Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 263
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 9168, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649777

RESUMEN

Fluorinated graphene, a two-dimensional nanomaterial composed of three atomic layers, a central carbon layer sandwiched between two layers of fluorine atoms, has attracted considerable attention across various fields, particularly for its potential use in biomedical applications. Nonetheless, scant effort has been devoted to assessing the potential toxicological implications of this nanomaterial. In this study, we scrutinize the potential impact of fluorinated graphene on a protein model, HP35 by utilizing extensive molecular dynamics (MD) simulation methods. Our MD results elucidate that upon adsorption to the nanomaterial, HP35 undergoes a denaturation process initiated by the unraveling of the second helix of the protein and the loss of the proteins hydrophobic core. In detail, substantial alterations in various structural features of HP35 ensue, including alterations in hydrogen bonding, Q value, and RMSD. Subsequent analyses underscore that hydrophobic and van der Waals interactions (predominant), alongside electrostatic energy (subordinate), exert influence over the adsorption of HP35 on the fluorinated graphene surface. Mechanistic scrutiny attests that the unrestrained lateral mobility of HP35 on the fluorinated graphene nanomaterial primarily causes the exposure of HP35's hydrophobic core, resulting in the eventual structural denaturation of HP35. A trend in the features of 2D nanostructures is proposed that may facilitate the denaturation process. Our findings not only substantiate the potential toxicity of fluorinated graphene but also unveil the underlying molecular mechanism, which thereby holds significance for the prospective utilization of such nanomaterials in the field of biomedicine.


Asunto(s)
Grafito , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Proteínas de Neurofilamentos , Fragmentos de Péptidos , Conformación Proteica en Hélice alfa , Grafito/química , Grafito/toxicidad , Interacciones Hidrofóbicas e Hidrofílicas , Desplegamiento Proteico/efectos de los fármacos , Halogenación , Adsorción , Nanoestructuras/química , Nanoestructuras/toxicidad
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 316: 124332, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38676982

RESUMEN

Studies on the interactions between ligands and proteins provide insights into how a possible medication alters the structures and activities of the target or carrier proteins. The natural flavonoid aglycone Chrysin (CHR) has demonstrated anti-inflammatory, antioxidant, antiapoptotic, neuroprotective, and antineoplastic effects, both in vitro and in vivo. In this work, we investigated the impact of CHR binding on the as-yet-unexplored conformation, dynamics, and unfolding mechanism of human serum albumin (HSA). We determined CHR binding to HSA domain-II with the association constant (Ka) of 2.70 ± 0.21 × 105 M-1. The urea-induced sequential unfolding mechanism of HSA was used to elucidate the debatable binding location of CHR. CHR binding induced both secondary and tertiary structural alterations in the protein as studied by far-UV circular dichroism and intrinsic fluorescence spectroscopy. Red edge excitation shift (REES) indicated a decrease in conformational dynamics of the protein on the complex formation. This suggested an ordered compact and spatial arrangement of the CHR-boundmolecule. The binding of CHR was found to significantly modulate the urea-induced unfolding pathway of HSA. Urea-induced unfolding pathway of HSA became a two-state process (N-U) from a three-state process (N-I-U). The interaction of CHR is found to increase the thermal stability of the protein by ∼4 °C. This study focuses on the fundamental sciences and demonstrates how prospective medication compounds can alter the dynamics and stability of protein structure.


Asunto(s)
Flavonoides , Unión Proteica , Desplegamiento Proteico , Albúmina Sérica Humana , Humanos , Flavonoides/química , Flavonoides/farmacología , Flavonoides/metabolismo , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo , Desplegamiento Proteico/efectos de los fármacos , Urea/farmacología , Urea/química , Dicroismo Circular , Espectrometría de Fluorescencia , Conformación Proteica
3.
Mol Pharmacol ; 101(2): 95-105, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34866045

RESUMEN

Folding-deficient mutants of solute carrier 6 (SLC6) family members have been linked to human diseases. The serotonin transporter [(SERT)/SLC6A4] is an important drug target in the treatment of depression, anxiety, and obsessive-compulsive disorders and-with structural information in several conformational states-one of the best understood transporters. Here, we surmised that thermal unfolding offered a glimpse on the folding energy landscape of SLC6 transporters. We carried out molecular dynamic (MD) simulations to understand the mechanistic basis for enhanced and reduced stability, respectively, of the thermostabilized variant SERT-Y110A/I291A/T439S, which had previously been used for crystallization of human SERT in the outward-facing state, and of the folding-deficient SERT-P601A/G602A. We also examined the hydrophobic mismatch caused by the absence of cholesterol to explore the contribution of cholesterol to protein stability. When compared with wild type SERT, the thermodynamic and kinetic stability of SERT-Y110A/I291A/T439S was enhanced. In the other instances, changes in these two components were not correlated: the mutations in SERT-P601A/G602A led to a drop in thermodynamic but an increase in kinetic stability. The divergence was even more pronounced after cholesterol depletion, which reduced thermodynamic stability but increased the kinetic stability of wild type SERT to a level comparable to that of SERT-Y110A/I291A/T439S. We conclude that the low cholesterol content of the endoplasmic reticulum facilitates progression of the folding trajectory by reducing the energy difference between folding intermediates and the native state. SIGNIFICANCE STATEMENT: Point mutations in solute carrier 6 (SLC6) family members cause folding diseases. The serotonin transporter [(SERT)/SLC6A4] is a target for antidepressants and the best understood SLC6. This study produced molecular dynamics simulations and examined thermal unfolding of wild type and mutant SERT variants to understand their folding energy landscape. In the folding-deficient SERT-P012A/G602A, changes in kinetic and thermodynamic stability were not correlated. Similarly, cholesterol depletion lowered thermodynamic but enhanced kinetic stability. These observations allow for rationalizing the action of pharmacochaperones.


Asunto(s)
Colesterol/metabolismo , Mutación/genética , Desplegamiento Proteico/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Termodinámica , Antidepresivos/metabolismo , Antidepresivos/farmacología , Variación Genética/genética , Células HEK293 , Humanos , Cinética , Simulación de Dinámica Molecular , Unión Proteica/fisiología , Estabilidad Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Proteínas de Transporte de Serotonina en la Membrana Plasmática/química
4.
J Am Chem Soc ; 143(45): 19137-19148, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34739240

RESUMEN

The assembly and maturation of human immunodeficiency virus type 1 (HIV-1) require proteolytic cleavage of the Gag polyprotein. The rate-limiting step resides at the junction between the capsid protein CA and spacer peptide 1, which assembles as a six-helix bundle (6HB). Bevirimat (BVM), the first-in-class maturation inhibitor drug, targets the 6HB and impedes proteolytic cleavage, yet the molecular mechanisms of its activity, and relatedly, the escape mechanisms of mutant viruses, remain unclear. Here, we employed extensive molecular dynamics (MD) simulations and free energy calculations to quantitatively investigate molecular structure-activity relationships, comparing wild-type and mutant viruses in the presence and absence of BVM and inositol hexakisphosphate (IP6), an assembly cofactor. Our analysis shows that the efficacy of BVM is directly correlated with preservation of 6-fold symmetry in the 6HB, which exists as an ensemble of structural states. We identified two primary escape mechanisms, and both lead to loss of symmetry, thereby facilitating helix uncoiling to aid access of protease. Our findings also highlight specific interactions that can be targeted for improved inhibitor activity and support the use of MD simulations for future inhibitor design.


Asunto(s)
Fármacos Anti-VIH/metabolismo , VIH-1/química , Succinatos/metabolismo , Triterpenos/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Simulación de Dinámica Molecular , Mutación , Ácido Fítico/metabolismo , Conformación Proteica en Hélice alfa/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
5.
Curr Top Med Chem ; 21(31): 2839-2850, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34749611

RESUMEN

BACKGROUND: Sphingosine kinase 1 (SPhK1) is a crucial signaling enzyme involved in cell proliferation, cellular survival, stimulation of angiogenesis, and apoptosis prevention. Recently, we have reported the unfolding kinetics of SPhK1 using molecular dynamics (MD) simulation, circular dichroism, and fluorescence spectroscopy. We found that SPhK1 showed a biphasic unfolding with an intermediate state (~ 4.0 M urea). OBJECTIVE: We aim to understand the impact of MD simulation duration on the structure, function, and dynamics of proteins. In order to get deeper insights into the folding mechanism, an extended MD simulation is required. METHODS: Here, we extended the MD simulations time scale from 100 to 300 ns on SPhK1 at increasing urea concentration to explore structural changes in the SPhK1. RESULTS: The results suggested a constant form of the unfolding of SPhK1 upon extending the simulation time scale at different urea concentrations. Furthermore, we showed step by step unfolding and percentage of secondary structure contents in SPhK1 under the influence of urea at each concentration. CONCLUSION: The results from the current work revealed a uniform pattern of the SPhK1 unfolding at different urea concentrations. This study provides deeper mechanistic insights into the urea-induced denaturation of SPhK1.


Asunto(s)
Simulación de Dinámica Molecular , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Desnaturalización Proteica , Desplegamiento Proteico , Urea , Dicroismo Circular , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Conformación Proteica , Desnaturalización Proteica/efectos de los fármacos , Pliegue de Proteína , Desplegamiento Proteico/efectos de los fármacos , Espectrometría de Fluorescencia , Urea/farmacología
6.
Nat Chem Biol ; 17(11): 1148-1156, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34556859

RESUMEN

The unfolded protein response (UPR) homeostatically matches endoplasmic reticulum (ER) protein-folding capacity to cellular secretory needs. However, under high or chronic ER stress, the UPR triggers apoptosis. This cell fate dichotomy is promoted by differential activation of the ER transmembrane kinase/endoribonuclease (RNase) IRE1α. We previously found that the RNase of IRE1α can be either fully activated or inactivated by ATP-competitive kinase inhibitors. Here we developed kinase inhibitors, partial antagonists of IRE1α RNase (PAIRs), that partially antagonize the IRE1α RNase at full occupancy. Biochemical and structural studies show that PAIRs promote partial RNase antagonism by intermediately displacing the helix αC in the IRE1α kinase domain. In insulin-producing ß-cells, PAIRs permit adaptive splicing of Xbp1 mRNA while quelling destructive ER mRNA endonucleolytic decay and apoptosis. By preserving Xbp1 mRNA splicing, PAIRs allow B cells to differentiate into immunoglobulin-producing plasma cells. Thus, an intermediate RNase-inhibitory 'sweet spot', achieved by PAIR-bound IRE1α, captures a desirable conformation for drugging this master UPR sensor/effector.


Asunto(s)
Adenosina Trifosfato/farmacología , Endorribonucleasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/química , Endorribonucleasas/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Desplegamiento Proteico/efectos de los fármacos
7.
Int J Mol Sci ; 22(7)2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808390

RESUMEN

When combined with NMR spectroscopy, high hydrostatic pressure is an alternative perturbation method used to destabilize globular proteins that has proven to be particularly well suited for exploring the unfolding energy landscape of small single-domain proteins. To date, investigations of the unfolding landscape of all-ß or mixed-α/ß protein scaffolds are well documented, whereas such data are lacking for all-α protein domains. Here we report the NMR study of the unfolding pathways of GIPC1-GH2, a small α-helical bundle domain made of four antiparallel α-helices. High-pressure perturbation was combined with NMR spectroscopy to unravel the unfolding landscape at three different temperatures. The results were compared to those obtained from classical chemical denaturation. Whatever the perturbation used, the loss of secondary and tertiary contacts within the protein scaffold is almost simultaneous. The unfolding transition appeared very cooperative when using high pressure at high temperature, as was the case for chemical denaturation, whereas it was found more progressive at low temperature, suggesting the existence of a complex folding pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Desplegamiento Proteico/efectos de los fármacos , Humanos , Cinética , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Conformación Proteica en Hélice alfa/fisiología , Desnaturalización Proteica , Dominios Proteicos , Temperatura , Termodinámica
8.
In Vitro Cell Dev Biol Anim ; 57(3): 359-371, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33559028

RESUMEN

We previously described a non-monotonic dose response curve at low copper concentrations where 3.125 µM CuSO4 (the early inflection point) was more toxic than 25 µM CuSO4 in Caco-2 cells. We employed global proteomics to investigate this observation. The altered expression levels of a small number of proteins displaying a temporal response may provide the best indication of the underlying mechanism; more well-known copper response proteins including the metal binding metallothioneins (MT1X, MT1F, MT2A) and antioxidant response proteins including Heme oxygenase were upregulated to a similar level in both copper concentrations and so are less likely to underpin this phenomenon.The temporal response proteins include Granulins, AN1-type zinc finger protein 2A (ZFAND2A), and the heat shock proteins (HSPA6 and HSPA1B). Granulins were decreased after 4 h only in 25 µM CuSO4 but from 24 h, were decreased in both copper concentrations to a similar level. Induction of ZFAND2A and increases in HSPA6 and HSPA1B were observed at 24 h only in 25 µM CuSO4 but were present at 48 h in both copper conditions. The early expression of ZFAND2A, HSPs, and higher levels of α-crystallin B (CRYAB) correlated with lower levels of misfolded proteins in 25 µM CuSO4 compared to 3.125 µM CuSO4 at 48 h. These results suggest that 3.125 µM CuSO4 at early time points was unable to activate the plethora of stress responses invoked by the higher copper concentration, paradoxically exposing the Caco-2 cells to higher levels of misfolded proteins and greater proteotoxic stress.


Asunto(s)
Cobre/toxicidad , Intestinos/patología , Células CACO-2 , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Glutatión/metabolismo , Humanos , Desplegamiento Proteico/efectos de los fármacos , Proteómica , Reproducibilidad de los Resultados , Factores de Tiempo
9.
Int J Biol Macromol ; 174: 153-161, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33484803

RESUMEN

Heavy metal based toxicity has a direct relation with the perturbation of protein structure. We have investigated the progressive unfolding of ovalbumin, in the presence of increasing concentration mercury (0-6.25 µM) using different spectroscopic techniques. Formation of amorphous aggregate has been observed at the physiological pH. Initial addition of HgCl2 resulted in the association of monomers to oligomers that proceeded to non-fibrillar aggregates on further addition. The sigmoidal curve obtained from the Stern-Volmer plot clearly divided into three stage transition. A strong lag phase is observed indicating the time dependence for the association of competent monomers. The second stage was resolved into non-cooperative binding. These results match very well with the data from atomic force microscopy and the free energy change observed in the regions. Raman spectroscopic studies indicated toxic antiparallel ß-sheets structure. Time dependent atomic force microscopy study revealed the off-pathway nature of amorphous aggregates. At molten globular state, similar quenching behaviour is observed. The atomic force microscopy images clearly indicate at pH 2.2 the initiation of fibril formation occurs at lower concentration of HgCl2 itself. Our results revealed the conformation switch of ovalbumin upon the contact of an environmental toxin and its possible way of toxicity.


Asunto(s)
Metales Pesados/toxicidad , Agregado de Proteínas/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos , Amiloide/química , Ambiente , Concentración de Iones de Hidrógeno , Cinética , Cloruro de Mercurio/química , Cloruro de Mercurio/toxicidad , Mercurio/química , Mercurio/toxicidad , Metales Pesados/química , Microscopía de Fuerza Atómica/métodos , Conformación Molecular , Ovalbúmina/química , Ovalbúmina/efectos de los fármacos , Transición de Fase , Proteínas/química , Proteínas/efectos de los fármacos , Espectrometría Raman/métodos
10.
Int J Biol Macromol ; 166: 687-693, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33130263

RESUMEN

Alzheimer's disease is characterized by important patho-proteins, which being composed of Amyloid-ß plaques and intracellular neurofibrillary tangles of Tau. Intrinsically disordered protein tau has several interacting partners, which are necessary for its normal functioning. Tau has been shown to interact with various proteins, nucleic acid, and lipids. α-Linolenic acid (ALA) a plant-based omega-3 fatty acid has been studied for its role as neuroprotective and beneficial fatty acid in the brain. In this study, we are focusing on the ability of ALA to induce spontaneous assembly in tau protein. ALA inhibited the Tau aggregation as indicated by reduced ThS fluorescence kinetics, which indicates no aggregation of Tau. Similarly, SDS-PAGE analysis supported that ALA exposure inhibited the aggregation as no higher-order tau species were observed. Along with its ability to impede the aggregation of Tau, ALA also maintains a native random coiled structure, which was estimated by CD spectroscopy. Finally, TEM analysis showed that the formation of Tau fibrils was found to be discouraged by ALA. Hence, conclusion of the study suggested that ALA profoundly inhibited aggregation of Tau and maintained it's the random-coil structure.


Asunto(s)
Multimerización de Proteína/efectos de los fármacos , Ácido alfa-Linolénico/farmacología , Proteínas tau/química , Humanos , Conformación Proteica , Desplegamiento Proteico/efectos de los fármacos , Ácido alfa-Linolénico/química , Proteínas tau/metabolismo
11.
Biochemistry ; 59(39): 3650-3659, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32924445

RESUMEN

Misfolding of Cu, Zn superoxide dismutase (SOD1) variants may lead to protein aggregation and ultimately amyotrophic lateral sclerosis (ALS). The mechanism and protein conformational changes during this process are complex and remain unclear. To study SOD1 variant aggregation at the molecular level and in solution, we chemically induced aggregation of a mutant variant (G93A SOD1) with trifluoroethanol (TFE) and used both native mass spectrometry (MS) to analyze the intact protein and fast photochemical oxidation of proteins (FPOP) to characterize the structural changes induced by TFE. We found partially unfolded G93A SOD1 monomers prior to oligomerization and identified regions of the N-terminus, C-terminus, and strands ß5, ß6 accountable for the partial unfolding. We propose that exposure of hydrophobic interfaces of these unstructured regions serves as a precursor to aggregation. Our results provide a possible mechanism and molecular basis for ALS-linked SOD1 misfolding and aggregation.


Asunto(s)
Agregado de Proteínas/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos , Superóxido Dismutasa/química , Trifluoroetanol/farmacología , Humanos , Espectrometría de Masas , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Huella de Proteína , Espectrometría de Masa por Ionización de Electrospray
12.
Chem Biodivers ; 17(10): e2000495, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32865330

RESUMEN

Human pancreatic tumor cells such as PANC-1 are known for their ability to tolerate nutrient starvation and thrive under the hypovascular tumor microenvironment, a phenomenon termed as 'austerity'. A search of agents that preferentially inhibit the cancer cell viability under the starvation condition without toxicity in the nutrient-rich condition is a promising approach in anticancer drug discovery. In this study, a triterpene lactone, 3ß-hydroxy-13,28-epoxyurs-11-en-28-one (ursenolide), isolated from a Callistemon citrinus extract has shown strong preferential cytotoxicity against PANC-1 cells under nutrient starvation with PC50 value of 0.4 µm. Ursenolide-induced rounding of PANC-1 cell morphology followed by rupture of the cell membrane leading to cell death. In a real-time cell migration study, ursenolide was found to inhibit PANC-1 cell migration significantly. Mechanistically, it inhibited GRP78 and GRP94 under the starvation condition suggesting inhibition of unfolded protein response (UPR), an adaptive process of cell survival during starvation. It also inhibited the phosphorylation of the key survival protein Akt and mTOR. Overall results suggested that ursenolide is a potential anticancer agent against pancreatic cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas de Choque Térmico/antagonistas & inhibidores , Lactonas/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , Myrtaceae/química , Neoplasias Pancreáticas/tratamiento farmacológico , Triterpenos/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Humanos , Lactonas/química , Lactonas/aislamiento & purificación , Glicoproteínas de Membrana/metabolismo , Conformación Molecular , Neoplasias Pancreáticas/patología , Desplegamiento Proteico/efectos de los fármacos , Triterpenos/química , Triterpenos/aislamiento & purificación , Células Tumorales Cultivadas
13.
Phys Chem Chem Phys ; 22(35): 19779-19786, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32844828

RESUMEN

Ionic liquids (ILs) are gaining attention as protein stabilizers and refolding additives. However, varying degrees of success with this approach motivates the need to better understand fundamental IL-protein interactions. A combination of experiment and simulation is used to investigate the thermal unfolding of lysozyme in the presence of two imidazolium-based ILs (1-ethyl-3-methylimidazolium ethylsulfate, [EMIM][EtSO4] and 1-ethyl-3-methylimidazolium diethylphosphate, [EMIM][Et2PO4]). Both ILs reduce lysozyme melting temperature Tm, but more gradually than strong denaturants. [EMIM][Et2PO4] lowers lysozyme Tm more readily than [EMIM][EtSO4], as well as requiring less energy to unfold the protein, as determined by the calorimetric enthalpy ΔH. Intrinsic fluorescence measurements indicate that both ILs bind to tryptophan residues in a dynamic mode, and furthermore, molecular dynamics simulations show a high density of [EMIM]+ near lysozyme's Trp62 residue. For both ILs approximately half of the [EMIM]+ cations near Trp62 show perfect alignment of their respective rings. The [EMIM]+ cations, having a "local" effect in binding to tryptophan, likely perturb a critically important Arg-Trp-Arg bridge through favorable π-π and cation-π interactions. Simulations show that the anions, [EtSO4]- and [Et2PO4]-, interact in a "global" manner with lysozyme, due to this protein's strong net positive charge. The anions also determine the local distribution of ions surrounding the protein. [Et2PO4]- is found to have a closer first coordination shell around the protein and stronger Coulomb interactions with lysozyme than [EtSO4]-, which could explain why the former anion is more destabilizing. Patching of ILs to the protein surface is also observed, suggesting there is no universal IL solvent for proteins, and highlighting the complexity of the IL-protein environment.


Asunto(s)
Líquidos Iónicos/química , Muramidasa/química , Desplegamiento Proteico/efectos de los fármacos , Animales , Pollos , Imidazoles/química , Simulación de Dinámica Molecular , Organofosfatos/química , Estabilidad Proteica/efectos de los fármacos , Termodinámica , Temperatura de Transición/efectos de los fármacos
14.
Biochemistry ; 59(31): 2870-2881, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32786888

RESUMEN

The properties of enzymes packaged within the coat protein shell of virus-like particles (VLPs) were studied to provide a comprehensive assessment of such factors. Such entrainment did not seem to perturb enzyme function, but it did significantly enhance enzyme stability against several denaturing stimuli such as heat, organic solvents, and chaotropic agents. This improvement in performance was found to be general and independent of the number of independent subunits required and of the number of catalytically active enzymes packaged. Packaged enzymes were found by measurements of intrinsic tryptophan fluorescence to retain some of their native folded structure even longer than their catalytic activity, suggesting that protein folding is a significant component of the observed catalytic benefits. While we are unable to distinguish between kinetic and thermodynamic effects - including inhibition of enzyme unfolding, acceleration of refolding, and biasing of folding equilibria - VLP packaging appears to represent a useful general strategy for the stabilization of enzymes that operate on diffusible substrates and products.


Asunto(s)
Enzimas/metabolismo , Leviviridae , Virión/metabolismo , Biocatálisis , Estabilidad de Enzimas/efectos de los fármacos , Enzimas/química , Desplegamiento Proteico/efectos de los fármacos , Solventes/farmacología
15.
J Inorg Biochem ; 211: 111179, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32736274

RESUMEN

We have investigated the structural stability of the SARS (Severe acute respiratory syndrome)-CoV-2 main protease monomer (Mpro). We quantified the spatial and angular changes in the structure using two independent analyses, one based on a spatial metrics (δ, ratio), the second on angular metrics. The order of unfolding of the 10 helices in Mpro is characterized by beta vs alpha plots similar to those of cytochromes and globins. The longest turning region is anomalous in the earliest stage of unfolding. In an investigation of excluded-volume effects, we found that the maximum spread in average molecular-volume values for Mpro, cytochrome c-b562, cytochrome c', myoglobin, and cytoglobin is ~10 Å3. This apparent universality is a consequence of the dominant contributions from six residues: ALA, ASP, GLU, LEU, LYS and VAL. Of the seven Mpro histidines, residues 41, 163, 164, and 246 are in stable H-bonded regions; metal ion binding to one or more of these residues could break up the H-bond network, thereby affecting protease function. Our analysis also indicated that metal binding to cysteine residues 44 and 145 could disable the enzyme.


Asunto(s)
Proteasas 3C de Coronavirus/química , SARS-CoV-2/enzimología , Cobalto/química , Cobalto/metabolismo , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Cisteína/química , Histidina/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Unión Proteica , Estabilidad Proteica/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos
16.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165922, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32800945

RESUMEN

Excessive production of immunoglobulins (Ig) causes endoplasmic reticulum (ER) stress and triggers the unfolded protein response (UPR). Hypergammaglobulinemia and lymphadenopathy are hallmarks of murine AIDS that develops in mice infected with the LP-BM5 murine leukemia retrovirus complex. In these mice, Th2 polarization and aberrant humoral response have been previously correlated to altered intracellular redox homeostasis. Our goal was to understand the role of the cell's redox state in Ig secretion and plasma cell (PC) maturation. To this aim, LP-BM5-infected mice were treated with I-152, an N-acetyl-cysteine and cysteamine supplier. Intraperitoneal I-152 administration (30 µmol/mouse three times a week for 9 weeks) decreased plasma IgG and increased IgG/Syndecan 1 ratio in the lymph nodes where IgG were in part accumulated within the ER. PC containing cytoplasmic inclusions filled with IgG were present in all animals, with fewer mature PC in those treated with I-152. Infection induced up-regulation of signaling molecules involved in the UPR, i.e. CHAC1, BiP, sXBP-1 and PDI, that were generally unaffected by I-152 treatment except for PDI and sXBP-1, which have a key role in protein folding and PC maturation, respectively. Our data suggest that one of the mechanisms through which I-152 can limit hypergammaglobulinemia in LP-BM5-infected mice is by influencing IgG folding/assembly as well as secretion and affecting PC maturation.


Asunto(s)
Acetilcisteína/análogos & derivados , Antivirales/farmacología , Cisteamina/análogos & derivados , Inmunoglobulinas/metabolismo , Células Plasmáticas/efectos de los fármacos , Infecciones por Retroviridae/tratamiento farmacológico , Infecciones Tumorales por Virus/tratamiento farmacológico , Respuesta de Proteína Desplegada/efectos de los fármacos , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Animales , Antivirales/administración & dosificación , Cisteamina/administración & dosificación , Cisteamina/farmacología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulinas/sangre , Inyecciones Intraperitoneales , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/metabolismo , Células Plasmáticas/virología , Desplegamiento Proteico/efectos de los fármacos , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/virología
17.
mBio ; 11(3)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32605984

RESUMEN

Antibiotic acyldepsipeptides (ADEPs) deregulate ClpP, the proteolytic core of the bacterial Clp protease, thereby inhibiting its native functions and concomitantly activating it for uncontrolled proteolysis of nonnative substrates. Importantly, although ADEP-activated ClpP is assumed to target multiple polypeptide and protein substrates in the bacterial cell, not all proteins seem equally susceptible. In Bacillus subtilis, the cell division protein FtsZ emerged to be particularly sensitive to degradation by ADEP-activated ClpP at low inhibitory ADEP concentrations. In fact, FtsZ is the only bacterial protein that has been confirmed to be degraded in vitro as well as within bacterial cells so far. However, the molecular reason for this preferred degradation remained elusive. Here, we report the unexpected finding that ADEP-activated ClpP alone, in the absence of any Clp-ATPase, leads to an unfolding and subsequent degradation of the N-terminal domain of FtsZ, which can be prevented by the stabilization of the FtsZ fold via nucleotide binding. At elevated antibiotic concentrations, importantly, the C terminus of FtsZ is notably targeted for degradation in addition to the N terminus. Our results show that different target structures are more or less accessible to ClpP, depending on the ADEP level present. Moreover, our data assign a Clp-ATPase-independent protein unfolding capability to the ClpP core of the bacterial Clp protease and suggest that the protein fold of FtsZ may be more flexible than previously anticipated.IMPORTANCE Acyldepsipeptide (ADEP) antibiotics effectively kill multidrug-resistant Gram-positive pathogens, including vancomycin-resistant enterococcus, penicillin-resistant Streptococcus pneumoniae (PRSP), and methicillin-resistant Staphylococcus aureus (MRSA). The antibacterial activity of ADEP depends on a new mechanism of action, i.e., the deregulation of bacterial protease ClpP that leads to bacterial self-digestion. Our data allow new insights into the mode of ADEP action by providing a molecular explanation for the distinct bacterial phenotypes observed at low versus high ADEP concentrations. In addition, we show that ClpP alone, in the absence of any unfoldase or energy-consuming system, and only activated by the small molecule antibiotic ADEP, leads to the unfolding of the cell division protein FtsZ.


Asunto(s)
Bacillus subtilis/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Depsipéptidos/farmacología , Endopeptidasa Clp/metabolismo , Desplegamiento Proteico/efectos de los fármacos , Antibacterianos/farmacología , Bacillus subtilis/enzimología , División Celular/efectos de los fármacos , Depsipéptidos/química
18.
NMR Biomed ; 33(10): e4367, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32656956

RESUMEN

The aim of this study was to investigate possible sodium triple-quantum (TQ) signal dependence on pH variation and protein unfolding which may happen in vivo. The model system, composed of bovine serum albumin (BSA), was investigated over a wide pH range of 0.70 to 13.05 and during urea-induced unfolding. In both experimental series, the sodium and BSA concentration were kept constant so that TQ signal changes solely arose from an environmental change. The experiments were performed using unique potential to detect weak TQ signals by implementing a TQ time proportional phase increment pulse sequence. At a pH of 0.70, in which case the effect of the negatively charged groups was minimized, the minimum TQ percentage relative to single-quantum of 1.34% ± 0.05% was found. An increase of the pH up to 13.05 resulted in an increase of the sodium TQ signal by 225%. Urea-induced unfolding of BSA, without changes in pH, led to a smaller increase in the sodium TQ signal of up to 40%. The state of BSA unfolding was verified by fluorescence microscopy. Results of both experiments were well fitted by sigmoid functions. Both TQ signal increases were in agreement with an increase of the availability of negatively charged groups. The results point to vital contributions of the biochemical environment to the TQ MR signals. The sodium TQ signal in vivo could be a valuable biomarker of cell viability, and therefore possible effects of pH and protein unfolding need to be considered for a proper interpretation of changes in sodium TQ signals.


Asunto(s)
Espectroscopía de Resonancia Magnética , Procesamiento de Señales Asistido por Computador , Sodio/química , Animales , Bovinos , Fluorescencia , Concentración de Iones de Hidrógeno , Fantasmas de Imagen , Conformación Proteica , Desplegamiento Proteico/efectos de los fármacos , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/metabolismo , Electricidad Estática , Urea/farmacología
19.
Mol Pharm ; 17(8): 2971-2986, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32687367

RESUMEN

Coformulations containing two therapeutic monoclonal antibodies (mAbs) could offer various benefits like enhanced therapeutic efficacy and better patient compliance. However, there are very few published studies on coformulations and binary mixtures of mAbs. It remains unclear to what extent mAbs with different physicochemical properties can be combined in solution without detrimental effects on protein stability. Here, we present a study including six model mAbs of the IgG1 subclass that are commercially available. In silico and biophysical characterization shows that the proteins have different physicochemical properties. Thus, their combinations represent various scenarios for coformulation development. We prepared all possible binary mixtures of the six mAbs and determined several biophysical parameters that are assessed during early-stage protein drug product development. The measured biophysical parameters are indicative of the conformational protein stability (inflection points of the thermal protein unfolding transitions) and the colloidal protein stability (aggregation onset temperatures and interaction parameter kD from dynamic light scattering). Remarkably, all 15 binary mAb mixtures do not exhibit biophysical parameters that indicate inferior conformational or colloidal stability compared to the least stable mAb in the mixture. Our findings suggest that the coformulation of some therapeutic monoclonal antibodies of the IgG1 subclass could be possible in a straightforward way as severe detrimental effects on the stability of these proteins in binary mixtures were not observed.


Asunto(s)
Anticuerpos Monoclonales/química , Preparaciones Farmacéuticas/química , Biofisica/métodos , Inmunoglobulina G/química , Estabilidad Proteica/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos
20.
Biochemistry ; 59(31): 2849-2858, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32667811

RESUMEN

The sterile α motif, also called the SAM domain, is known to form homo or heterocomplexes that modulate diverse biological functions through the regulation of specific protein-protein interactions. The MAPK pathway of budding yeast Saccharomyces cerevisiae is comprised of a three-tier kinase system akin to mammals. The MAPKKK Ste11 protein of yeast contains a homodimer SAM domain, which is critical for transmitting cues to the downstream kinases. The structural stability of the dimeric Ste11 SAM is maintained by hydrophobic and ionic interactions at the interfacial amino acids. The urea-induced equilibrium-unfolding process of the Ste11 SAM domain is cooperative without evidence of any intermediate states. The native-state H/D exchange under subdenaturing conditions is a useful method for the detection of intermediate states of proteins. In the present study, we investigated the effect of ionic strength on the conformational stability of the dimer using the H/D exchange experiments. The hydrogen exchange behavior of the Ste11 dimer under physiological salt concentrations reveals two partially unfolded metastable intermediate states, which may be generated by a sequential and cooperative unfolding of the five helices present in the domain. These intermediates appear to be significant for the reversible unfolding kinetics via hydrophobic collapse. In contrast, higher ionic concentrations eliminate this cooperative interactions that stabilize the pairs of helices.


Asunto(s)
Medición de Intercambio de Deuterio , Quinasas Quinasa Quinasa PAM/química , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Multimerización de Proteína/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/química , Cloruro de Sodio/farmacología , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas/efectos de los fármacos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Estructura Cuaternaria de Proteína , Desplegamiento Proteico/efectos de los fármacos , Urea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA