Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Chem Biol Interact ; 351: 109718, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34717915

RESUMEN

The ABCG2 transporter plays a pivotal role in multidrug resistance, however, no clinical trial using specific ABCG2 inhibitors have been successful. Although ABC transporters actively extrude a wide variety of substrates, photodynamic therapeutic agents with porphyrinic scaffolds are exclusively transported by ABCG2. In this work, we describe for the first time a porphyrin derivative (4B) inhibitor of ABCG2 and capable to overcome multidrug resistance in vitro. The inhibition was time-dependent and 4B was not itself transported by ABCG2. Independently of the substrate, the porphyrin 4B showed an IC50 value of 1.6 µM and a mixed type of inhibition. This compound inhibited the ATPase activity and increased the binding of the conformational-sensitive antibody 5D3. A thermostability assay confirmed allosteric protein changes triggered by the porphyrin. Long-timescale molecular dynamics simulations revealed a different behavior between the ABCG2 porphyrinic substrate pheophorbide a and the porphyrin 4B. Pheophorbide a was able to bind in three different protein sites but 4B showed one binding conformation with a strong ionic interaction with GLU446. The inhibition was selective toward ABCG2, since no inhibition was observed for P-glycoprotein and MRP1. Finally, this compound successfully chemosensitized cells that overexpress ABCG2. These findings reinforce that substrates may be a privileged source of chemical scaffolds for identification of new inhibitors of multidrug resistance-linked ABC transporters.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Adenosina Trifosfatasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Porfirinas/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Irinotecán/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Porfirinas/química , Porfirinas/metabolismo , Unión Proteica , Conformación Proteica/efectos de los fármacos
2.
Front Endocrinol (Lausanne) ; 12: 711906, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867774

RESUMEN

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two neuropeptides that contribute to the regulation of intestinal motility and secretion, exocrine and endocrine secretions, and homeostasis of the immune system. Their biological effects are mediated by three receptors named VPAC1, VPAC2 and PAC1 that belong to class B GPCRs. VIP and PACAP receptors have been identified as potential therapeutic targets for the treatment of chronic inflammation, neurodegenerative diseases and cancer. However, pharmacological use of endogenous ligands for these receptors is limited by their lack of specificity (PACAP binds with high affinity to VPAC1, VPAC2 and PAC1 receptors while VIP recognizes both VPAC1 and VPAC2 receptors), their poor oral bioavailability (VIP and PACAP are 27- to 38-amino acid peptides) and their short half-life. Therefore, the development of non-peptidic small molecules or specific stabilized peptidic ligands is of high interest. Structural similarities between VIP and PACAP receptors are major causes of difficulties in the design of efficient and selective compounds that could be used as therapeutics. In this study we performed structure-based virtual screening against the subset of the ZINC15 drug library. This drug repositioning screen provided new applications for a known drug: ticagrelor, a P2Y12 purinergic receptor antagonist. Ticagrelor inhibits both VPAC1 and VPAC2 receptors which was confirmed in VIP-binding and calcium mobilization assays. A following analysis of detailed ticagrelor binding modes to all three VIP and PACAP receptors with molecular dynamics revealed its allosteric mechanism of action. Using a validated homology model of inactive VPAC1 and a recently released cryo-EM structure of active VPAC1 we described how ticagrelor could block conformational changes in the region of 'tyrosine toggle switch' required for the receptor activation. We also discuss possible modifications of ticagrelor comparing other P2Y12 antagonist - cangrelor, closely related to ticagrelor but not active for VPAC1/VPAC2. This comparison with inactive cangrelor could lead to further improvement of the ticagrelor activity and selectivity for VIP and PACAP receptor sub-types.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Reposicionamiento de Medicamentos/métodos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/efectos de los fármacos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/efectos de los fármacos , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/efectos de los fármacos , Ticagrelor/farmacología , Sitios de Unión , Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Estructura Molecular , Conformación Proteica/efectos de los fármacos , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/química , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/química , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/química , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Ticagrelor/química
3.
Int J Biol Macromol ; 187: 976-987, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34333006

RESUMEN

Coronavirus 3C-like protease (3CLpro) is a crucial target for treating coronavirus diseases including COVID-19. Our preliminary screening showed that Ampelopsis grossedentata extract (AGE) displayed potent SARS-CoV-2-3CLpro inhibitory activity, but the key constituents with SARS-CoV-2-3CLpro inhibitory effect and their mechanisms were unrevealed. Herein, a practical strategy via integrating bioactivity-guided fractionation and purification, mass spectrometry-based peptide profiling and time-dependent biochemical assay, was applied to identify the crucial constituents in AGE and to uncover their inhibitory mechanisms. The results demonstrated that the flavonoid-rich fractions (10-17.5 min) displayed strong SARS-CoV-2-3CLpro inhibitory activities, while the constituents in these fractions were isolated and their SARS-CoV-2-3CLpro inhibitory activities were investigated. Among all isolated flavonoids, dihydromyricetin, isodihydromyricetin and myricetin strongly inhibited SARS-CoV-2 3CLpro in a time-dependent manner. Further investigations demonstrated that myricetin could covalently bind on SARS-CoV-2 3CLpro at Cys300 and Cys44, while dihydromyricetin and isodihydromyricetin covalently bound at Cys300. Covalent docking coupling with molecular dynamics simulations showed the detailed interactions between the orthoquinone form of myricetin and two covalent binding sites (surrounding Cys300 and Cys44) of SARS-CoV-2 3CLpro. Collectively, the flavonoids in AGE strongly and time-dependently inhibit SARS-CoV-2 3CLpro, while the newly identified SARS-CoV-2 3CLpro inhibitors in AGE offer promising lead compounds for developing novel antiviral agents.


Asunto(s)
Proteasas Virales 3C/química , Proteasas Virales 3C/metabolismo , Ampelopsis/química , Antivirales/farmacología , Flavonoides/farmacología , SARS-CoV-2/enzimología , Antivirales/química , Sitios de Unión/efectos de los fármacos , Cisteína/metabolismo , Flavonoides/química , Flavonoles/química , Flavonoles/farmacología , Espectrometría de Masas , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Extractos Vegetales/química , Extractos Vegetales/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos
4.
J Mol Model ; 27(8): 231, 2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34312718

RESUMEN

The Retinoid X Receptor (RXR) is an attractive target in the treatment of colon cancer. Different therapeutic binders with high potency have been used to specifically target RXR. Among these compounds is a novel analogue of berberine, B12. We provided structural and molecular insights into the therapeutic activity properties of B12 relative to its parent compound, berberine, using force field estimations and thermodynamic calculations. Upon binding of B12 to RXR, the high instability elicited by RXR was markedly reduced; similar observation was seen in the berberine-bound RXR. However, our analysis revealed that B12 could have a more stabilizing effect on RXR when compared to berberine. Interestingly, the mechanistic behaviour of B12 in the active site of RXR opposed its impact on RXR protein. This disparity could be due to the bond formation and breaking elicited between B12/berberine and the active site residues. We observed that B12 and berberine could induce a disparate conformational change in regions Gly250-Asp258 located on the His-RXRα/LBD domain. Comparatively, the high agonistic and activation potential reported for B12 compared to berberine might be due to its superior binding affinity as evidenced in the thermodynamic estimations. The total affinity for B12 (-25.76 kcal/mol) was contributed by electrostatic interactions from Glu243 and Glu239. Also, Arg371, which plays a crucial role in the activity of RXR, formed a strong hydrogen bond with B12; however, a weak interaction was elicited between Arg371 and berberine. Taken together, our study has shown the RXRα activating potential of B12, and findings from this study could provide a framework in the future design of RXRα binders specifically tailored in the selective treatment of colon cancer.


Asunto(s)
Berberina/química , Neoplasias del Colon/tratamiento farmacológico , Enlace de Hidrógeno/efectos de los fármacos , Receptores X Retinoide/genética , Berberina/análogos & derivados , Berberina/uso terapéutico , Dominio Catalítico/efectos de los fármacos , Neoplasias del Colon/genética , Humanos , Terapia Molecular Dirigida , Conformación Proteica/efectos de los fármacos , Receptores X Retinoide/antagonistas & inhibidores , Termodinámica
5.
Food Funct ; 11(7): 6595-6607, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32648885

RESUMEN

We extracted and purified oxyresveratrol (OXY) from Artocarpus heterophyllus Lam. and identified its structure. The kinetics and mechanisms of OXY-induced mushroom tyrosinase inhibition were studied using fluorescence spectroscopy, copper ion chelation, and circular dichroism (CD). We found that OXY significantly inhibited tyrosinase with a half maximal inhibitory concentration (IC50) of 0.03 mM. The inhibitory effect of OXY on tyrosinase was almost 25 times that of kojic acid, which had an IC50 of 0.78 mM. Additionally, OXY and the tyrosinase substrate L-dopa did not have a competitive relationship; OXY is a non-competitive inhibitor. Using a fluorescence quenching experiment, we determined the corresponding rate constant (Kq) values at 298, 303, and 310 K to be 2.24 × 1012, 1.08 × 1012 and 1.44 × 1012 L mol-1 s-1, respectively. The OXY and tyrosinase interaction occured mainly through van der Waals forces and a hydrogen bond between the -OH group and its amino acid residue. Furthermore, we investigated the effects of OXY on murine melanoma B16 cells and on age pigments in Caenorhabditis elegans (C. elegans). OXY decreased melanin production by inhibiting the tyrosinase activity in murine melanoma B16 cells, which decreased superoxide dismutase (SOD) and glutathione peroxidase (GSH) and increased catalase (CAT), leading to apoptosis. The lifespan of nematodes in the 50 ml resveratrol-treated group was significantly longer than that in the blank group by 5%. The mean lifespan of nematodes in the 50 µM OXY-treated group was significantly longer than that in the blank group by 6.82%.The fluorescence intensity of C. elegans pigments decreased by 30.43%, 47.35% and 64.42% after the treatment with a low, middle, and high OXY dose, respectively, showing that OXY has a significant inhibitory effect on melanin and age pigment production.


Asunto(s)
Monofenol Monooxigenasa/antagonistas & inhibidores , Extractos Vegetales/farmacología , Estilbenos/farmacología , Agaricales/enzimología , Animales , Apoptosis/efectos de los fármacos , Artocarpus/química , Caenorhabditis elegans , Melaninas/biosíntesis , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental , Ratones , Monofenol Monooxigenasa/química , Monofenol Monooxigenasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/aislamiento & purificación , Conformación Proteica/efectos de los fármacos , Pigmentación de la Piel/efectos de los fármacos , Estilbenos/aislamiento & purificación , Factores de Tiempo
6.
Molecules ; 25(12)2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32580406

RESUMEN

Despite extensive efforts in the development of drugs for complex neurodegenerative diseases, treatment often remains challenging or ineffective, and hence new treatment strategies are necessary. One approach is the design of multi-target drugs, which can potentially address the complex nature of disorders such as Alzheimer's disease. We report a method for high throughput virtual screening aimed at identifying new dual target hit molecules. One of the identified hits, N,N-dimethyl-1-(4-(3-methyl-[1,2,4]triazolo[4,3-a]pyrimidin-6-yl)phenyl)ethan-1-amine (Ý;mir-2), has dual-activity as an acetylcholinesterase (AChE) inhibitor and as an α7 nicotinic acetylcholine receptor (α7 nAChR) agonist. Using computational chemistry methods, parallel and independent screening of a virtual compound library consisting of 3,848,234 drug-like and commercially available molecules from the ZINC15 database, resulted in an intersecting set of 57 compounds, that potentially possess activity at both of the two protein targets. Based on ligand efficiency as well as scaffold and molecular diversity, 16 of these compounds were purchased for in vitro validation by Ellman's method and two-electrode voltage-clamp electrophysiology. Ý;mir-2 was shown to exhibit the desired activity profile (AChE IC50 = 2.58 ± 0.96 µM; α7 nAChR activation = 7.0 ± 0.9% at 200 µM) making it the first reported compound with this particular profile and providing further evidence of the feasibility of in silico methods for the identification of novel multi-target hit molecules.


Asunto(s)
Acetilcolinesterasa/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Inhibidores de la Colinesterasa/aislamiento & purificación , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Acetilcolinesterasa/química , Acetilcolinesterasa/ultraestructura , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Inhibidores de la Colinesterasa/química , Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida , Conformación Proteica/efectos de los fármacos , Relación Estructura-Actividad , Interfaz Usuario-Computador , Receptor Nicotínico de Acetilcolina alfa 7/química , Receptor Nicotínico de Acetilcolina alfa 7/ultraestructura
7.
J Recept Signal Transduct Res ; 40(6): 605-612, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32476594

RESUMEN

Recently, a pathogen has been identified as a novel coronavirus (SARS-CoV-2) and found to trigger novel pneumonia (COVID-19) in human beings and some other mammals. The uncontrolled release of cytokines is seen from the primary stages of symptoms to last acute respiratory distress syndrome (ARDS). Thus, it is necessary to find out safe and effective drugs against this deadly coronavirus as soon as possible. Here, we downloaded the three-dimensional model of NSP10/NSP16 methyltransferase (PDB-ID: 6w6l) and main protease (PDB-ID: 6lu7) of COVID-19. Using these molecular models, we performed virtual screening with our anti-viral, inti-infectious, and anti-protease compounds, which are attractive therapeutics to prevent infection of the COVID-19. We found that top screened compound binds with protein molecules with good dock score with the help of hydrophobic interactions and hydrogen bonding. We observed that protease complexed with Cyclocytidine hydrochloride (anti-viral and anti-cancer), Trifluridine (anti-viral), Adonitol, and Meropenem (anti-bacterial), and Penciclovir (anti-viral) bound with a good docking score ranging from -6.8 to -5.1 (Kcal/mol). Further, NSP10/NSP16 methyltransferase complexed with Telbivudine, Oxytetracycline dihydrate (anti-viral), Methylgallate (anti-malarial), 2-deoxyglucose and Daphnetin (anti-cancer) from the docking score of -7.0 to -5.7 (Kcal/mol). In conclusion, the selected compounds may be used as a novel therapeutic agent to combat this deadly pandemic disease, SARS-CoV-2 infection, but needs further experimental research.HighlightsNSP10/NSP16 methyltransferase and main protease complex of SARS CoV-2 bind with selected drugs.NSP10/NSP16 methyltransferase and protease interacted with drugs by hydrophobic interactions.Compounds show good DG binging free energy with protein complexes.Ligands were found to follow the Lipinski rule of five.


Asunto(s)
Antivirales/química , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Proteínas no Estructurales Virales/química , Proteínas Reguladoras y Accesorias Virales/química , Aciclovir/análogos & derivados , Aciclovir/química , Aciclovir/uso terapéutico , Ancitabina/química , Ancitabina/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/virología , Evaluación Preclínica de Medicamentos , Guanina , Humanos , Meropenem/química , Meropenem/uso terapéutico , Metiltransferasas , Modelos Moleculares , Simulación del Acoplamiento Molecular , Pandemias , Neumonía Viral/virología , Conformación Proteica/efectos de los fármacos , Ribitol/química , Ribitol/uso terapéutico , SARS-CoV-2 , Trifluridina/química , Trifluridina/uso terapéutico , Interfaz Usuario-Computador , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/ultraestructura , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/ultraestructura
8.
Sci Rep ; 10(1): 8661, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457393

RESUMEN

High risk human papillomaviruses are highly associated with the cervical carcinoma and the other genital tumors. Development of cervical cancer passes through the multistep process initiated from benign cyst to increasingly severe premalignant dysplastic lesions in an epithelium. Replication of this virus occurs in the fatal differentiating epithelium and involves in the activation of cellular DNA replication proteins. The oncoprotein E7 of human papillomavirus expressed in the lower epithelial layers constrains the cells into S-phase constructing an environment favorable for genome replication and cell proliferation. To date, no suitable drug molecules exist to treat HPV infection whereas anticipation of novel anti-HPV chemotherapies with distinctive mode of actions and identification of potential drugs are crucial to a greater extent. Hence, our present study focused on identification of compounds analogue to EGCG, a green tea molecule which is considered to be safe to use for mammalian systems towards treatment of cancer. A three dimensional similarity search on the small molecule library from natural product database using EGCG identified 11 potential small molecules based on their structural similarity. The docking strategies were implemented with acquired small molecules and identification of the key interactions between protein and compounds were carried out through binding free energy calculations. The conformational changes between the apoprotein and complexes were analyzed through simulation performed thrice demonstrating the dynamical and structural effects of the protein induced by the compounds signifying the domination. The analysis of the conformational stability provoked us to describe the features of the best identified small molecules through electronic structure calculations. Overall, our study provides the basis for structural insights of the identified potential identified small molecules and EGCG. Hence, the identified analogue of EGCG can be potent inhibitors against the HPV 16 E7 oncoprotein.


Asunto(s)
Catequina/análogos & derivados , Evaluación Preclínica de Medicamentos/métodos , Papillomavirus Humano 16/efectos de los fármacos , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Infecciones por Papillomavirus/tratamiento farmacológico , Neoplasias del Cuello Uterino/prevención & control , Antivirales/farmacología , Catequina/química , Catequina/farmacología , Proliferación Celular/genética , Quimioprevención/métodos , Descubrimiento de Drogas , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Análisis de Componente Principal , Conformación Proteica/efectos de los fármacos , Neoplasias del Cuello Uterino/virología , Internalización del Virus/efectos de los fármacos
9.
Langmuir ; 36(7): 1804-1812, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32011894

RESUMEN

Deposition of amyloid-ß (Aß) aggregates in the brain is a main pathological hallmark of Alzheimer's disease (AD), so inhibition of Aß aggregation has been considered as a promising strategy for AD prevention and treatment. Black phosphorus (BP) is a 2D nanomaterial with high biocompatibility and unique biodegradability, but its potential application in biomedicine suffers from the rapid degradability and unfunctionability. To overcome the drawbacks and broaden its application, we have herein designed an Aß inhibitor (LK7)-coupled and polyethylene glycol (PEG)-stabilized BP-based nanosystem. The PEGylated-LK7-BP nanosheets (PEG-LK7@BP) not only exhibited a good stability but also demonstrated a significantly enhanced inhibitory potency on Aß42 fibrillogenesis in comparison with its counterparts. This elaborately designed PEG-LK7@BP stopped the conformational transition and suppressed the fibrillization of Aß42, so it could completely rescue cultured cells from the toxicity of Aß42 (by increasing the cell viability from 72 to 100%) at 100 µg/mL. It is considered that PEG-LK7@BP could bind Aß species by enhanced electrostatic and hydrophobic interactions and thus efficiently alleviated Aß-Aß interactions. Meanwhile, the coupled LK7 on the BP surface formed a high local concentration that enhanced the affinity between the nanosystem and Aß species. Finally, PEG could improve the stability and dispersibility of the nanoplatform to make it show an increased inhibitory effect on the amyloid formation. Hence, this work proved that PEG-LK7@BP is a promising nanosystem for the development of amyloid inhibitors fighting against AD.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Nanoestructuras/química , Fragmentos de Péptidos/efectos de los fármacos , Péptidos/farmacología , Fósforo/farmacología , Polietilenglicoles/farmacología , Péptidos beta-Amiloides/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Nanoestructuras/toxicidad , Fragmentos de Péptidos/toxicidad , Péptidos/química , Péptidos/toxicidad , Fósforo/química , Fósforo/toxicidad , Polietilenglicoles/química , Polietilenglicoles/toxicidad , Conformación Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos
10.
Redox Biol ; 28: 101331, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31568923

RESUMEN

Iodide ions (I-) are an essential dietary mineral, and crucial for mental and physical development, fertility and thyroid function. I- is also a high affinity substrate for the heme enzyme myeloperoxidase (MPO), which is involved in bacterial cell killing during the immune response, and also host tissue damage during inflammation. In the presence of H2O2 and Cl-, MPO generates the powerful oxidant hypochlorous acid (HOCl), with excessive formation of this species linked to multiple inflammatory diseases. In this study, we have examined the hypothesis that elevated levels of I- would decrease HOCl formation and thereby protein damage induced by a MPO/Cl-/H2O2 system, by acting as a competitive substrate. The presence of increasing I- concentrations (0.1-10 µM; i.e. within the range readily achievable by oral supplementation in humans), decreased damage to both model proteins and extracellular matrix components as assessed by gross structural changes (SDS-PAGE), antibody recognition of parent and modified protein epitopes (ELISA), and quantification of both parent amino acid loss (UPLC) and formation of the HOCl-biomarker 3-chlorotyrosine (LC-MS) (reduced by ca. 50% at 10 µM I-). Elevated levels of I- ( > 1 µM) also protected against functional changes as assessed by a decreased loss of adhesion (eg. 40% vs. < 22% with >1 µM I-) of primary human coronary artery endothelial cells (HCAECs), to MPO-modified human plasma fibronectin. These data indicate that low micromolar concentrations of I-, which can be readily achieved in humans and are readily tolerated, may afford protection against cell and tissue damage induced by MPO.


Asunto(s)
Susceptibilidad a Enfermedades , Hemo/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Yoduros/metabolismo , Peroxidasa/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Fibronectinas/química , Fibronectinas/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Ácido Hipocloroso/metabolismo , Inflamación/patología , Yoduros/farmacología , Oxidación-Reducción , Conformación Proteica/efectos de los fármacos
11.
Anal Chem ; 91(19): 12560-12567, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31487155

RESUMEN

Signaling proteins exemplified by calmodulin usually bind cooperatively to multiple ligands. Intermediate states and allosteric behavior are difficult to characterize. Here we extend a recently reported mass spectrometry (MS)-based method named LITPOMS (ligand titration, fast photochemical oxidation of proteins and mass spectrometry) that characterizes complex binding systems typically found as signaling proteins. As reported previously, calmodulin's response to binding four Ca2+ can be determined by LITPOMS to reveal binding sites, binding order, and most importantly composite binding behavior. Modeling this behavior provides site-specific binding affinities. In this article, we dissect the composite, peptide-level conformational changes at several regions either by digestion with a different protease or by tandem MS of LITPOMS behavior at the amino-acid residue level. Such dissection greatly elevates spatial resolution and increases the confidence of binding-order assignment. These complementary views of complex protein conformational change recapitulate the cumulative understanding via a single approach, providing new insights on poorly understood yet important allostery and underpin an approach applicable for exploring other signaling systems.


Asunto(s)
Calcio/metabolismo , Calcio/farmacología , Calmodulina/química , Calmodulina/metabolismo , Ligandos , Modelos Moleculares , Unión Proteica , Conformación Proteica/efectos de los fármacos , Proteolisis
12.
Int J Biol Macromol ; 138: 800-809, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31351960

RESUMEN

Herein, we explored the interaction of Al2O3 NPs with RBCs and Hb to determine the effect of Al2O3 NPs on hemolytic activity and Hb denaturation. The percentage of hemolysis of extracts and direct contact assays triggered by Al2O3 NPs was calculated by determining supernatant Hb concentration at 540 nm. Far-UV CD and Trp/ANS/acrylamide fluorescence spectroscopic methods were used to determine the structural changes of Hb upon interaction with Al2O3 NPs. Theoretical studies were carried out to display the residues involved in the binding site of Hb with Al2O3 nanocluster as well as the structural changes of Hb after interaction. The results showed that the percentage of hemolysis of extract and direct contact assays induced by Al2O3 NPs were 1.16 and 0.46, respectively. Fluorescence spectroscopy revealed that Al2O3 NPs alter the quaternary structure of the protein; however, CD spectroscopy indicated that the secondary structure of Hb remains almost unchanged. Theoretical study displayed that Al2O3 nanocluster interacts with different residues of protein, and Hb tends to be destabilized at the binding site with nanocluster. This study may be significant in exploring the toxicity profile of Al2O3 NPs for their in vivo implementations.


Asunto(s)
Óxido de Aluminio/química , Óxido de Aluminio/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemoglobinas/química , Conformación Proteica/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Modelos Moleculares , Conformación Molecular , Nanopartículas/química , Nanopartículas/ultraestructura , Análisis Espectral
13.
Int J Biol Macromol ; 133: 945-956, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31005690

RESUMEN

Abiotic stress induced by heavy metals retards the growth and development of plants. Therefore, it is essential to have an insight into the potential toxic effects of heavy metals. The present article investigates the effect of zinc and cadmium on the structure and function of garlic phytocystatin (GPhyCys). The cysteine proteinase inhibitory assay showed a reduction in the inhibitory activity upon binding with zinc and cadmium. UV-vis absorption spectroscopy revealed the complex formation of zinc and cadmium with garlic phytocystatin. Fluorescence quenching experiment confirmed the quenching of fluorophores upon binding of zinc and cadmium. Synchronous and 3-dimensional fluorescence spectroscopy suggest the alteration in the microenvironment around aromatic residues of garlic phytocystatin upon binding with the above metals. Circular dichroism showed a reduction in the alpha-helical content of native garlic phytocystatin. Scanning electron micrographs showed the morphological changes in the native garlic phytocystatin upon addition of zinc and cadmium. The observations confirmed the alteration in structure and conformation of garlic phytocystatin upon interaction with zinc and cadmium. It can be safely concluded that the high concentration of zinc and cadmium can alter the functioning of cysteine proteinase present in garlic and affects the growth and development of plants.


Asunto(s)
Cadmio/metabolismo , Cadmio/farmacología , Cistatinas/metabolismo , Ajo/metabolismo , Estrés Fisiológico/efectos de los fármacos , Zinc/metabolismo , Zinc/farmacología , Cistatinas/química , Ajo/efectos de los fármacos , Ajo/fisiología , Unión Proteica , Conformación Proteica/efectos de los fármacos
14.
Sci Rep ; 9(1): 4073, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30858542

RESUMEN

Vasculogenesis and angiogenesis are process of formation of blood vessels. Blood vessels are evolved to distribute nutrients and oxygen to distant organs. These vessels are crucial for growth and repair of wounded tissue. During tumor condition there occurs imbalance in the growth of blood vessels which leads to neo-angiogenesis. Neo-angiogenesis is major perpetrator behind the establishment of tumor. Tumor cells secrete pro-angiogenic factor VEGFA which binds to VEGFR2 present over surface of endothelial cells and triggers formation of new blood vessels. To inhibit tumor-angiogenesis, a physiologically-safe small molecule inhibitor was screened which can potentially interact with kinase domain of VEGFR2 and inhibit its activity. Molecular-docking module and biochemical analysis identified andrographolide as one of the best docking molecules that binds to ATP-binding pocket of VEGFR2 and inhibits its kinase activity. Thus, for a more radical approach towards safe VEGFR2 inhibitor, andrographolide was repurposed to inhibit tumor-angiogenesis and reduce tumor burden.


Asunto(s)
Diterpenos/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Adenosina Trifosfato/química , Adenosina Trifosfato/genética , Andrographis paniculata , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Proteínas Portadoras/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/farmacología , Diterpenos/química , Combinación de Medicamentos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Laminina/farmacología , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Extractos Vegetales/química , Conformación Proteica/efectos de los fármacos , Proteoglicanos/farmacología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química
15.
J Pharmacol Exp Ther ; 369(2): 282-290, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30846619

RESUMEN

Transient, reversible blockade of complex I during early reperfusion after ischemia limits cardiac injury. We studied the cardioprotection of high dose of metformin in cultured cells and mouse hearts via the novel mechanism of acute downregulation of complex I. The effect of high dose of metformin on complex I activity was studied in isolated heart mitochondria and cultured H9c2 cells. Protection with metformin was evaluated in H9c2 cells at reoxygenation and at early reperfusion in isolated perfused mouse hearts and in vivo regional ischemia reperfusion. Acute, high-dose metformin treatment inhibited complex I in ischemia-damaged mitochondria and in H9c2 cells following hypoxia. Accompanying the complex I modulation, high-dose metformin at reoxygenation decreased death in H9c2 cells. Acute treatment with high-dose metformin at the end of ischemia reduced infarct size following ischemia reperfusion in vitro and in vivo, including in the AMP kinase-dead mouse. Metformin treatment during early reperfusion improved mitochondrial calcium retention capacity, indicating decreased permeability transition pore (MPTP) opening. Acute, high-dose metformin therapy decreased cardiac injury through inhibition of complex I accompanied by attenuation of MPTP opening. Moreover, in contrast to chronic metformin treatment, protection by acute, high-dose metformin is independent of AMP-activated protein kinase activation. Thus, a single, high-dose metformin treatment at reperfusion reduces cardiac injury via modulation of complex I.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Metformina/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Citoprotección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Masculino , Metformina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Conformación Proteica/efectos de los fármacos
16.
Phytomedicine ; 52: 225-237, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30599902

RESUMEN

BACKGROUND: Native protein conformation is essential for the functional activity of the proteins and enzymes. Defects in conformation or alterations in expression of the proteins have been reported in various diseases. PURPOSE: The aim of this study is to review the molecular insight into the therapeutic potential of phytoconstituents targeting protein conformations or expressions. METHODS: Published literatures were searched in PubMed, Scopus, Web of Science; Article published till Dec 2017 were extracted. The literature was assessed from the Central University of Rajasthan, India. Present study evaluate article based on the role of active plant constituents on the conformation and expression of the different proteins. RESULTS: Plant components play their role either at the molecular level or cellular level and exhibit antibacterial, antiviral, anti-neurodegenerative and other activities. Plant active compounds isolated from different plants may either stabilize or destabilize the conformation of proteins or alter expression level of the protein involved in these diseases, therefore, can play a significant role in preventing diseases caused by the alteration in these proteins. CONCLUSION: In the present article, we have reviewed the molecular mechanism of plant active compounds, their target proteins, methods of extraction and identification, and their biological significances. Therefore, a proper understanding of the effect of these herbal molecules on the concerned proteins may help to develop new herbal-based therapeutics for various diseases.


Asunto(s)
Fitoquímicos/farmacología , Conformación Proteica/efectos de los fármacos , Animales , Humanos , Fitoterapia , Plantas Medicinales/química
17.
Nucleic Acids Res ; 47(4): 1637-1652, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30649466

RESUMEN

The DNA binding domain (DBD) of the tumor suppressor p53 is the site of several oncogenic mutations. A subset of these mutations lowers the unfolding temperature of the DBD. Unfolding leads to the exposure of a hydrophobic ß-strand and nucleates aggregation which results in pathologies through loss of function and dominant negative/gain of function effects. Inspired by the hypothesis that structural changes that are associated with events initiating unfolding in DBD are likely to present opportunities for inhibition, we investigate the dynamics of the wild type (WT) and some aggregating mutants through extensive all atom explicit solvent MD simulations. Simulations reveal differential conformational sampling between the WT and the mutants of a turn region (S6-S7) that is contiguous to a known aggregation-prone region (APR). The conformational properties of the S6-S7 turn appear to be modulated by a network of interacting residues. We speculate that changes that take place in this network as a result of the mutational stress result in the events that destabilize the DBD and initiate unfolding. These perturbations also result in the emergence of a novel pocket that appears to have druggable characteristics. FDA approved drugs are computationally screened against this pocket.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas Mutantes/química , Bibliotecas de Moléculas Pequeñas/química , Proteína p53 Supresora de Tumor/química , Proteínas de Unión al ADN/genética , Evaluación Preclínica de Medicamentos/métodos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Mutación/genética , Conformación Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Dominios Proteicos/genética , Desplegamiento Proteico/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
18.
Inorg Chem ; 57(21): 13533-13543, 2018 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-30345755

RESUMEN

Alzheimer's disease (AD) is a typical protein-misfolding disease. Aggregation of amyloid ß-peptide (Aß) plays a key role in the etiology of AD. The misfolding of Aß results in the formation of ß-sheet-rich aggregates and damages the function of neurons. A modified polyoxometalate (POM), [CoL(H2O)]2[CoL]2[HAsVMoV6MoVI6O40] [CAM, L = 2-(1 H-pyrazol-3-yl)pyridine], was designed to disaggregate the Aß aggregates, where L acts as an Aß-targeting group and POM as a conformational modulator. X-ray crystallography shows that CAM is composed of a ε-Keggin unit and four coordination units. CAM can disaggregate the ß-sheet-rich fibrils and metal-induced or self-aggregated Aß aggregates, and it further inhibits the production of ROS; as a result, it can protect the neurons from synaptic toxicity induced by Zn2+- or Cu2+-Aß aggregates or Aß self-aggregation. The mechanism of disaggregation involves a transformation of Aß conformation from ß-sheet to other conformers. The nature of the process is an interference of the ß-sheet conformation by CAM via hydrogen bonding. CAM specifically interacts with Aß aggregates but does not disturb the cerebral metal homeostasis and enzymatic systems. Molecular simulation suggests that the appropriate size of CAM and the cavity of ß-sheets facilitate the interaction between CAM and Aß aggregates; additionally, the H-bonding-favored amino acid residues in the cavity provide a precondition for the interaction. Moreover, CAM is lipophilic and capable of penetrating the blood-brain barrier, and it is metabolizable without causing an untoward effect to mice at high dosages. In view of the significant inhibitory effect on the Aß aggregation and related neurotoxicity, CAM represents a new type of leading compounds with a distinctive mechanism of action for the treatment of Alzheimer' disease. The conception of this study may be applied to other protein-misfolding diseases caused by conformational changes.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/química , Cobre/farmacología , Compuestos Organometálicos/farmacología , Pliegue de Proteína , Zinc/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Cobre/química , Simulación de Dinámica Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Compuestos Organometálicos/química , Células PC12 , Agregado de Proteínas/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Ratas , Zinc/química
19.
Int J Biol Macromol ; 120(Pt A): 1140-1148, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30179693

RESUMEN

The application of nanomaterials in the healthy system may induce some neurodegenerative diseases initiated by tau folding and neuronal cell death. Herein, aluminium oxide nanoparticles (Al2O3 NPs) were synthesized and characterized by XRD, TEM, DLS and zeta potential investigations. Afterwards, the interaction of Al2O3 NPs with tau protein was investigated by fluorescence and CD spectroscopic methods. The molecular docking and molecular dynamic were also run to explore the binding site and conformational changes of tau after interaction with Al2O3 cluster. Moreover, the MTT, LDH, caspase-9/-3 and flow cytometry assays were done to explore the Al2O3 NPs-induced cytotoxicity against SH-SY5Y cells. It was revealed that Al2O3 NPs bind to tau protein and form a static complex and fold the structure of tau toward a more packed structure. Molecular docking and molecular dynamic investigations revealed that NPs bind to the hydrophilic residues of the tau segments and promote some marginal structural folding of tau segment. The cellular assays displayed that Al2O3 NPs can elicit cell mortality through membrane leakage, caspase-9/-3 activations, and induction of both apoptosis and necrosis. This data may indicate that NPs can induce some adverse effects on the biological systems.


Asunto(s)
Óxido de Aluminio/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Neuroblastoma/tratamiento farmacológico , Proteínas tau/química , Óxido de Aluminio/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Nanopartículas del Metal/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neuroblastoma/química , Neuroblastoma/patología , Conformación Proteica/efectos de los fármacos , Proteínas tau/efectos de los fármacos
20.
Anal Chem ; 90(16): 10039-10048, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30041521

RESUMEN

This study reports on the real-time binding assessment between heavy metal ions and blood proteins immobilized onto nanoporous anodic alumina photonic crystals (NAA-PCs) by reflectometric interference spectroscopy (RIfS). The surface of NAA-PCs is chemically functionalized with γ-globulin (GG), transferrin (TFN), and serum albumin (HSA), the major proteins present in human blood plasma. Protein-modified NAA-PC platforms are exposed to analytical solutions of mercury ions of different concentrations. Dynamic changes in the effective optical thickness of protein-modified NAA-PCs in response to heavy metal ions are assessed in real time to evaluate the binding kinetics, affinity, and mechanism. Protein molecules undergo conformational changes upon exposure to mercury ions, with HSA exhibiting the strongest affinity. The combination of protein-modified NAA-PCs with RIfS allows real-time monitoring of protein-heavy metal ions interactions under dynamic flow conditions. This system is capable of detecting dynamic conformational changes in these proteins upon exposure to heavy metal ions. Our results provide new insights into these binding events, which could enable new methodologies to study the toxicity of heavy metal ions and other biomolecular interactions.


Asunto(s)
Óxido de Aluminio/química , Metales Pesados/metabolismo , Albúmina Sérica Humana/metabolismo , Transferrina/metabolismo , gammaglobulinas/metabolismo , Humanos , Porosidad , Unión Proteica , Conformación Proteica/efectos de los fármacos , Análisis Espectral/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA