Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Chem Biodivers ; 18(2): e2000863, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33411971

RESUMEN

Glucokinase (GLK) and Hexokinase (HK) have been characterized as essential targets in Trypanosoma cruzi (Tc)-mediated infection. A recent study reported the propensity of the concomitant inhibition of TcGLK and TcHK by compounds GLK2-003 and GLK2-004, thereby presenting an efficient approach in Chagas disease treatment. We investigated this possibility using atomic and molecular scaling methods. Sequence alignment of TcGLK and TcHK revealed that both proteins shared approximately 33.3 % homology in their glucose/inhibitor binding sites. The total binding free energies of GLK2-003 and GLK2-004 were favorable in both proteins. PRO92 and THR185 were pivotal to the binding and stabilization of the ligands in TcGLK, likewise their conserved counterparts, PRO163 and THR237 in TcHK. Both compounds also induced a similar pattern of perturbations in both TcGLK and TcHK secondary structure. Findings from this study therefore provide insights into the underlying mechanisms of dual inhibition exhibited by the compounds. These results can pave way to discover and optimize novel dual Tc inhibitors with favorable pharmacokinetics properties eventuating in the mitigation of Chagas disease.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glucoquinasa/antagonistas & inhibidores , Hexoquinasa/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma cruzi/enzimología , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Inhibidores Enzimáticos/química , Glucoquinasa/química , Glucoquinasa/metabolismo , Hexoquinasa/química , Hexoquinasa/metabolismo , Humanos , Modelos Moleculares , Termodinámica , Tripanocidas/química , Trypanosoma cruzi/efectos de los fármacos
2.
Chem Biodivers ; 18(9): e2100204, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34252268

RESUMEN

Parkinson's disease (PD) is one of the most targeted neurodegenerative diseases in clinical research. Awareness of research is due to its increasing number of affected people worldwide. The pathology of PD has been linked to several key proteins upregulation such as the catechol O-Methyltransferase (COMT). Hence, the synthesis of compounds possessing inhibitory capacity has been the frontline of research in recent years. Several compounds have been synthesized among which is the nitrocatechol. However, major limitations associated with the nitrocatechol scaffold include the inability to possess adequate CNS penetration properties and hepatic toxicity associated with the compounds. However, a series of bicyclic hydroxypyridones compounds were synthesized to evaluate their inhibitory potentials on COMT protein with compound 38 (c38) 2-[(2,4-dichlorophenyl)methyl]-7-hydroxy-1,2,3,4-tetrahydro-8H-pyrido[1,2-a]pyrazin-8-one shown to have a 40 fold increase level coverage in its IC50 over brain exposure when compared to the other synthesized compound. The molecular dynamics method was employed to understand the nature of interaction exhibited by c38. Molecular mechanics of c38 revealed a disruptive effect on the secondary structure of COMT protein. Per residue decomposition analysis revealed similar crucial residues involved in the favorable binding of c38 and tolcapone implicated its increased inhibitory capacity on COMT in preventing PD. Free binding energy (ΔGbind ) of c38 further revealed the inhibitory capacity towards COMT protein in comparison to the FDA approved tolcapone. Ligand mobility analysis of both compounds showed a timewise different mobility pattern across the simulation time frame at the active site pocket of the protein connoting the different inhibitory potency exhibited by c38 and tolcapone. Findings from this study revealed optimization of c38 could facilitate the discovery of new compounds with enhanced inhibitory properties towards COMT in treating PD.


Asunto(s)
Antiparkinsonianos/farmacología , Inhibidores de Catecol O-Metiltransferasa/farmacología , Catecol O-Metiltransferasa/metabolismo , Simulación de Dinámica Molecular , Enfermedad de Parkinson/tratamiento farmacológico , Antiparkinsonianos/química , Inhibidores de Catecol O-Metiltransferasa/química , Humanos , Estructura Molecular , Enfermedad de Parkinson/metabolismo , Termodinámica
3.
Chem Biodivers ; 18(1): e2000802, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33289285

RESUMEN

Numerous studies have established the involvement of Poly (ADP-ribose) Polymerase-1 (PARP-1) in cancer presenting it as an important therapeutic target over recent years. Although homology among the PARP protein family makes selective targeting difficult, two compounds [d11 (0.939 µM) and d21 (0.047 µM)] with disparate inhibitory potencies against PARP-1 were recently identified. In this study, free energy calculations and molecular simulations were used to decipher underlying mechanisms of differential PARP-1 inhibition exhibited by the two compounds. The thermodynamics calculation revealed that compound d21 had a relatively higher ΔGbind than d11. High involvement of van der Waal and electrostatic effects potentiated the affinity of d21 at PARP-1 active site. More so, incorporated methyl moiety in d11 accounted for steric hindrance which, in turn, prevented complementary interactions of key site residues such as TYR889, MET890, TYR896, TYR907. Conformational studies also revealed that d21 is more stabilized for interactions in the active site compared to d11. We believe that findings from this study would provide an important avenue for the development of selective PARP-1 inhibitors.


Asunto(s)
Azepinas/química , Oxadiazoles/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Azepinas/metabolismo , Sitios de Unión , Dominio Catalítico , Halógenos/química , Humanos , Simulación de Dinámica Molecular , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Análisis de Componente Principal , Electricidad Estática , Termodinámica
4.
Chem Biodivers ; 17(3): e1900548, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32034875

RESUMEN

Chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) has been involved in several inflammation dependent diseases by mediating the chemotaxis of pro-inflammatory cells in response to allergy and other responses through PGD2 ligation. This CRTH2-PGD2 signaling pathway has become a target for treating allergic and type 2 inflammation dependent diseases, with many inhibitors developed to target the PGD2 binding pocket. One of such inhibitors is the ramatroban analog, CT-133, which exhibited therapeutic potency cigarette smoke-induced acute lung injury in patients. Nonetheless, the molecular mechanism and structural dynamics that accounts for its therapeutic prowess remain unclear. Employing computational tools, this study revealed that although the carboxylate moiety in CT-133 and the native agonist PGD2 aided in their stability within the CRTH2 binding pocket, the tetrahydrocarbazole group of CT-133 engaged in strong interactions with binding pocket residues which could have formed as the basis of the antagonistic advantage of CT-133. Tetrahydrocarbazole group interactions also enhanced the relative stability CT-133 within the binding pocket which consequently favored CT-133 binding affinity. CT-133 binding also induced an inactive or 'desensitized' state in the helix 8 of CRTH2 which could conversely favor the recruitment of arrestin. These revelations would aid in the speedy development of small molecule inhibitors of CRTH2 in the treatment of type 2 inflammation dependent diseases.


Asunto(s)
Ácidos Borónicos/farmacología , Inflamación/tratamiento farmacológico , Lípidos/química , Simulación de Dinámica Molecular , Prostaglandina D2/agonistas , Ácidos Borónicos/química , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Relación Estructura-Actividad , Células Th2
5.
Molecules ; 25(18)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947886

RESUMEN

Mycobacterium tuberculosis (Mtb) serine/threonine (Ser/Thr) Protein kinases A (PknA) and B (PknB) have been identified as highly attractive targets for overcoming drug resistant tuberculosis. A recent lead series optimization study yielded compound 33 which exhibited potencies ~1000 times higher than compound 57. This huge discrepancy left us curious to investigate the mechanistic 'dual' (in)activities of the compound using computational methods, as carried out in this study. Findings revealed that 33 stabilized the PknA and B conformations and reduced their structural activities relative to 57. Optimal stability of 33 in the hydrophobic pockets further induced systemic alterations at the P-loops, catalytic loops, helix Cs and DFG motifs of PknA and B. Comparatively, 57 was more surface-bound with highly unstable motions. Furthermore, 33 demonstrated similar binding patterns in PknA and B, involving conserved residues of their binding pockets. Both π and hydrogen interactions played crucial roles in the binding of 33, which altogether culminated in high ΔGs for both proteins. On the contrary, the binding of 57 was characterized by unfavorable interactions with possible repulsive effects on its optimal dual binding to both proteins, as evidenced by the relatively lowered ΔGs. These findings would significantly contribute to the rational structure-based design of novel and highly selective dual inhibitors of Mtb PknA and B.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Mycobacterium tuberculosis/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/química , Secuencia de Aminoácidos , Proteínas Bacterianas/antagonistas & inhibidores , Sitios de Unión , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Diseño de Fármacos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Análisis de Componente Principal , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Teoría Cuántica , Quinazolinas/metabolismo , Alineación de Secuencia , Termodinámica
6.
J Cell Biochem ; 120(1): 951-966, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30160791

RESUMEN

The DNA-binding ability of p53 represents the crux of its tumor suppressive activities, which involves transcriptional activation of target genes responsible for apoptosis and cell-cycle arrest. Mutational occurrences within or in close proximity to the DNA-binding surface of p53 have accounted for the loss of direct DNA-binding ability and inactivation implicated in many cases of cancer. Moreover, the design of therapeutic compounds that can restore DNA-binding ability in p53 mutants has been identified as a way forward in curtailing their oncogenic activities. However, there is still the need for more insights into evaluate the perturbations that occur at the DNA-binding interface of mp53 relative to DNA-binding loss, inactivation, and design of potent reactivators, hence the purpose of this study. Therefore, we evaluated p53-structural (R175H) and contact (R273C) mutational effects using tunnel perturbation analysis and other computational tools. We identified significant perturbations in the active tunnels of p53, which resulted in altered geometry and loss, unlike in the wild-type p53. This corroborated with structural, DNA-binding, and interaction network analysis, which showed that loss of flexibility, repulsion of DNA-interactive residues, and instability occurred at the binding interface of both mutants. Also, these mutations altered bonding interactions and network topology at the DNA-binding interface, resulting in the reduction of p53-DNA binding proximity and affinity. Therefore, these findings would aid the structure-based design of novel chemical entities capable of restoring p53-DNA binding and activation.


Asunto(s)
Diseño de Fármacos , Mutación , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Acetilación , ADN/química , Genes Supresores de Tumor , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas Mutantes/química , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional/genética , Estructura Secundaria de Proteína , Activación Transcripcional/genética
7.
J Cell Biochem ; 120(9): 16108-16119, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31125144

RESUMEN

Therapeutic targeting of the adenosine triphosphate (ATP) machinery of Mycobacterium tuberculosis (Mtb) has recently presented a potent and alternative measure to halt the pathogenesis of tuberculosis. This has been potentiated by the development of bedaquiline (BDQ), a novel small molecule inhibitor that selectively inhibits mycobacterial F1 Fo -ATP synthase by targeting its rotor c-ring, resulting in the disruption of ATP synthesis and consequential cell death. Although the structural resolution of the mycobacterial C9 ring in co`mplex with BDQ provided the first-hand detail of BDQ interaction at the c-ring region of the ATP synthase, there still remains a need to obtain essential and dynamic insights into the mechanistic activity of this drug molecule towards crucial survival machinery of Mtb. As such, for the first time, we report an atomistic model to describe the structural dynamics that explicate the experimentally reported antagonistic features of BDQ in halting ion shuttling by the mycobacterial c-ring, using molecular dynamics simulation and the Molecular Mechanics/Poisson-Boltzmann Surface Area methods. Results showed that BDQ exhibited a considerably high ΔG while it specifically maintained high-affinity interactions with Glu65B and Asp32B , blocking their crucial roles in proton binding and shuttling, which is required for ATP synthesis. Moreover, the bulky nature of BDQ induced a rigid and compact conformation of the rotor c-ring, which impedes the essential rotatory motion that drives ion exchange and shuttling. In addition, the binding affinity of a BDQ molecule was considerably increased by the complementary binding of another BDQ molecule, which indicates that an increase in BDQ molecule enhances inhibitory potency against Mtb ATP synthase. Taken together, findings provide atomistic perspectives into the inhibitory mechanisms of BDQ coupled with insights that could enhance the structure-based design of novel ATP synthase inhibitors towards the treatment of tuberculosis.


Asunto(s)
Antituberculosos/farmacología , Diarilquinolinas/farmacología , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mycobacterium tuberculosis/enzimología , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Sinergismo Farmacológico , Modelos Moleculares , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Conformación Proteica , Dominios Proteicos
8.
Org Biomol Chem ; 17(5): 1176-1190, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30644960

RESUMEN

The influence of chirality on the therapeutic activities of drug molecules has remained an interesting subject matter in drug design. The recent identification of two chiral irreversible inhibitors with differential inhibitory activities towards oncogenic fibroblast growth factor receptor 4 (FGFR4) presented an avenue to investigate the underlying mechanisms that accounted for their disparate activities. Accordingly, the S-configured form (9g) exhibited '15 times' potency than the corresponding R-configured (9h) form. Nonetheless, the big question remains how does chirality influence their inhibitory potencies? Therefore, in this study, we seek to provide useful insights into this interesting phenomenon using molecular dynamics simulations and free binding energy calculations. Interestingly, we observed that the inhibitory 9g activity correlates with a coordinated movement of the active site p-loop, as specifically induced by the S-configuration, which allowed the rotation of three dihedral angles; φ1(CNCO), φ2(CCC*N) and φ3(CCCC), thereby achieving optimal orientations suitable for interactions with crucial active site residues such as LEU473, LYS503, ASP641 and TYR643. Consequentially, while the 9h-bound FGFR4 active site was highly unstable, 9g exerted an inward pulling effect which accounted for active site stability and compactness. Also, the positional movement of 9h (R-configuration) at the active site was restricted, thereby preventing interactions with key residues. Moreover, 9g exhibited the most favorable binding as compared to 9h which showed a relatively lower ΔGbind. The higher binding affinity of 9g to FGFR4 can be mainly attributed to the increase in van der Waals energy by -4.12 kcal mol-1 and electrostatic by -2.89 kcal mol-1. The difference in van der Waals interactions is mainly determined by two residues; ASP641 and TYR643, whilst, the difference in electrostatic interactions is primarily determined by two residues LEU473 and LYS503.

9.
Phys Chem Chem Phys ; 21(27): 15120-15132, 2019 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-31241637

RESUMEN

The concept of chirality has become prominent over the years, particularly with regards to the design of therapeutic molecules. This phenomenon was recently reported for pro-carcinogenic fibroblast growth factor receptor 1 (FGFR1), wherein two inhibitors exhibited disparate inhibitory potencies due to the effects of chirality. Therefore, the ability of the R-enantiomer (R-21c) to possess a potency 10.44 times that of the S-enantiomer (S-21c) leaves us with a curiosity to investigate the underlying mechanisms using computational methods. Hence, presented in this study are insights that clearly explain the systematic effects of chirality on the differential activities of (R)-21c and (S)-21c towards FGFR1. The findings showed that the "R-configured" (R)-21c induced a notable conformational change in the active site P-loop, which enhanced its motion, as complemented by rotation of two dihedral angles: φ1(CNCC) and φ2(CC*OC), providing a favorable orientation. Likewise, optimal positioning of (R)-21c at the binding cavity allowed adequate interspaces that facilitated the formation of strong interactions with target residues. Moreover, the estimated ΔG binding correlated with bioactivity data (IC50) and, when decomposed, we observed that van der Waals (vdW) interactions were the major highlight of the binding process of both 21c enantiomers and also accounted for their differential activities. Active site interactions of (R)-21c with residues Phe489 and Arg629 stabilized its two benzimidazole motifs, while Arg570 and Pro663 formed two strong NH-N hydrogen bonds and one π-alkyl interaction, which altogether accounted for its inhibitory prowess towards FGFR1. In contrast, these interactions were not observed in (S)-21c due to its non-flexible S-configuration, which disallowed its extension into the active site region and prevented interaction with crucial residues. These results are expected to facilitate the discovery and rational design of novel and specific FGFR1 inhibitors.


Asunto(s)
Indazoles/química , Indazoles/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Concentración 50 Inhibidora , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
10.
Biotechnol Lett ; 41(1): 115-128, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30377869

RESUMEN

OBJECTIVE: Bacterial RNA polymerase (bRNAP) represent a crucial target for curtailing microbial activity but its structural and sequence similarities with human RNA polymerase II (hRNAPII) makes it difficult to target. Recently, Pseudouridimycin (PUM), a novel nucleoside analogue was reported to selectively inhibit bRNAP and not hRNAP. Till date, underlying mechanisms of PUM selectivity remains unresolved, hence the aim of this study. RESULTS: Using sequence alignment method, we observed that the ß' of bRNAP and the RPB1 subunits of hRNAPII were highly conserved while the ß and RPB2 subunits of both proteins were also characterized by high sequence variations. Furthermore, the impact of these variations on the differential binding of PUM was evaluated using MMPB/SA binding free energy and per-residue decomposition analysis. These revealed that PUM binds better to bRNAP than hRNAP with prominent bRNAP active site residues that contributed the most to PUM binding and stabilization lacking in hRNAPII active site due to positional substitution. Also, the binding of PUM to hRNAP was characterized by the formation of unfavorable interactions. In addition, PUM assumed favorable orientations that possibly enhanced its mobility towards the hydrophobic core region of bRNAP. On the contrary, unfavorable intramolecular interactions characterize PUM orientations at the binding site of hRNAPII, which could restrict its movement due to electrostatic repulsions. CONCLUSION: These findings would enhance the design of potent and selective drugs for broad-spectrum antimicrobial activity.


Asunto(s)
Proteínas Bacterianas/química , Nucleósidos/análogos & derivados , ARN Polimerasa II/química , Alineación de Secuencia , Análisis de Secuencia de Proteína , Proteínas Bacterianas/genética , Dominio Catalítico , Humanos , Nucleósidos/química , ARN Polimerasa II/genética
11.
Chem Biodivers ; 16(12): e1900365, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31589372

RESUMEN

The quest for reliable dihydroorotate dehydrogenase (DHODH) inhibitors has engendered the discovery of potential therapeutic compounds at different stages of clinical trials. Although promising, high attrition rates and unfavorable bioactivities have limited their drug developmental progress. A recent structural modification of DSM265, a triazolopyrimidine-based inhibitor, yielded DSM421, derived by the substitution of the SF5 -aniline group on DSM265 with a CF3 -pyridinyl moiety. Consequently, DSM421 exhibited improved pharmacological and pharmacokinetics attributes relative to DSM265. The improved bioactivity mediated by the CF3 -pyridinyl group leaves us with a curiosity to investigate underlying ligand-binding mechanisms and dynamics using computational methods. Presented in this study are insights that clearly explain the effects of structural SF5 -aniline→CF3 -pyridinyl modifications on pfDHODH inhibition. Findings showed that the CF3 -pyridinyl group induced an optimal and stabilized positioning of DSM421 within the binding pocket, allowing for steady and strong intermolecular interactions which favored its stronger binding affinity as estimated and correlated with bioactivity data. These interactions consequently induced a pronounced stabilization of the structural conformation of pfDHODH by restricting residue motions, which possibly underpinned its enhanced inhibitory activity relative to DSM265. Active site interactions of the CF3 -pyrinidyl group with residues Ser236, Ile237, and Phe188 characterized by strong π-π stacking and halogen interactions also stabilized its positioning which altogether accounted for its enhanced inhibitory prowess towards pfDHODH. On the contrary, fewer and weaker interactions characterized DSM265 binding which could explain its relatively lower binding affinity. Findings will facilitate the design of novel pfDHODH inhibitors with enhanced properties.


Asunto(s)
Antimaláricos/química , Inhibidores Enzimáticos/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Plasmodium falciparum/enzimología , Proteínas Protozoarias/química , Purinas/química , Piridinas/química , Antimaláricos/metabolismo , Sitios de Unión , Dominio Catalítico , Dihidroorotato Deshidrogenasa , Inhibidores Enzimáticos/metabolismo , Simulación de Dinámica Molecular , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Proteínas Protozoarias/metabolismo , Purinas/metabolismo , Teoría Cuántica , Termodinámica
12.
Chem Biodivers ; 16(6): e1900085, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30990952

RESUMEN

The discovery of J147 represented a significant milestone in the treatment of age-related disorders, which was further augmented by the recent identification of mitochondrial ATP synthase as the therapeutic target. However, the underlying molecular events associated with the modulatory activity of J147 have remained unresolved till date. Herein, we present, for the first time, a dynamical approach to investigate the allosteric regulation of mATP synthase by J147, using a reliable human αÎ³ß protein model. The highlight of our findings is the existence of the J147-bound protein in distinct structural associations at different MD simulation periods coupled with concurrent open↔close transitions of the ß catalytic and α allosteric (ATP5A) sites as defined by Cα distances (d), TriCα (Θ) and dihedral (φ) angular parameters. Firstly, there was an initial pairing of the αγ subunits away from the ß subunit followed by the formation of the 'non-catalytic' αß pair at a distance from the γ subunit. Interestingly, J147-induced structural arrangements were accompanied by the systematic transition of the ß catalytic site from a closed to an open state, while there was a concurrent transition of the allosteric site from an open αE conformation to a closed state. Consequentially, J147 reduced the structural activity of the whole αÎ³ß complex, while the unbound system exhibited high atomistic deviations and structural flexibility. Furthermore, J147 exhibited favorable binding at the allosteric site of mATP synthase with considerable electrostatic energy contributions from Gln215, Gly217, Thr219, Asp312, Asp313, Glu371 and Arg406. These findings provide details on the possible effects of J147 on mitochondrial bioenergetics, which could facilitate the structure-based design of novel small-molecule modulators of mATP synthase in the management of Alzheimer's disease and other neurodegenerative disorders.


Asunto(s)
Curcumina/análogos & derivados , Hidrazinas/química , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Regulación Alostérica , Enfermedad de Alzheimer/tratamiento farmacológico , Sitios de Unión , Dominio Catalítico , Curcumina/farmacología , Humanos , Hidrazinas/metabolismo , Hidrazinas/uso terapéutico , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/química , Simulación del Acoplamiento Molecular , Análisis de Componente Principal , Electricidad Estática , Termodinámica
13.
J Cell Biochem ; 119(3): 2646-2652, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29058783

RESUMEN

Various evidence has revealed that mutations in p53 exert activities that go beyond simply inactivation of wildtype functions but rather elicits downstream interactions that promote malignancy described as mutant p53 gain-of-function (GOF). Here we report the first account of the dynamics of mutation-induced structural transition of native p53 to an aberrant gain-of-function state, studying the wildtype (WT) and high incidence contact (R273C) and structural (R175H) mutant p53 (mutp53) through molecular dynamics simulation. Result analysis revealed that both mutants exhibited structural distortion and reduced flexibility, indicative of rigidity and kinetic stability. In addition, surface analysis revealed an increase in the accessible surface area in the p53 mutants. This suggests that the GOF transition involves protein unfolding and exposure of buried hydrophobic surface essential for interaction with HSF-1 oncogenic partner and wildtype p63, and p73 homologs. Further validation revealed binding cavities, similar in the mutants but dissimilar to the WT. Taken together, this study complements experimental findings and reveals the interplay between mutation-induced structural distortion, loss of flexibility, rigidity, enhanced stability, protein unfolding and ultimately, exposure of binding surfaces as conformational attributes that characterize mutP53 structure-GOF activities. This insight is, therefore, of great importance as it opens up a novel therapeutic approach toward the structure based targeting of mutP53 oncogenic involvement beyond wildtype inactivation. Furthermore, "exposed" binding site information obtained from this study can be explored for structure-based design of substances best described as "destabilizers" to disrupt the GOF interaction of mutp53.


Asunto(s)
Carcinogénesis/genética , Mutación con Ganancia de Función/genética , Proteína p53 Supresora de Tumor/genética , Animales , Humanos , Simulación de Dinámica Molecular , Conformación Proteica , Proteína p53 Supresora de Tumor/química
14.
J Cell Biochem ; 119(11): 9364-9379, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30129224

RESUMEN

Aggregatibacter actinomycetemcomitans is a Gram-negative bacteria that has gained wide recognition for its causative role in the development of various immune diseases, which includes localized aggressive periodontitis. Its ability to evade host defense mechanisms is mediated by the secretion of leukotoxin (LtxA), which induces death of white blood cells (leukocytes) by specific binding to their surface-expressed leukocyte function-associated receptor (LFA-1) in its active state. Therapeutic compounds that interfere with this pathogenic process and abrogate A. actinomycetemcomitans virulence have been reported in literature. These include doxycycline, and more recently phytochemical compounds such as hamamelitanin, resveratrol, naringin, and quercetin. However, the question remains how do they work? Therefore, with the aid of computational tools, we explore the molecular mechanisms by which they possibly elicit their therapeutic functions. Molecular mechanics Poisson/Boltzmann surface area analyses revealed that these compounds bind favorably to active LFA-1 with high affinity and considerable stability, indicative of their ability to occupy the LtxA binding site (LBS) and prevent LtxA binding. The conformational transition of open LFA-1 to its closed state further describe the mechanistic activity of these compounds. In addition to notable reductions in structural mobility and flexibility, the burial of surface-exposed interactive side chains at the LBS was observed, an occurrence that could alter the complementary binding of LtxA. It is also important to mention that these occurrences were induced more prominently by the phytochemicals. We believe that these findings will enhance the scope of drug design and discovery for potent LtxA antagonists with improved activities and therapeutic efficacies in the treatment of virulent A. actinomycetemcomitans diseases.


Asunto(s)
Exotoxinas/metabolismo , Enfermedades Periodontales/metabolismo , Animales , Conjuntos de Datos como Asunto , Exotoxinas/química , Humanos , Enfermedades Periodontales/genética , Unión Proteica , Estructura Secundaria de Proteína , Termodinámica
15.
Chem Biodivers ; 15(12): e1800437, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30290062

RESUMEN

The collapsin response mediator protein (CRMP-2) is hyperphosphorylated in Alzheimer's disease (AD). These phosphorylation events are mediated by specific kinase proteins, GSK3ß and Cdk5, and occur at target phosphorylation sites majorly located at the C-terminal tail of CRMP-2. The abilities of naringenin (NAR) and naringenin-7-O-glucuronide (NAR-7-O-G) to selectively bind CRMP-2 and reduce its phosphorylation have been previously demonstrated; the molecular interplay between these events remains unresolved. Using computational tools, we unravel the possible mechanisms by which these molecules disrupt CRMP-2 phosphorylation. Structural and dynamic analyses revealed that while the C-terminal tail of unbound CRMP-2 was extended and subtly organized, notable conformational disarray and rigidity characterized this region when bound by NAR and NAR-7-O-G. Consequentially, atomistic motions of constituent phosphorylation sites were restricted, indicative of structural occurrences that could distort the accessibility of interactive kinase proteins. A similar pattern was observed at a target phosphorylation site located in the globular domain of CRMP-2. MM/PBSA analyses revealed that both compounds interacted favorably with CRMP-2 while crucial residues that enhanced their selective binding include Glu353, Thr349, Lys254, Asp140 and Arg75. These structural insights provide mechanistic events that could contribute towards the structure-based design of anti-AD molecules which can bind CRMP2 selectively and alter its phosphorylation process.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Flavanonas/uso terapéutico , Glucósidos/uso terapéutico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Flavanonas/farmacología , Glucósidos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Simulación de Dinámica Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Termodinámica
16.
Chem Biodivers ; 15(3): e1700533, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29325229

RESUMEN

Bcr-Abl is an oncogenic fusion protein which expression enhances tumorigenesis, and has been highly associated with chronic myeloid leukemia (CML). Acquired drug resistance in mutant Bcr-Abl has enhanced pathogenesis with the use of single therapy agents such as nilotinib. Moreover, allosteric targeting has been identified to consequentially inhibit Bcr-Abl activity, which led to the recent development of ABL-001 (asciminib) that selectively binds the myristoyl pocket. Experimental studies have revealed that the combination of nilotinib and ABL-001 induced a 'bent' conformation in the C-terminal helix of Bcr-Abl; a benchmark of inhibition, thereby exhibiting a greater potency in the treatment of CML, surmounting the setbacks of drug resistance, disease regression and relapse. Therefore, we report the first account of the dynamics and conformational analysis of oncogenic T334I Bcr-Abl by dual targeting. Our findings revealed that unlike in the Bcr-Abl-Nilotinib complex, dual targeting by both inhibitors induced the bent conformation in the C-terminal helix that varied with time. This was coupled with significant alteration in Bcr-Abl stability, flexibility, and compactness and an overall structural re-orientation inwards towards the hydrophobic core, which reduced the solvent-exposed residues indicative of protein folding. This study will facilitate allosteric targeting and the design of more potent allosteric inhibitors for resistive target proteins in cancer.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas/efectos de los fármacos , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Simulación de Dinámica Molecular , Estructura Molecular , Mutación , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
17.
Biotechnol Lett ; 39(12): 1843-1851, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28918491

RESUMEN

OBJECTIVES: To provide insight into the dynamics of the shape-shifting mechanistic events associated with the opening (activation) of Lymphocyte Function Associated Antigen-1 upon allosteric modulation by an activator, ICAM Binding Enhancer-667 (IBE-667), using molecular dynamics simulation. RESULTS: Various parameters were used to appropriately describe and understand the sequence of events that characterized its activation across the simulation period such as residual distances, TriCα angles; as well as the dihedral angle. Our findings revealed a significant residual fluctuation and stability difference between both systems. Also, there was a synergistic coordination of the active MIDAS site by the downward pull of the α7 helix upon ligand binding, which appeared to be directly proportional to each other. CONCLUSION: Allosteric binding of IBE-667, activated LFA-1 integrin as evidenced by residual motion at the MIDAS region which appears to be synergistically coordinated by the downward pull of the α7 helix.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito/química , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Transducción de Señal/fisiología , Azepinas/química , Azepinas/metabolismo , Biología Computacional , Humanos , Indazoles/química , Indazoles/metabolismo , Simulación de Dinámica Molecular , Unión Proteica
18.
J Biomol Struct Dyn ; 41(6): 2419-2430, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35105282

RESUMEN

The specific inhibition of aberrant Fibroblast Growth Factor Receptors (FGFRs) has been identified as a feasible strategy to therapeutically ameliorate their respective carcinogenic involvements. High homology among these proteins has however limited efforts towards the discovery of selective small-molecule compounds due to undesirable effects elicited by pan-FGFR inhibitors. A recent study showed the selective activity of a new compound C11 which was >52 times more potent against FGFR1 than FGFR2 and FGFR3, and 4 times than FGFR4. This C11 selective non-covalency was investigated in this study using computational methods since it has remained unresolved. Structural findings revealed that C11 enhanced structural perturbations in FGFR1 with less prominent effects in other FGFRs. High deviations also characterized the C11-bound active pocket of FGFR1 with notable fluctuations across the constituent P-loop, αC helix, hinge region, catalytic, and activation loops. These induced motions were essential for optimal C11 motion an d positioning of its phenalenone ring and prop-2-en-l-yl moiety at the FGFR1 active pocket to interact stably and strongly with A564FGFR1, L484FGFR1, Y563FGFR1, and E562FGFR1 which as well had high energy contributions. C11 exhibited highly unstable binding in F GFRs2-3 with a more steady interaction with FGFR4. Free binding energy (ΔGbind) analyses further estimated the highest interaction energy for C11-FGFR1 with favorable desolvation energy that indicated a deep hydrophobic pocket binding for C11 in FGFR1 compared to other FGFRs. We believe rational insights from this study will contribute to the structure-based design of highly specific FGFR1 inhibitors.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Transducción de Señal , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores
19.
J Biomol Struct Dyn ; 40(21): 10878-10886, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34463214

RESUMEN

PARP-1 has become an attractive target in cancer treatment owing to its significant role in breast and ovarian cancers. The design of highly selective and effective poly (ADP ribose) polymerase-1 inhibitors has significant therapeutic advantages and has remained the core of several PARP-1-based drug discovery research. The pharmacophoric relevance of a chlorine substituent in a recent study led to the design of compounds 11c (meta-chlorophenyl) and 11d (para-chlorophenyl). In this study, we resolved the mechanistic effects of the changes in chlorine positional orientation, which underlie the inhibitory potencies and selectivity exhibited disparately by 11c and 11d. Compared to 11d, among other multiple higher-affinity complementary interactions with key site residues, the meta-Cl positioning in 11c facilitated its optimal motion and orientation towards conserved residues Arg878 and Asp766 with consistent pi-cation and pi-anion interactions, respectively, thereby favoring the stability of the ligand towards PARP-1. These could account for the higher inhibitory potency exhibited by 11c relative to 11d against PARP-1. The thermodynamics calculation revealed that 11c had a relatively higher total binding energy (ΔGbind) than 11d. We also observed that 11d displayed high deviations, compared to 11c, indicative of its unstable binding orientation. Furthermore, we reported in this study that the high involvement of electrostatic and van der Waal effects potentiated the binding affinity and strength of 11c (ΔEvdW = -50.58 and ΔEele = -27.20) relative to 11d (ΔEvdW = -49.46 and ΔEele = -19.96) at PARP-1 binding pocket. We believe the findings in this current study would provide valuable insights into the design of selective PARP-1 inhibitors containing chlorine substituent for cancer treatment, including lung cancer.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Cloro , Neoplasias Pulmonares/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasa-1/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
20.
Artículo en Inglés | MEDLINE | ID: mdl-35616668

RESUMEN

BACKGROUND: Parkinson's disease (PD) is one of the most prominent neurodegenerative diseases hence the continual search for viable and effective treatment options. The pathogeny of PD is driven by many key proteins among which is the recently identified Leucine-rich repeated kinase 2 (LRRK2). Going forward, the onus lies on identifying small-molecule inhibitors that can halt its pathogenic involvement, and, importantly, possess the capacity to cross the blood-brain barrier (BBB). Although several compounds have been identified over the past decade for their potencies, a major limitation remains the inability of the majority to cross the blood-brain barrier (BBB). A novel series of benzothiazole-based compounds with varying LRRK2 inhibitory activities were recently synthesized, with one compound 14 (CPD14) that notably inhibited LRRK2 and promoted neuronal progenitor proliferation. METHODS: Here, we implemented molecular modelling and computational simulation methods to characterize CPD14 inhibitory mechanisms and dynamics against LRRK2. More so, we employed pharmacokinetic parameters to evaluate the biological activity and CNS-suitability of CPD14. RESULTS: Molecular dynamics evaluation revealed that CPD14 elicited disruptive effects on the secondary structure of LRRK2, including its catalytic kinase domain. Interaction analyses at the binding site further revealed crucial residues for the affinity binding and stability of CPD14, further supported by a highly favorable binding energy (ΔG). Pharmacokinetic predictions revealed the CNS-suitability of CPD14 based on its adherence to Lipinski's rule of 5 for neurogenic compounds. Also, CPD14 exhibited inhibitory tendencies against transcription proteins such as signal transducer and activation transcription (STAT) protein and STAT3; complementary mechanisms that could account for its in vitro potency. CONCLUSION: These findings, taken together, will aid the pharmacological and pharmacokinetic optimization of novel LRRK2 inhibitors for the treatment of PD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA