassing the D and E helices in the second repeat of the protein showed that this region refolded and formed a continuous amphipathic alpha helix following Ca2+-independent binding to membranes at mildly acidic pH. At pH 4.0, this helix assumed a transmembrane topography, but at pH approximately 5.0-5.5, it was peripheral and approximately parallel to the membrane. The peripheral form was reversibly converted into the transmembrane form by lowering the pH and vice versa. Furthermore, analysis of vesicles incubated with annexin B12 using freeze-fracture electron microscopy methods showed classical intramembrane particles at pH 4.0 but none at pH 5.3. Together, these data raise the possibility that the peripheral-bound form of annexin B12 could act as a kinetic intermediate in the formation of the transmembrane form of the protein.


Asunto(s)
Anexinas/química , Anexinas/metabolismo , Membrana Celular/metabolismo , Anexinas/genética , Calcio/metabolismo , Técnica de Fractura por Congelación , Concentración de Iones de Hidrógeno , Modelos Moleculares , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA