Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
AAPS PharmSciTech ; 25(5): 127, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844724

RESUMEN

The success of obtaining solid dispersions for solubility improvement invariably depends on the miscibility of the drug and polymeric carriers. This study aimed to categorize and select polymeric carriers via the classical group contribution method using the multivariate analysis of the calculated solubility parameter of RX-HCl. The total, partial, and derivate parameters for RX-HCl were calculated. The data were compared with the results of excipients (N = 36), and a hierarchical clustering analysis was further performed. Solid dispersions of selected polymers in different drug loads were produced using solvent casting and characterized via X-ray diffraction, infrared spectroscopy and scanning electron microscopy. RX-HCl presented a Hansen solubility parameter (HSP) of 23.52 MPa1/2. The exploratory analysis of HSP and relative energy difference (RED) elicited a classification for miscible (n = 11), partially miscible (n = 15), and immiscible (n = 10) combinations. The experimental validation followed by a principal component regression exhibited a significant correlation between the crystallinity reduction and calculated parameters, whereas the spectroscopic evaluation highlighted the hydrogen-bonding contribution towards amorphization. The systematic approach presented a high discrimination ability, contributing to optimal excipient selection for the obtention of solid solutions of RX-HCl.


Asunto(s)
Química Farmacéutica , Excipientes , Polímeros , Clorhidrato de Raloxifeno , Solubilidad , Difracción de Rayos X , Polímeros/química , Excipientes/química , Clorhidrato de Raloxifeno/química , Análisis Multivariante , Difracción de Rayos X/métodos , Química Farmacéutica/métodos , Portadores de Fármacos/química , Composición de Medicamentos/métodos , Microscopía Electrónica de Rastreo/métodos , Enlace de Hidrógeno , Cristalización/métodos
2.
J Neurochem ; 166(1): 76-86, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36621842

RESUMEN

Aggregation of the microtubule-associated protein tau is implicated in several neurodegenerative tauopathies including Alzheimer's disease (AD). Recent studies evidenced tau liquid-liquid phase separation (LLPS) into droplets as an early event in tau pathogenesis with the potential to enhance aggregation. Tauopathies like AD are accompanied by sustained neuroinflammation and the release of alarmins at early stages of inflammatory responses encompass protective functions. The Ca2+ -binding S100B protein is an alarmin augmented in AD that was recently implicated as a proteostasis regulator acting as a chaperone-type protein, inhibiting aggregation and toxicity through interactions of amyloidogenic clients with a regulatory surface exposed upon Ca2+ -binding. Here we expand the regulatory functions of S100B over protein condensation phenomena by reporting its Ca2+ -dependent activity as a modulator of tau LLPS induced by crowding agents (PEG) and metal ions (Zn2+ ). We observe that apo S100B has a negligible effect on PEG-induced tau demixing but that Ca2+ -bound S100B prevents demixing, resulting in a shift of the phase diagram boundary to higher crowding concentrations. Also, while incubation with apo S100B does not compromise tau LLPS, addition of Ca2+ results in a sharp decrease in turbidity, indicating that interactions with S100B-Ca2+ promote transition of tau to the mixed phase. Further, electrophoretic analysis and FLIM-FRET studies revealed that S100B incorporates into tau liquid droplets, suggesting an important stabilizing and chaperoning role contributing to minimize toxic tau aggregates. Resorting to Alexa488-labeled tau we observed that S100B-Ca2+ reduces the formation of tau fluorescent droplets, without compromising liquid-like behavior and droplet fusion events. The Zn2+ -binding properties of S100B also contribute to regulate Zn2+ -promoted tau LLPS as droplets are decreased by Zn2+ buffering by S100B, in addition to the Ca2+ -triggered interactions with tau. Altogether this work uncovers the versatility of S100B as a proteostasis regulator acting on protein condensation phenomena of relevance across the neurodegeneration continuum.


Asunto(s)
Enfermedad de Alzheimer , Proteínas tau , Humanos , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100
3.
J Mol Biol ; 434(19): 167791, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35970403

RESUMEN

Alzheimer's disease (AD) hallmarks include the aggregation of amyloid-ß (Aß), tau and neuroinflammation promoted by several alarmins. Among these is S100B, a small astrocytic homodimeric protein, upregulated in AD, whose multiple biological activities depend on localization, concentration, and assembly state. S100B was reported to inhibit the aggregation and toxicity of Aß42 and tau similarly to a holdase-type chaperone. This activity is dependent of Ca2+-binding, which triggers the exposure of a regulatory binding cleft at the S100B dimer interface with which amyloidogenic clients dynamically interact. Although the dimer prevails, a significant portion of secreted S100B in the human brain occurs as higher order multimers, whose protective functions remain uncharacterized and which we here investigate. Resorting to ThT-monitored aggregation kinetics, we determined that unlike the dimer, tetrameric S100B inhibits Aß42 aggregation at sub/equimolar ratios, an effect that persists in the absence of Ca2+ binding. Structural analysis revealed that S100B tetramerization spawns a novel extended cleft accommodating an aggregation-prone surface that mediates interactions with monomeric Aß client via hydrophobic interactions, as corroborated by Bis-ANS fluorescence and docking analysis. Correspondingly, at high ionic strength that reduces solvation and favours hydrophobic contacts, the inhibition of Aß42 aggregation by tetrameric S100B is 3-fold increased. Interestingly, this extended Ca2+-independent surface favours Aß42 as substrate, as tau K18 aggregation is not inhibited by the apo tetramer. Overall, results illustrate a mechanism through which oligomerization of the S100B chaperone fine-tunes anti-aggregation activity and client specificity, highlighting the potential functional relevance of S100B multimers in the regulation of AD proteotoxicity.


Asunto(s)
Enfermedad de Alzheimer , Calcio , Chaperonas Moleculares , Agregación Patológica de Proteínas , Subunidad beta de la Proteína de Unión al Calcio S100 , Alarminas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , Calcio/metabolismo , Humanos , Chaperonas Moleculares/química , Agregación Patológica de Proteínas/metabolismo , Conformación Proteica , Multimerización de Proteína , Subunidad beta de la Proteína de Unión al Calcio S100/química
4.
Nat Commun ; 12(1): 6292, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725360

RESUMEN

The microtubule-associated protein tau is implicated in the formation of oligomers and fibrillar aggregates that evade proteostasis control and spread from cell-to-cell. Tau pathology is accompanied by sustained neuroinflammation and, while the release of alarmin mediators aggravates disease at late stages, early inflammatory responses encompass protective functions. This is the case of the Ca2+-binding S100B protein, an astrocytic alarmin which is augmented in AD and which has been recently implicated as a proteostasis regulator, acting over amyloid ß aggregation. Here we report the activity of S100B as a suppressor of tau aggregation and seeding, operating at sub-stoichiometric conditions. We show that S100B interacts with tau in living cells even in microtubule-destabilizing conditions. Structural analysis revealed that tau undergoes dynamic interactions with S100B, in a Ca2+-dependent manner, notably with the aggregation prone repeat segments at the microtubule binding regions. This interaction involves contacts of tau with a cleft formed at the interface of the S100B dimer. Kinetic and mechanistic analysis revealed that S100B inhibits the aggregation of both full-length tau and of the microtubule binding domain, and that this proceeds through effects over primary and secondary nucleation, as confirmed by seeding assays and direct observation of S100B binding to tau oligomers and fibrils. In agreement with a role as an extracellular chaperone and its accumulation near tau positive inclusions, we show that S100B blocks proteopathic tau seeding. Together, our findings establish tau as a client of the S100B chaperone, providing evidence for neuro-protective functions of this inflammatory mediator across different tauopathies.


Asunto(s)
Chaperonas Moleculares/metabolismo , Enfermedades Neurodegenerativas/prevención & control , Agregación Patológica de Proteínas/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Proteínas tau/metabolismo , Fenómenos Biofísicos , Línea Celular , Humanos , Cinética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Resonancia Magnética Nuclear Biomolecular/métodos , Unión Proteica , Elementos Estructurales de las Proteínas
5.
Chem Commun (Camb) ; 57(3): 379-382, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33326534

RESUMEN

S100B is an extracellular protein implicated in Alzheimer's Disease and a suppressor of amyloid-ß aggregation. Herein we report a mechanism tying Cu2+ binding to a change in assembly state yielding disulfide cross-linked oligomers with higher anti-aggregation activity. This chemical control of chaperone function illustrates a regulatory process relevant under metal and proteostasis dysfunction as in neurodegeneration.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Cobre/farmacología , Reactivos de Enlaces Cruzados/farmacología , Disulfuros/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Subunidad beta de la Proteína de Unión al Calcio S100/química , Péptidos beta-Amiloides/metabolismo , Sitios de Unión , Cobre/química , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/química , Disulfuros/química , Humanos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Polimerizacion , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/metabolismo
6.
Int J Mol Sci ; 20(23)2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31783644

RESUMEN

Metal ions are well known modulators of protein aggregation and are key players in Alzheimer's Disease, being found to be associated to pathologic protein deposits in diseased brains. Therefore, understanding how metals influence amyloid aggregation is critical in establishing molecular mechanisms that underlie disease onset and progression. Here, we report data on the interaction of full-length human Tau protein with calcium and zinc ions, evidencing that Tau self-assembly is differently regulated, depending on the type of bound metal ion. We established that Tau binds 4 Zn2+ and 1 Ca2+ per monomer while using native mass spectrometry analysis, without inducing order or substantial conformational changes in the intrinsically disordered Tau, as determined by structural analysis using circular dichroism and Attenuated Total Reflectance-Fourier Transform Infrared (ATR-FTIR) spectroscopies. However, Tau aggregation is found to proceed differently in the calcium- and -zinc bound forms. While the rate of aggregation, as determined from thioflavin-T (ThT) fluorescence kinetics, is highly increased in both cases, the reaction proceeds via different mechanisms, as evidenced by the absence of the lag phase in the reaction of zinc-bound Tau. Monitoring Tau aggregation using native mass spectrometry indeed evidenced a distinct distribution of Tau conformers along the reaction, as confirmed by dynamic light scattering analysis. We propose that such differences arise from zinc binding at distinct locations within the Tau sequence that prompt both the rapid formation of seeding oligomers through interactions at high affinity sites within the repeat domains, as well as amorphous aggregation, through low affinity interactions with residues elsewhere in the sequence, including at the fuzzy coat domain.


Asunto(s)
Agregado de Proteínas/fisiología , Zinc/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Benzotiazoles/metabolismo , Calcio/metabolismo , Dicroismo Circular , Humanos , Cinética , Conformación Proteica , Espectroscopía Infrarroja por Transformada de Fourier/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA