Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Intervalo de año de publicación
1.
PLoS One ; 18(11): e0294511, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37972144

RESUMEN

Cardiovascular disorders are the world's major cause of death nowadays. To treat cardiovascular diseases especially coronary artery diseases and hypertension, researchers found potential ROCK2 (Rho-associated coiled-coil-containing protein kinase 2) target due to its substantial role in NO-cGMP and RhoA/ROCK pathway. Available drugs for ROCK2 are less effective and some of them depict side effects. Therefore, a set of novel compounds were screened that can potentially inhibit the activity of ROCK2 and help to treat cardiovascular diseases by employing In-silico techniques. In this study, we undertook ligand based virtual screening of 50 million compound's library, to that purpose shape and features (contain functional groups) based pharmacophore query was modelled and validated by Area Under Curve graph (AUC). 2000 best hits were screened for Lipinski's rule of 5 compliance. Subsequently, these selected compounds were docked into the binding site of ROCK2 to gain insights into the interactions between hit compounds and the target protein. Based on binding affinity and RMSD scores, a final cohort of 15 compounds were chosen which were further refined by pharmacokinetics, ADMET and bioactivity scores. 2 potential hits were screened using density functional theory, revealing remarkable biological and chemical activity. Potential inhibitors (F847-0007 and 9543495) underwent rigorous examination through MD Simulations and MMGBSA analysis, elucidating their stability and strong binding affinities. Results of current study unveil the potential of identified novel hits as promising lead compounds for ROCK2 associated with cardiovascular diseases. These findings will further investigate via In-vitro and In-vivo studies to develop novel druglike molecules against ROCK2.


Asunto(s)
Enfermedades Cardiovasculares , Simulación de Dinámica Molecular , Humanos , Simulación del Acoplamiento Molecular , Enfermedades Cardiovasculares/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento , Sitios de Unión , Quinasas Asociadas a rho
2.
J Biomol Struct Dyn ; : 1-14, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37814544

RESUMEN

Ostrinia furnacalis is a species of moth in the Crambidae family that is harmful to maize and other corn crops in Southeast Asia and the Western Pacific regions. Ostrinia furnacalis causes devastating losses to economically important corn fields. The ß-N-acetyl-D-hexosaminidase is an essential enzyme in O. furnacalis and its substrate binding +1 active site is different from that of the plants and humans ß-N-acetyl-D-hexosaminidases. To develop environment-friendly insecticides against OfHex1, we conducted structure-guided computational insecticide discovery to identify potential inhibitors that can bind the active site and inhibit the substrate binding and activity of the enzyme. We adopted a three-pronged strategy to conduct virtual screening using Glide and virtual screening workflow (VSW) in Schrödinger Suite-2022-3, against crystal structures of OfHex1 (PDB Id:3NSN), its homologue in humans (PDB Id: 1NP0) and Alphafold model of ß-N-acetyl-D-hexosaminidase from Trichogramma pretiosum, an egg parasitoid that protects the crops from O. furnacalis. A library of 20,313 commercially available and "insecticide-like" compounds was extracted from published literature. LigPrep enabled 44,943 ready-to-dock conformers generation. Glide docking revealed 18 OfHex1-specific hits that were absent in human and T. pretiosum screens. Reference docking was conducted using inhibitors/natural ligands in the crystal structures and hits with better docking scores than the reference were selected for MD simulations using Desmond to understand the stability of hit-target interactions. We noted five compounds that bound to OfHex1 TMX active-site based on their docking scores, consistent binding as noted by MD simulations and their insecticide/pesticide likeliness as noted by the Comprehensive Pesticide Likeness Analysis.Communicated by Ramaswamy H. Sarma.

3.
J Biomol Struct Dyn ; : 1-16, 2023 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-37545162

RESUMEN

The inhibition of Phosphodiesterase 5A (PDEA5) has the potential to modulate pulmonary arterial hypertension and cardiovascular diseases. Exploring the cross-reactivity of clinically available PDE5A therapeutics with PDE6A is intriguing in order to develop highly selective PDE5A compounds in cardiovascular arena. In the current study, we leveraged e-pharmacophore based screening and molecular dynamics (MD) simulation to discover more selective PDE5A inhibitors as compared to the PDE6A catalytic domain. e-Pharmacophore based mapping of the CoCoCo database (7 million compounds: ∼ 150,000,000 conformers), followed by Glide docking, MM-GBSA, and protein-inhibitor interaction analysis, revealed 1536427, 4832637 and 6788240 as stable, tight binders of PDE5A instead of PDE6A. These compounds adhere to Lipinski Rule of Five (RO5) and ADME/Tox criteria. MD simulations analysis showed that 1536427 stays stable and tightly binds to catalytic (Q-region) core of PDE5A catalytic domain as compared to sildenafil. Pronounced inward motions of the hydrophobic (H-region) and Lid region indicate the closure of PDE5A-1536427 complex, whereas this region in PDE6A-1536427 is more open. Significant differences in the interactions, stability, and dynamics of 1536427 were observed in the catalytic domain of PDE6A, demonstrating less specificity for PDE6A in comparison to PDE5A. After lead optimization and therapeutic interventions, this proposed lead may emerge as a promising PDE5A selective inhibitor.Communicated by Ramaswamy H. Sarma.

4.
J Biomol Struct Dyn ; 40(13): 6128-6150, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-33522438

RESUMEN

Modulating the activity of human soluble guanylate cyclase (hsGC) through allosteric regulation of the ßH-NOX domain has been considered as an immediate treatment for cardiovascular disorder (CVDs). Currently available ßH-NOX domain-specific agonists including cinaciguat are unable to deal with the conundrum raised due to oxidative stress in the case of CVDs and their associated comorbidities. Therefore, the idea of investigating novel compounds for allosteric regulation of hsGC activation has been rekindled to circumvent CVDs. Current study aims to identify novel ßH-NOX domain-specific compounds that can selectively turn on sGC functions by modulating the conformational dynamics of the target protein. Through a comprehensive computational drug-discovery approach, we first executed a target-based performance assessment of multiple docking (PLANTS, QVina, LeDock, Vinardo, Smina) scoring functions based on multiple performance metrices. QVina showed the highest capability of selecting true-positive ligands over false positives thus, used to screen 4.8 million ZINC15 compounds against ßH-NOX domain. The docked ligands were further probed in terms of contact footprint and pose reassessment through clustering analysis and PLANTS docking, respectively. Subsequently, energy-based AMBER rescoring of top 100 low-energy complexes, per-residue energy decomposition analysis, and ADME-Tox analysis yielded the top three compounds i.e. ZINC000098973660, ZINC001354120371, and ZINC000096022607. The impact of three selected ligands on the internal structural dynamics of the ßH-NOX domain was also investigated through molecular dynamics simulations. The study revealed potential electrostatic interactions for better conformational dialogue between ßH-NOX domain and allosteric ligands that are critical for the activation of hsGC as compared to the reference compound.


Asunto(s)
Enfermedades Cardiovasculares , Simulación de Dinámica Molecular , NADPH Oxidasas , Guanilil Ciclasa Soluble , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Ligandos , Simulación del Acoplamiento Molecular , NADPH Oxidasas/química , Unión Proteica , Guanilil Ciclasa Soluble/química
5.
J Biomol Struct Dyn ; 39(7): 2302-2317, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32299297

RESUMEN

The need of circumventing life-threatening cardiovascular disorders (CVDs) and pulmonary hypertension (PHT) worldwide prompts researchers to develop effective therapeutic agents. Crucial role of cyclic nucleotide phosphodiesterase-5 (PDE5A) and cyclic nucleotide phosphodiesterase-3 (PDE3A) in cardiovascular signaling makes them potential drug targets for the treatment of CVDs and PHT. In this study, one-drug-multiple-target strategy has been employed to screen inhibitors exhibiting dual specificity through Phase-generated and statistically validated e-pharmacophore models of PDE5A and PDE3A. An extensive CoCoCo database of 7 million compounds with ∼150,000,000 conformations was virtually screened by sequential e-pharmacophore mapping followed by Lipinski Rule of Five (RO5) evaluation and hierarchical docking simulations. Finally, docked hits were subjected to rigorous MMGBSA analysis to estimate the relative spatial affinity of the drug-like compounds. The hits (354 and 366 ligands against PDE5A and PDE3A, respectively) were further optimized through 2D clustering followed by a comprehensive 2D and 3D interaction analysis. Five structurally diverse hits mapped equally well with the e-pharmacophore models and showed promising inhibitory interactions with conserved four catalytic features of PDE5A and PDE3A, thus exhibiting dual specificity. Proposed lead compounds exhibited the lowest MMGBSA binding energies and were found to be in agreement with Lipinski Rule of Five (RO5) and ADME/Tox criteria as compared to sildenafil. The proposed dual inhibitors could thus provide promising outcomes for the discovery of dual as well as multipotent drug like compounds after lead optimization and primary therapeutic interventions.


Asunto(s)
Enfermedades Cardiovasculares , Inhibidores de Fosfodiesterasa 3 , Enfermedades Cardiovasculares/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 3/farmacología
6.
Molecules ; 25(16)2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32824118

RESUMEN

Pseudomonas aeruginosa is a Gram-negative pathogenic bacterium that is present commonly in soil and water and is responsible for causing septic shock, pneumonia, urinary tract and gastrointestinal infections, etc. The multi-drug resistance (MDR) phenomenon has increased dramatically in past years and is now considered a major threat globally, so there is an urgent need to develop new strategies to overcome drug resistance by P. aeruginosa. In P. aeruginosa, a major factor of drug resistance is associated to the formation of biofilms by the LasR enzyme, which regulates quorum sensing and has been reported as a new therapeutic target for designing novel antibacterial molecules. In this study, virtual screening and molecular docking were performed against the ligand binding domain (LBD) of LasR by employing a pharmacophore hypothesis for the screening of 2373 FDA-approved compounds to filter top-scoring hit compounds. Six inhibitors out of 2373 compounds were found to have binding affinities close to that of known LasR inhibitors. The binding modes of these compounds to the binding site in LasR-LBD were analyzed to identify the key interactions that contribute to the inhibition of LasR activity. Then, 50 ns simulations of top hit compounds were performed to elucidate the stability of their binding conformations with the LasR-LBD. This study, thus concluded that sulfamerazine showed the highest binding affinity for the LasR-LBD binding pocket exhibiting strong inhibitory binding interactions during molecular dynamics (MD) simulation.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Biopelículas/crecimiento & desarrollo , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Preparaciones Farmacéuticas/metabolismo , Pseudomonas aeruginosa/crecimiento & desarrollo , Transactivadores/antagonistas & inhibidores , Biopelículas/efectos de los fármacos , Simulación por Computador , Aprobación de Drogas , Reposicionamiento de Medicamentos , Simulación del Acoplamiento Molecular , Unión Proteica , Pseudomonas aeruginosa/efectos de los fármacos , Estados Unidos , United States Food and Drug Administration
7.
Comput Struct Biotechnol J ; 18: 1625-1638, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32670503

RESUMEN

Protein kinase Iα (PKGIα) is a pivotal cyclic guanosine monophosphate (cGMP) signalling protein. Major steps related to the structural plasticity of PKGIα have been inferred but the structural aspects of the auto-inhibition and multidomain tertiary organization of human PKGIα in active and inactive form are not clear. Here we combine computational comparative modelling, protein-protein docking and molecular dynamics (MD) simulations to investigate structural details of the repressed state of the catalytic domain of PKGIα. Exploration of the potential inhibitory conformation of the auto-inhibitory domain (AI) within the catalytic cleft reveals that the pseudo-substrate motif binds with residues of the glycine rich loop and substrate-binding lobe. Dynamic changes as a result of coupling of the catalytic and AI domains are also investigated. The three-dimensional homodimeric models of PKGIα in the active and inactive state indicate that PKGIα in its inactive-state attains a compact globular structure where cyclic nucleotide binding (CNB-A/B) domains are buried, whereas the catalytic domains are inaccessible with their substrate-binding pockets facing the N-terminal of CNB-A. Contrary to this, the active-state model of PKGIα shows an extended conformation where CNB-A/B domains are slightly rearranged and the catalytic domains of homodimer flanking the C-terminal with their substrate binding lobes free to entrap downstream proteins. These findings are consistent with previously reported static images of the multidomain organization of PKGIα. Structural insights pertaining to the conformational heterogeneity and auto-inhibition of PKGIα provided in this study may help to understand the dynamics-driven effective regulation of PKGIα.

8.
Sci Rep ; 10(1): 9488, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32528025

RESUMEN

In the nitric oxide (NO) signaling pathway, human soluble guanylate cyclase (hsGC) synthesizes cyclic guanosine monophosphate (cGMP); responsible for the regulation of cGMP-specific protein kinases (PKGs) and phosphodiesterases (PDEs). The crystal structure of the inactive hsGC cyclase dimer is known, but there is still a lack of information regarding the substrate-specific internal motions that are essential for the catalytic mechanism of the hsGC. In the current study, the hsGC cyclase heterodimer complexed with guanosine triphosphate (GTP) and cGMP was subjected to molecular dynamics simulations, to investigate the conformational dynamics that have functional implications on the catalytic activity of hsGC. Results revealed that in the GTP-bound complex of the hsGC heterodimer, helix 1 of subunit α (α:h1) moves slightly inwards and comes close to helix 4 of subunit ß (ß:h4). This conformational change brings loop 2 of subunit ß (ß:L2) closer to helix 2 of subunit α (α:h2). Likewise, loop 2 of subunit α (α:L2) comes closer to helix 2 of subunit ß (ß:h2). These structural events stabilize and lock GTP within the closed pocket for cyclization. In the cGMP-bound complex, α:L2 detaches from ß:h2 and establishes interactions with ß:L2, which results in the loss of global structure compactness. Furthermore, with the release of pyrophosphate, the interaction between α:h1 and ß:L2 weakens, abolishing the tight packing of the binding pocket. This study discusses the conformational changes induced by the binding of GTP and cGMP to the hsGC catalytic domain, valuable in designing new therapeutic strategies for the treatment of cardiovascular diseases.


Asunto(s)
Dominio Catalítico/fisiología , Guanilil Ciclasa Soluble/metabolismo , Sitios de Unión/fisiología , GMP Cíclico/metabolismo , Dimerización , Guanosina Trifosfato/metabolismo , Humanos , Óxido Nítrico/metabolismo , Unión Proteica/fisiología , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología
9.
World J Microbiol Biotechnol ; 36(1): 9, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31858269

RESUMEN

Campylobacter jejuni is the one of the leading cause of bacterial food borne gastroenteritis. PglB, a glycosyltransferase, plays a crucial role of mediating glycosylation of numerous periplasmic proteins. It catalyzes N-glycosylation at the sequon D/E-X1-N-X2-S/T in its substrate proteins. Here we report that the PglB itself is a glycoprotein which self-glycosylates at N534 site in its DYNQS sequon by its own catalytic WWDYG motif. Site-directed mutagenesis, lectin Immunoblot, and mobility shift assays confirmed that the DYNQS is an N-glycosylation motif. PglB's N-glycosylation motif is structurally and functionally similar to its widely studied glycosylation substrate, the OMPH1. Its DYNQS motif forms a solvent-exposed crest. This motif is close to a cluster of polar and hydrophilic residues, which form a loop flanked by two α helices. This arrangement extremely apposite for auto-glycosylation at N534. This self-glycosylation ability of PglB could mediate C. jejuni's ability to colonize the intestinal epithelium. Further this capability may also bear significance for the development of novel conjugated vaccines and diagnostic tests.


Asunto(s)
Campylobacter jejuni/enzimología , Glicoproteínas/química , Hexosiltransferasas/química , Proteínas de la Membrana/química , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Glicosilación , Hexosiltransferasas/genética , Hexosiltransferasas/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Filogenia , Conformación Proteica , Alineación de Secuencia , Análisis de Secuencia de Proteína , Vacunas
10.
J Biol Chem ; 294(32): 11980-11991, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31160323

RESUMEN

The reversible adenine phosphoribosyltransferase enzyme (APRT) is essential for purine homeostasis in prokaryotes and eukaryotes. In humans, APRT (hAPRT) is the only enzyme known to produce AMP in cells from dietary adenine. APRT can also process adenine analogs, which are involved in plant development or neuronal homeostasis. However, the molecular mechanism underlying substrate specificity of APRT and catalysis in both directions of the reaction remains poorly understood. Here we present the crystal structures of hAPRT complexed to three cellular nucleotide analogs (hypoxanthine, IMP, and GMP) that we compare with the phosphate-bound enzyme. We established that binding to hAPRT is substrate shape-specific in the forward reaction, whereas it is base-specific in the reverse reaction. Furthermore, a quantum mechanics/molecular mechanics (QM/MM) analysis suggests that the forward reaction is mainly a nucleophilic substitution of type 2 (SN2) with a mix of SN1-type molecular mechanism. Based on our structural analysis, a magnesium-assisted SN2-type mechanism would be involved in the reverse reaction. These results provide a framework for understanding the molecular mechanism and substrate discrimination in both directions by APRTs. This knowledge can play an instrumental role in the design of inhibitors, such as antiparasitic agents, or adenine-based substrates.


Asunto(s)
Adenina Fosforribosiltransferasa/metabolismo , Adenina/química , Adenina/metabolismo , Adenina Fosforribosiltransferasa/química , Biocatálisis , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Estructura Terciaria de Proteína , Teoría Cuántica , Especificidad por Sustrato
11.
J Mol Graph Model ; 90: 109-119, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31055154

RESUMEN

The human soluble Guanylate Cyclase (hsGC) is a heterodimeric heme-containing enzyme which regulates many important physiological processes. In eukaryotes, hsGC is the only known receptor for nitric oxide (NO) signaling. Improper NO signaling results in various disease conditions such as neurodegeneration, hypertension, stroke and erectile dysfunction. To understand the mechanisms of these diseases, structure determination of the hsGC dimer complex is crucial. However, so far all the attempts for the experimental structure determination of the protein were unsuccessful. The current study explores the possibility to model the quaternary structure of hsGC using a hybrid approach that combines state-of-the-art protein structure prediction tools with cryo-EM experimental data. The resultant 3D model shows close consistency with structural and functional insights extracted from biochemistry experiment data. Overall, the atomic-level complex structure determination of hsGC helps to unveil the inter-domain communication upon NO binding, which should be of important usefulness for elucidating the biological function of this important enzyme and for developing new treatments against the hsGC associated human diseases.


Asunto(s)
Óxido Nítrico/química , Óxido Nítrico/metabolismo , Guanilil Ciclasa Soluble/química , Guanilil Ciclasa Soluble/metabolismo , Secuencia de Aminoácidos , Simulación por Computador , Microscopía por Crioelectrón/métodos , Hemo/química , Hemo/metabolismo , Humanos , Alineación de Secuencia , Transducción de Señal/fisiología
12.
Comput Struct Biotechnol J ; 17: 378-389, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30962868

RESUMEN

In the cyclic guanosine monophosphate (cGMP) signaling pathway, phosphodiesterase 6 (PDE6) maintains a critical balance of the intracellular concentration of cGMP by catalyzing it to 5' guanosine monophosphate (5'-GMP). To gain insight into the mechanistic impacts of the PDE6 somatic mutations that are implicated in cancer and retinitis pigmentosa, we first defined the structure and organization of the human PDE6 heterodimer using computational comparative modelling. Each subunit of PDE6αß possesses three domains connected through long α-helices. The heterodimer model indicates that the two chains are likely related by a pseudo two-fold axis. The N-terminal region of each subunit is comprised of two allosteric cGMP-binding domains (Gaf-A & Gaf-B), oriented in the same way and interacting with the catalytic domain present at the C-terminal in a way that would allow the allosteric cGMP-binding domains to influence catalytic activity. Subsequently, we applied an integrated knowledge-driven in silico mutation analysis approach to understand the structural and functional implications of experimentally identified mutations that cause various cancers and retinitis pigmentosa, as well as computational saturation mutagenesis of the dimer interface and cGMP-binding residues of both Gaf-A, and the catalytic domains. We studied the impact of mutations on the stability of PDE6αß structure, subunit-interfaces and Gaf-cGMP interactions. Further, we discussed the changes in interatomic interactions of mutations that are destabilizing in Gaf-A (R93L, V141 M, F162 L), catalytic domain (D600N, F742 L, F776 L) and at the dimer interface (F426A, F248G, F424 N). This study establishes a possible link of change in PDE6αß structural stability to the experimentally observed disease phenotypes.

13.
Int J Mol Sci ; 20(3)2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30736292

RESUMEN

Soluble guanylate cyclase (sGC) regulates numerous physiological processes. The ß subunit Heme Nitric Oxide/Oxygen (HNOX) domain makes this protein sensitive to small gaseous ligands. The structural basis of the activation mechanism of sGC under the influence of ligands (NO, O2, CO) is poorly understood. We examine the effect of different ligands on the human sGC HNOX domain. HNOX systems with gaseous ligands were generated and explored using Molecular Dynamics (MD). The distance between heme Fe2+ and histidine in the NO-ligated HNOX (NO-HNOX) system is larger compared to the O2, CO systems. NO-HNOX rapidly adopts the conformation of the five-group metal coordination system. Loops α, ß, γ and helix-f exhibit increased mobility and different hydrogen bond networks in NO-HNOX compared to the other systems. The removal of His from the Fe coordination sphere in NO-HNOX is assisted by interaction of the imidazole ring with the surrounding residues which in turn leads to the release of signaling helix-f and activation of the sGC enzyme. Insights into the conformational dynamics of a human sGC HNOX domain, especially for regions which are functionally critical for signal transduction, are valuable in the understanding of cardiovascular diseases.


Asunto(s)
Hemo/química , Óxido Nítrico/química , Oxígeno/química , Hemo/metabolismo , Humanos , Enlace de Hidrógeno , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Unión Proteica , Guanilil Ciclasa Soluble/química , Guanilil Ciclasa Soluble/metabolismo
14.
Sci Rep ; 9(1): 1433, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723263

RESUMEN

Emergence of Dengue as one of the deadliest viral diseases prompts the need for development of effective therapeutic agents. Dengue virus (DV) exists in four different serotypes and infection caused by one serotype predisposes its host to another DV serotype heterotypic re-infection. We undertook virtual ligand screening (VLS) to filter compounds against DV that may inhibit inclusively all of its serotypes. Conserved non-structural DV protein targets such as NS1, NS3/NS2B and NS5, which play crucial role in viral replication, infection cycle and host interaction, were selected for screening of vital antiviral drug leads. A dataset of plant based natural antiviral derivatives was developed. Molecular docking was performed to estimate the spatial affinity of target compounds for the active sites of DV's NS1, NS3/NS2B and NS5 proteins. The drug likeliness of the screened compounds was followed by ADMET analysis whereas the binding behaviors were further elucidated through molecular dynamics (MD) simulation experiments. VLS screened three potential compounds including Canthin-6-one 9-O-beta-glucopyranoside, Kushenol W and Kushenol K which exhibited optimal binding with all the three conserved DV proteins. This study brings forth novel scaffolds against DV serotypes to serve as lead molecules for further optimization and drug development against all DV serotypes with equal effect against multiple disease causing DV proteins. We therefore anticipate that the insights given in the current study could be regarded valuable towards exploration and development of a broad-spectrum natural anti-dengue therapy.


Asunto(s)
Antivirales/química , Inhibidores Enzimáticos/química , Simulación del Acoplamiento Molecular , Fitoquímicos/química , Proteínas no Estructurales Virales/química , Antivirales/farmacología , Sitios de Unión , Virus del Dengue/enzimología , Virus del Dengue/genética , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Fitoquímicos/farmacología , Plantas Medicinales/química , Unión Proteica , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/química , ARN Helicasas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Serogrupo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo
15.
Pak J Pharm Sci ; 31(6): 2443-2451, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30473516

RESUMEN

Hypodermosis is a parasitic disease of cattle. The pathogenicity of the disease is attributed to Hypodermin proteins (Hypodermin A, Hypodermin B and Hypodermin C). Studies suggest that Hypodermin proteins may be defined as Serine proteases and collagenases. The structure of both proteases Hypodermin A and Hypodermin B were modeled using the Swiss-model server followed by its validation using Procheck, Errat and Verify-3D. Afterwards, both Hypodermin A and Hypodermin B were docked against collagen in order to study its interaction with respective Hypodermin proteins. The structure of both Hypodermin A and Hypodermin B showed more bent towards hydrophobic nature as more beta sheets were present in them. Both structures were also superimposed to check out similarities and differences present between them. Serine, Aspartic acid, Histidine, Glutamic acid and Lysine are found as interacting residues that are involved in hydrogen bonding with collagen. The interactions are found in the active domain region of Hypodermin proteins. The interacting residues were present in the active region of the hypodermin proteins thus validating the docking studies. This study may help in the drug development against hypodermosis with least side effects.


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Diseño Asistido por Computadora , Diseño de Fármacos , Hipodermosis/prevención & control , Simulación del Acoplamiento Molecular , Serina Endopeptidasas/química , Vacunas/química , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/parasitología , Hipodermosis/inmunología , Hipodermosis/parasitología , Conformación Proteica , Serina Endopeptidasas/inmunología , Serina Endopeptidasas/farmacología , Relación Estructura-Actividad , Vacunas/inmunología , Vacunas/farmacología
16.
Molecules ; 23(9)2018 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-30149624

RESUMEN

Soluble guanylate cyclase (sGC) is a key enzyme implicated in various physiological processes such as vasodilation, thrombosis and platelet aggregation. The enzyme's Heme-Nitric oxide/Oxygen (H-NOX) binding domain is the only sensor of nitric oxide (NO) in humans, which on binding with NO activates sGC to produce the second messenger cGMP. H-NOX is thus a hot target for drug design programs. BAY60-2770 and BAY58-2667 are two widely studied activators of sGC. Here we present comparative molecular dynamics studies to understand the molecular details characterizing the binding of BAY60-2770 and BAY58-2667 with the human H-NOX (hH-NOX) and bacterial H-NOX (bH-NOX) domains. HartreeFock method was used for parametrization of both the activators. A 50 ns molecular dynamics (MD) simulation was run to identify the functionally critical regions of the H-NOX domains. The CPPTRAJ module was used for analysis. BAY60-2770 on binding with bH-NOX, triggered rotational movement in signaling helix F and significant dynamicity in loops α and ß, but in hH-NOX domain the compound showed relatively lesser aforementioned structural fluctuations. Conversely, hH-NOX ligated BAY58-2667 experienced highest transitions in its helix F due to electrostatic interactions with D84, T85 and R88 residues which are not conserved in bH-NOX. These conformational transformations might be essential to communicate with downstream PAS, CC and cyclase domains of sGC. Comparative MD studies revealed that BAY bound bHNOX dynamics varied from that of hH-NOX, plausibly due to some key residues such as R40, F74 and Y112 which are not conserved in bacteria. These findings will help to the design of novel drug leads to cure diseases associated to human sGC.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hemo/química , Óxido Nítrico/química , Oxígeno/química , Dominios y Motivos de Interacción de Proteínas , Guanilil Ciclasa Soluble/antagonistas & inhibidores , Guanilil Ciclasa Soluble/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Humanos , Enlace de Hidrógeno , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Guanilil Ciclasa Soluble/metabolismo
17.
EXCLI J ; 17: 169-180, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29743855

RESUMEN

This study reports three novel sulfonamide derivatives 4-Chloro-N-[(4-methylphenyl) sulphonyl]-N-propyl benzamide (1A), N-(2-hydroxyphenyl)-4-methyl benzene sulfonamide (1B) and 4-methyl-N-(2-nitrophenyl) benzene sulfonamide (1C). The compounds were synthesised from starting material 4-methylbenzenesulfonyl chloride and their structure was studied through 1H-NMR and 13C-NMR spectra. Computational docking was performed to estimate their binding energy against bacterial p-amino benzoic acid (PABA) receptor, the dihydropteroate synthase (DHPS). The derivatives were tested in vitro for their antimicrobial activity against Gram+ and Gram- bacteria including E. coli, B. subtilis, B. licheniformis and B. linen. 1A was found active only against B. linen; 1B was effective against E. coli, B. subtilis and B. linen whereas 1C showed activity against E. coli, B. licheniformis and B. linen. 1C showed maximum activity with minimum inhibitory concentration (MIC) of 50, 100 and 150 µg/mL against E. coli, B. licheniformis and B. linen respectively. 1C exhibited maximum affinity to DHPS with binding free energy of -8.1 kcal/mol. It enriched in the top 0.5 % of a library of 7663 compounds, ranked in order of their binding affinity against DHPS. 1C was followed by 1B which showed a moderate to low level MIC of 100, 250 and 150 µg/mL against E. coli, B. subtilis and B. linen respectively, whereas 1A showed a moderate level MIC of 100 µg/mL but only against B. linen. These derivatives may thus serve as potential anti-bacterial alternatives against resistant pathogens.

18.
Cell Chem Biol ; 25(6): 666-676.e4, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29576532

RESUMEN

Phosphoribosyltransferases catalyze the displacement of a PRPP α-1'-pyrophosphate to a nitrogen-containing nucleobase. How they control the balance of substrates/products binding and activities is poorly understood. Here, we investigated the human adenine phosphoribosyltransferase (hAPRT) that produces AMP in the purine salvage pathway. We show that a single oxygen atom from the Tyr105 side chain is responsible for selecting the active conformation of the 12 amino acid long catalytic loop. Using in vitro, cellular, and in crystallo approaches, we demonstrated that Tyr105 is key for the fine-tuning of the kinetic activity efficiencies of the forward and reverse reactions. Together, our results reveal an evolutionary pressure on the strictly conserved Tyr105 and on the dynamic motion of the flexible loop in phosphoribosyltransferases that is essential for purine biosynthesis in cells. These data also provide the framework for designing novel adenine derivatives that could modulate, through hAPRT, diseases-involved cellular pathways.


Asunto(s)
Adenina Fosforribosiltransferasa/metabolismo , Adenina Fosforribosiltransferasa/química , Adenina Fosforribosiltransferasa/aislamiento & purificación , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica
19.
Microbiol Res ; 174: 56-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25946329

RESUMEN

Bordetella pertussis, the causative agent of whooping cough, attaches to mucosal surface in upper respiratory tract, where it produces a variety of surface associated and secreted autotransporter molecules among others. In this study we have cloned newly identified member of autotransporter family BapC (B. pertussis autotransporter protein C); expressed it in Escherichia coli and characterized it for its different properties. We have also raised antisera to BapC protein; the antisera were used in immunofluorescence assay to determine the surface association of the protein. Results suggest that BapC in B. pertussis Taberman parent is surface exposed when compared with the respective BapC mutant. The neutralizing effect of anti-BapC serum was also evaluated in the presence of active complement proteins and results suggest that antiserum can potentiate the killing of B. pertussis cells in the presence of added source of complement. Structure of the protein was also studied, both α and ß domains of the protein were modeled, ß domain exhibits typical transmembrane ß-barrel porin topology whereas α domain behaves as a characteristic bacterial autotransporter passenger domain.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bordetella pertussis/genética , Bordetella pertussis/fisiología , Sistemas de Secreción Tipo V/genética , Sistemas de Secreción Tipo V/metabolismo , Proteínas Bacterianas/química , Actividad Bactericida de la Sangre , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Modelos Moleculares , Conformación Proteica , Sistemas de Secreción Tipo V/química
20.
Int J Anal Chem ; 2015: 164974, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25838824

RESUMEN

Currently, the clinical use of sweat as biofluid is limited. The collection of sweat and its analysis for determining ethanol, drugs, ions, and metals have been encompassed in this review article to assess the merits of sweat compared to other biofluids, for example, blood or urine. Moreover, sweat comprises various biomarkers of different diseases including cystic fibrosis and diabetes. Additionally, the normalization of sampled volume of sweat is also necessary for getting efficient and useful results.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...