Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Biochem ; 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37796176

RESUMEN

In recent years, it has been shown that the liquid-liquid phase separation (LLPS) of virus proteins plays a crucial role in their life cycle. It promotes the formation of viral replication organelles, concentrating viral components for efficient replication and facilitates the assembly of viral particles. LLPS has emerged as a crucial process in the replication and assembly of herpes simplex virus-1 (HSV-1). Recent studies have identified several HSV-1 proteins involved in LLPS, including the myristylated tegument protein UL11 and infected cell protein 4; however, a complete proteome-level understanding of the LLPS-prone HSV-1 proteins is not available. We provide a comprehensive analysis of the HSV-1 proteome and explore the potential of its proteins to undergo LLPS. By integrating sequence analysis, prediction algorithms and an array of tools and servers, we identified 10 HSV-1 proteins that exhibit high LLPS potential. By analysing the amino acid sequences of the LLPS-prone proteins, we identified specific sequence motifs and enriched amino acid residues commonly found in LLPS-prone regions. Our findings reveal a diverse range of LLPS-prone proteins within the HSV-1, which are involved in critical viral processes such as replication, transcriptional regulation and assembly of viral particles. This suggests that LLPS might play a crucial role in facilitating the formation of specialized viral replication compartments and the assembly of HSV-1 virion. The identification of LLPS-prone proteins in HSV-1 opens up new avenues for understanding the molecular mechanisms underlying viral pathogenesis. Our work provides valuable insights into the LLPS landscape of HSV-1, highlighting potential targets for further experimental validation and enhancing our understanding of viral replication and pathogenesis.

2.
Biochem Biophys Res Commun ; 629: 54-60, 2022 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-36113178

RESUMEN

Shortly after the onset of the COVID-19 pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has acquired numerous variations in its intracellular proteins to adapt quickly, become more infectious, and ultimately develop drug resistance by mutating certain hotspot residues. To keep the emerging variants at bay, including Omicron and subvariants, FDA has approved the antiviral nirmatrelvir for mild-to-moderate and high-risk COVID-19 cases. Like other viruses, SARS-CoV-2 could acquire mutations in its main protease (Mpro) to adapt and develop resistance against nirmatrelvir. Employing a unique high-throughput protein design technique, the hotspot residues, and signatures of adaptation of Mpro having the highest probability of mutating and rendering nirmatrelvir ineffective were identified. Our results show that ∼40% of the designed mutations in Mpro already exist in the globally circulating SARS-CoV-2 lineages and several predicted mutations. Moreover, several high-frequency, designed mutations were found to be in corroboration with the experimentally reported nirmatrelvir-resistant mutants and are naturally occurring. Our work on the targeted design of the nirmatrelvir-binding site offers a comprehensive picture of potential hotspot sites and resistance mutations in Mpro and is thus crucial in comprehending viral adaptation, robust antiviral design, and surveillance of evolving Mpro variations.


Asunto(s)
COVID-19 , SARS-CoV-2 , Antivirales/química , Sitios de Unión , COVID-19/genética , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/metabolismo , Genoma Viral , Humanos , Mutación , Pandemias , Inhibidores de Proteasas/química , SARS-CoV-2/genética , Proteínas no Estructurales Virales/química
3.
Phys Chem Chem Phys ; 24(16): 9141-9145, 2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35411366

RESUMEN

Dimerization of SARS-CoV-2 main protease (Mpro) is a prerequisite for its processing activity. With >2000 mutations already reported in Mpro, SARS-CoV-2 may accumulate mutations in the Mpro dimeric interface to stabilize it further. We employed high-throughput protein design strategies to design the symmetrical dimeric interface of Mpro (300 000 designs) to identify mutational hotspots that render the Mpro more stable. We found that ∼22% of designed mutations that yield stable Mpro dimers already exist in SARS-CoV-2 genomes and are currently circulating. Our multi-parametric analyses highlight potential Mpro mutations that SARS-CoV-2 may develop, providing a foundation for assessing viral adaptation and mutational surveillance.


Asunto(s)
Proteasas 3C de Coronavirus , Ingeniería de Proteínas , SARS-CoV-2 , COVID-19 , Proteasas 3C de Coronavirus/genética , Dimerización , Humanos , SARS-CoV-2/enzimología , SARS-CoV-2/genética
4.
Molecules ; 27(6)2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35335141

RESUMEN

Most neurodegenerative diseases such as Alzheimer's disease, type 2 diabetes, Parkinson's disease, etc. are caused by inclusions and plaques containing misfolded protein aggregates. These protein aggregates are essentially formed by the interactions of either the same (homologous) or different (heterologous) sequences. Several experimental pieces of evidence have revealed the presence of cross-seeding in amyloid proteins, which results in a multicomponent assembly; however, the molecular and structural details remain less explored. Here, we discuss the amyloid proteins and the cross-seeding phenomena in detail. Data suggest that targeting the common epitope of the interacting amyloid proteins may be a better therapeutic option than targeting only one species. We also examine the dual inhibitors that target the amyloid proteins participating in the cross-seeding events. The future scopes and major challenges in understanding the mechanism and developing therapeutics are also considered. Detailed knowledge of the amyloid cross-seeding will stimulate further research in the practical aspects and better designing anti-amyloid therapeutics.


Asunto(s)
Amiloidosis , Diabetes Mellitus Tipo 2 , Amiloide/química , Péptidos beta-Amiloides/metabolismo , Proteínas Amiloidogénicas , Amiloidosis/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos
5.
Biochem Biophys Res Commun ; 555: 147-153, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813274

RESUMEN

Several existing drugs are currently being tested worldwide to treat COVID-19 patients. Recent data indicate that SARS-CoV-2 is rapidly evolving into more transmissible variants. It is therefore highly possible that SARS-CoV-2 can accumulate adaptive mutations modulating drug susceptibility and hampering viral antigenicity. Thus, it is vital to predict potential non-synonymous mutation sites and predict the evolution of protein structural modifications leading to drug tolerance. As two FDA-approved anti-hepatitis C virus (HCV) drugs, boceprevir, and telaprevir, have been shown to effectively inhibit SARS-CoV-2 by targeting the main protease (Mpro), here we used a high-throughput interface-based protein design strategy to identify mutational hotspots and potential signatures of adaptation in these drug binding sites of Mpro. Several mutants exhibited reduced binding affinity to these drugs, out of which hotspot residues having a strong tendency to undergo positive selection were identified. The data further indicated that these anti-HCV drugs have larger footprints in the mutational landscape of Mpro and hence encompass the highest potential for positive selection and adaptation. These findings are crucial in understanding the potential structural modifications in the drug binding sites of Mpro and thus its signatures of adaptation. Furthermore, the data could provide systemic strategies for robust antiviral design and discovery against COVID-19 in the future.


Asunto(s)
Adaptación Fisiológica/genética , Antivirales/química , Proteasas 3C de Coronavirus/química , Diseño de Fármacos , Farmacorresistencia Viral/genética , Mutación , SARS-CoV-2/enzimología , SARS-CoV-2/genética , Secuencia de Aminoácidos , Antivirales/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/genética , Proteasas 3C de Coronavirus/metabolismo , Aptitud Genética/genética , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Ligandos , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/farmacología , Prolina/análogos & derivados , Prolina/química , Prolina/farmacología , Reproducibilidad de los Resultados , SARS-CoV-2/efectos de los fármacos , Selección Genética/genética , Relación Estructura-Actividad , Tratamiento Farmacológico de COVID-19
6.
Mol Divers ; 25(3): 1929-1943, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33575983

RESUMEN

The H1N1 influenza virus causes a severe disease that affects the human respiratory tract leading to millions of deaths every year. At present, certain vaccines and few drugs are used to control the virus during seasonal outbreaks. However, high mutation rates and genetic reassortment make it challenging to prevent and mitigate outbreaks, leading to pandemics. Thus, alternate therapies are required for its management and control. Here, we report that a bacterial protein, azurin, and its peptide derivatives p18 and p28 target critical proteins of the influenza virus in an effective manner. The molecular docking studies show that the p28 peptide could target C-PB1, NS1-ED, PB2-CBD, PB2-RBD, NP, and PA proteins. These complexes were further subjected to the simulation of molecular dynamics and binding free energy calculations. The data indicate that p28 has an unusually high affinity and forms stable complexes with the viral proteins C-PB1, PB2-CBD, PB2-RBD, and NP. We suggest that the azurin derivative p28 peptide can act as an anti-influenza agent as it can bind to multiple targets and neutralize the virus. Additional experimental studies need to be conducted to evaluate its safety and efficacy as an anti-H1N1 molecule.


Asunto(s)
Antivirales/química , Azurina/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Fragmentos de Péptidos/química , Proteínas Virales/química , Antivirales/farmacología , Azurina/farmacología , Sitios de Unión , Dominio Catalítico , Descubrimiento de Drogas , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Conformación Molecular , Fragmentos de Péptidos/farmacología , Unión Proteica , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores
7.
Biochemistry ; 59(4): 341-342, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31944100

RESUMEN

The deposition of amyloid-ß (Aß) plaques and tau-based neurofibrillary tangles is a neuropathological feature of Alzheimer's disease (AD). While studies have shown that the Aß and tau interaction results in elevated AD pathology, the molecular linkage and mechanism of interaction of Aß and tau are unclear. A recent study demonstrated the direct interaction between the Aß core and specific regions of tau that facilitates pathological cross-seeding via a shared epitope. The data suggest that targeting the common epitope could be a more effective treatment strategy rather than targeting only Aß or only tau. The findings have an important clinical significance for AD and related tauopathies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Animales , Epítopos , Humanos , Ovillos Neurofibrilares/inmunología , Tauopatías/patología , Proteínas tau/inmunología
8.
Microb Pathog ; 145: 104236, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32376359

RESUMEN

Coronavirus disease 2019 (COVID-19) is an emerging infectious disease that was first reported in Wuhan, China, and has subsequently spread worldwide. In the absence of any antiviral or immunomodulatory therapies, the disease is spreading at an alarming rate. A possibility of a resurgence of COVID-19 in places where lockdowns have already worked is also developing. Thus, for controlling COVID-19, vaccines may be a better option than drugs. An mRNA-based anti-COVID-19 candidate vaccine has entered a phase 1 clinical trial. However, its efficacy and potency have to be evaluated and validated. Since vaccines have high failure rates, as an alternative, we are presenting a new, designed multi-peptide subunit-based epitope vaccine against COVID-19. The recombinant vaccine construct comprises an adjuvant, cytotoxic T-lymphocyte (CTL), helper T-lymphocyte (HTL), and B-cell epitopes joined by linkers. The computational data suggest that the vaccine is non-toxic, non-allergenic, thermostable, with the capability to elicit a humoral and cell-mediated immune response. The stabilization of the vaccine construct is validated with molecular dynamics simulation studies. This unique vaccine is made up of 33 highly antigenic epitopes from three proteins that have a prominent role in host-receptor recognition, viral entry, and pathogenicity. We advocate this vaccine must be synthesized and tested urgently as a public health priority.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Proteínas de la Nucleocápside/inmunología , Pandemias/prevención & control , Neumonía Viral/prevención & control , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas de Subunidad/inmunología , Vacunas Virales/inmunología , Antígenos Virales/inmunología , COVID-19 , Infecciones por Coronavirus/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Humanos , Simulación de Dinámica Molecular , Neumonía Viral/inmunología , SARS-CoV-2
9.
J Cell Physiol ; 234(4): 4491-4503, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30192010

RESUMEN

Glutathione S-transferases (GSTs) are enzymes that are involved in the detoxification of harmful electrophilic endogenous and exogenous compounds by conjugating with glutathione (GSH). The liver fluke GSTs have multifunctional roles in the host-parasite interaction, such as general detoxification and bile acid sequestration to synthase activity. The GSTs have been highlighted as vaccine candidates towards parasitic flukes. In this study, we have thoroughly examined the urea-induced unfolding of a mu-class Fasciola gigantica GST1 (FgGST1) using spectroscopic techniques and molecular dynamic simulations. FgGST1 is a highly cooperative molecule, because during urea-induced equilibrium unfolding, a concurrent unfolding of the protein without stabilization of any folded intermediate was observed. The protein was stabilized with conformational free energy of about ~12.36 kcal/mol. The protein loses its activity with increasing urea concentration, as the GSH molecule is not able to bind to the protein. We also studied the fluorescence quenching of Trp residues and the obtained K SV data that provided additional information on the unfolding of FgGST1. Molecular dynamic trajectories simulated in different urea concentrations and temperatures indicated that urea destabilizes FgGST1 structure by weakening hydrophobic interactions and the hydrogen bond network. We observed a precise correlation between the in vitro and in silico studies.


Asunto(s)
Fasciola/enzimología , Glutatión Transferasa/química , Proteínas del Helminto/química , Desplegamiento Proteico , Urea/química , Animales , Dicroismo Circular , Fasciola/genética , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Desnaturalización Proteica , Estructura Secundaria de Proteína , Espectrometría de Fluorescencia , Relación Estructura-Actividad
10.
J Cell Biochem ; 120(2): 2469-2482, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30206985

RESUMEN

Persistent infection by Mycobacterium tuberculosis requires the glyoxylate shunt. This is a bypass to the tricarboxylic acid cycle in which isocitrate lyase (ICL) and malate synthase (MS) catalyze the net incorporation of carbon during mycobacterial growth on acetate or fatty acids as the primary carbon source. To identify a potential antitubercular compound, we performed a structure-based screening of natural compounds from the ZINC database (n = 1 67 740) against the M tuberculosis MS (MtbMS) structure. The ligands were screened against MtbMS, and 354 ligands were found to have better docking score. These compounds were assessed for Lipinski and absorption, distribution, metabolism, excretion, and toxicity prediction where 15 compounds were found to fit well for redocking studies. After refinement by molecular docking and drug-likeness analysis, four potential inhibitors (ZINC1483899, ZINC1754310, ZINC2269664, and ZINC15729522) were identified. These four ligands with phenyl-diketo acid were further subjected to molecular dynamics simulation to compare the dynamics and stability of the protein structure after ligand binding. The binding energy analysis was calculated to determine the intermolecular interactions. Our results suggested that the four compounds had a binding free energy of -201.96, -242.02, -187.03, and -169.02 kJ·mol-1 , for compounds with IDs ZINC1483899, ZINC1754310, ZINC2269664, and ZINC15729522, respectively. We concluded that two compounds (ZINC1483899 and ZINC1754310) displayed considerable structural and pharmacological properties and could be probable drug candidates to fight against M tuberculosis parasites.

11.
Arch Biochem Biophys ; 674: 108098, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31494118

RESUMEN

Thioredoxin glutathione reductase (TGR), a potential anthelminthic drug target causes NADPH-dependent transfer of electrons to both thioredoxins and glutathione systems. In the present study, we showed that a single point mutation conserved at Arg451 position is critical for maintaining the structure-function of FgTGR. The current biochemical results showed that R451A mutation significantly decreases both oxidoreductase activities (glutathione reductase and thioredoxin reductase) of the enzyme. Computational analyses using molecular dynamics simulation provided an in-depth insight into the structural alterations caused as a result of the mutation. Furthermore, the different regions of the mutant FgTGR structure were found to be altered in flexibility/rigidity as a result of the mutation. This led to mutant-specific conformational alterations and dominant differential motions that contributed to the abrogated function of mutant FgTGR. These results were confirmed using GdnHCl-induced denaturation-based stability studies. Moreover, mutation reduced the free energy of stabilization of the protein, thereby destabilizing the mutant protein structure. Therefore, these findings displayed differential dynamics in the FgTGR structure and highlighted the relevance of residue-level interactions in the protein. Thus, the current study provided a basis for exploiting regions other than the active site of TGR for inhibitory effect and development of novel antihelminthics.


Asunto(s)
Arginina/química , Proteínas del Helminto/química , Complejos Multienzimáticos/química , NADH NADPH Oxidorreductasas/química , Animales , Dominio Catalítico , Fasciola/enzimología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Simulación de Dinámica Molecular , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , Mutación Puntual , Análisis de Componente Principal , Estabilidad Proteica , Estructura Secundaria de Proteína/genética , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinámica
12.
Biochem J ; 475(21): 3377-3391, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30287492

RESUMEN

The asparaginyl-tRNA synthetase (NRS) catalyzes the attachment of asparagine to its cognate tRNA during translation. NRS first catalyzes the binding of Asn and ATP to form the NRS-asparaginyl adenylate complex, followed by the esterification of Asn to its tRNA. We investigated the role of constituent domains in regulating the structure and activity of Fasciola gigantica NRS (FgNRS). We cloned the full-length FgNRS, along with its various truncated forms, expressed, and purified the corresponding proteins. Size exclusion chromatography indicated a role of the anticodon-binding domain (ABD) of FgNRS in protein dimerization. The N-terminal domain (NTD) was not essential for cognate tRNA binding, and the hinge region between the ABD and the C-terminal domain (CTD) was crucial for regulating the enzymatic activity. Molecular docking and fluorescence quenching experiments elucidated the binding affinities of the substrates to various domains. The molecular dynamics simulation of the modeled protein showed the presence of an unstructured region between the NTD and ABD that exhibited a large number of conformations over time, and further analysis indicated this region to be intrinsically disordered. The present study provides information on the structural and functional regulation, protein-substrate(s) interactions and dynamics, and the role of non-catalytic domains in regulating the activity of FgNRS.


Asunto(s)
Aspartato-ARNt Ligasa/metabolismo , Fasciola/metabolismo , Proteínas del Helminto/metabolismo , Aminoacil-ARN de Transferencia/metabolismo , ARN de Transferencia/metabolismo , Animales , Aspartato-ARNt Ligasa/química , Aspartato-ARNt Ligasa/genética , Sitios de Unión/genética , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Fasciola/enzimología , Fasciola/genética , Proteínas del Helminto/química , Proteínas del Helminto/genética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , ARN de Transferencia/genética , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/genética , Especificidad por Sustrato
13.
Parasitol Res ; 118(3): 861-872, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30706165

RESUMEN

Fasciola gigantica is an important food-borne trematode responsible for the hepatobiliary disease, commonly known as fascioliasis. In F. gigantica, the glyceraldehyde 3-phosphate dehydrogenase (FgGAPDH) is a key enzyme of the glycolytic pathway and catalyzes the reversible oxidative phosphorylation of D-glyceraldehyde-3-phosphate (G-3-P) to 1,3-bisphosphoglycerate (1,3-BPG), with the simultaneous reduction of NAD+ to NADH. In the present study, we analyzed the sequence of FgGAPDH and investigated its structural, binding, and catalytic properties. Sequence alignment of FgGAPDH showed 100% identity with the sister fluke Fasciola hepatica GAPDH. The gapdh gene was cloned and expressed in Escherichia coli, and the recombinant protein was purified. The purified FgGAPDH exists as a homo-tetramer, composed of a ~ 37-kDa subunit under non-dissociating conditions at 300 mM salt concentration indicating that higher salt stabilizes the tetrameric state. The binding of the cofactor NAD+ caused a conformational rearrangement in the enzyme structure, leading to the stabilization of the enzyme. A homology model of FgGAPDH was constructed, the cofactor (NAD+) and substrate (G-3-P) were docked, and the binding sites were identified in a single chain. The inter-subunit cleft of GAPDH that has been exploited for structure-based drug design in certain protozoan parasites is closed in the case of FgGAPDH, similar to the human GAPDH. Thus, the conformation of FgGAPDH in this region is similar to the human enzyme. Therefore, GAPDH may not be a suitable target for drug discovery against fascioliasis. Still, the analysis of the structural and functional attributes of GAPDH will be significant in understanding the various roles of this enzyme in the parasite as well as provide new insights into the biochemistry of flukes.


Asunto(s)
Fasciola/enzimología , Fasciola/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Fascioliasis/parasitología , Humanos , Proteínas Recombinantes/genética , Alineación de Secuencia
14.
Molecules ; 24(18)2019 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-31527516

RESUMEN

Amyloids are fibrous proteins aggregated into toxic forms that are implicated in several chronic disorders. More than 30 diseases show deposition of fibrous amyloid proteins associated with cell loss and degeneration in the affected tissues. Evidence demonstrates that amyloid diseases result from protein aggregation or impaired amyloid clearance, but the connection between amyloid accumulation and tissue degeneration is not clear. Common examples of amyloid diseases are Alzheimer's disease (AD), Parkinson's disease (PD) and tauopathies, which are the most common forms of neurodegenerative diseases, as well as polyglutamine disorders and certain peripheral metabolic diseases. In these diseases, increased accumulation of toxic amyloid proteins is suspected to be one of the main causative factors in the disease pathogenesis. It is therefore important to more clearly understand how these toxic amyloid proteins accumulate as this will aide in the development of more effective preventive and therapeutic strategies. Protein homeostasis, or proteostasis, is maintained by multiple cellular pathways-including protein synthesis, quality control, and clearance-which are collectively responsible for preventing protein misfolding or aggregation. Modulating protein degradation is a very complex but attractive treatment strategy used to remove amyloid and improve cell survival. This review will focus on autophagy, an important clearance pathway of amyloid proteins, and strategies for using it as a potential therapeutic target for amyloid diseases. The physiological role of autophagy in cells, pathways for its modulation, its connection with apoptosis, cell models and caveats in developing autophagy as a treatment and as a biomarker is discussed.


Asunto(s)
Proteínas Amiloidogénicas/química , Autofagia/efectos de los fármacos , Amiloide/química , Amiloide/metabolismo , Proteínas Amiloidogénicas/antagonistas & inhibidores , Proteínas Amiloidogénicas/metabolismo , Animales , Apoptosis , Biomarcadores , Susceptibilidad a Enfermedades , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/tratamiento farmacológico , Transducción de Señal
16.
J Cell Biochem ; 119(4): 3067-3080, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29052925

RESUMEN

Fascioliasis is caused by the helminth parasites of genus Fasciola. Thioredoxin glutathione reductase (TGR) is an important enzyme in parasitic helminths and plays an indispensable role in its redox biology. In the present study, we conducted a structure-based virtual screening of natural compounds against the Fasciola gigantica TGR (FgTGR). The compounds were docked against FgTGR in four sequential docking modes. The screened ligands were further assessed for Lipinski and ADMET prediction so as to evaluate drug proficiency and likeness property. After refinement, three potential inhibitors were identified that were subjected to 50 ns molecular dynamics simulation and free energy binding analyses to evaluate the dynamics of protein-ligand interaction and the stability of the complexes. Key residues involved in the interaction of the selected ligands were also determined. The results suggested that three top hits had a negative binding energy greater than GSSG (-91.479 KJ · mol-1 ), having -152.657, -141.219, and -92.931 kJ · mol-1 for compounds with IDs ZINC85878789, ZINC85879991, and ZINC36369921, respectively. Further analysis showed that the compound ZINC85878789 and ZINC85879991 displayed substantial pharmacological and structural properties to be a drug candidate. Thus, the present study might prove useful for the future design of new derivatives with higher potency and specificity.


Asunto(s)
Antiplatelmínticos/química , Inhibidores Enzimáticos/química , Fasciola/enzimología , Complejos Multienzimáticos/química , NADH NADPH Oxidorreductasas/química , Animales , Antiplatelmínticos/farmacología , Sitios de Unión , Simulación por Computador , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Fasciola/efectos de los fármacos , Proteínas del Helminto/química , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Complejos Multienzimáticos/antagonistas & inhibidores , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , Análisis de Componente Principal , Multimerización de Proteína , Homología Estructural de Proteína
17.
Arch Biochem Biophys ; 656: 38-45, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30205085

RESUMEN

Thioredoxin glutathione reductase (TGRsec) is a multi-domain flavoprotein that plays a principal role in redox homeostasis maintenance. We have previously demonstrated the role of selenocysteine in maintaining TGRsec structure-function, but the role of the glutaredoxin (Grx) domain and FAD is still unclear. In the present study, the urea-induced unfolding of recombinant Fasciola gigantica TGRsec (FgTGRsec) and its N-terminal truncated variant (ΔNTD-FgTGRsec) were examined to understand the role of the Grx domain and FAD in the stabilization of FgTGRsec and ΔNTD-FgTGRsec. Our results showed that both proteins underwent unfolding in a three state manner. First, the protein undergoes a conformational transition rendering a near-native state with no FAD bound, and then full unfolding of the apo-dimer occurs without dissociation. The Grx domain stabilized the global FgTGRsec structure and positively regulated FgTGRsec activity, and alteration in the FAD microenvironment was directly proportional to the loss of thioredoxin reductase (TrxR) and glutathione reductase activities. Based on these results, we concluded that the Grx domain stabilizes the full-length FgTGRsec protein for efficient catalysis. Thus, we suggest that in platyhelminth parasites, during evolution, the Grx domain merged with the TrxR domain to confer higher catalytic activity and provide additional structural stability to the full-length TGR.


Asunto(s)
Flavina-Adenina Dinucleótido/química , Glutarredoxinas/química , Proteínas del Helminto/química , Complejos Multienzimáticos/química , NADH NADPH Oxidorreductasas/química , Dominios Proteicos , Animales , Catálisis , Ácido Ditionitrobenzoico/metabolismo , Fasciola/enzimología , Flavina-Adenina Dinucleótido/metabolismo , Glutarredoxinas/genética , Glutarredoxinas/aislamiento & purificación , Glutarredoxinas/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/aislamiento & purificación , Proteínas del Helminto/metabolismo , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/aislamiento & purificación , Complejos Multienzimáticos/metabolismo , Mutación , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/aislamiento & purificación , NADH NADPH Oxidorreductasas/metabolismo , NADP/metabolismo , Unión Proteica , Conformación Proteica/efectos de los fármacos , Estabilidad Proteica , Desplegamiento Proteico/efectos de los fármacos , Tiorredoxinas/química , Tiorredoxinas/genética , Tiorredoxinas/aislamiento & purificación , Tiorredoxinas/metabolismo , Triptófano/química , Urea/química
18.
Protein Expr Purif ; 143: 9-13, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29031680

RESUMEN

We report the molecular cloning, expression, and single-step homogeneous purification of a full-length asparaginyl tRNA synthetase (NRS) from Fasciola gigantica (FgNRS). Fasciola gigantica is a parasitic liver fluke of the class Trematoda. It causes fascioliasis that infects the liver of various mammals, including humans. Aminoacyl tRNA synthetases (AARS) catalyze the first step of protein synthesis. They attach an amino acid to its cognate tRNA, forming an amino acid-tRNA complex. The gene that codes for FgNRS was generated by amplification by polymerase chain reaction. It was then inserted in the expression vector pQE30 under the transcriptional control of the bacteriophage T5 promoter and lac operator. M15 Escherichia coli strain transformed with the FgNRS expression vector pQE30-NRS accumulates large amounts of a soluble protein of about 61 kDa. The protein was purified to homogeneity using immobilized metal affinity chromatography. The recombinant protein was further confirmed by immunoblotting with anti-His antibody. Following size exclusion chromatography, the FgNRS was stable and observed to be a dimeric protein. In this study, the expression and purification procedures have provided a simple and efficient method to obtain full-length FgNRS in large quantities. This will provide an opportunity to study the structure, dynamics and function of NRS.


Asunto(s)
Aspartato-ARNt Ligasa/metabolismo , Fasciola/enzimología , Fasciola/genética , Aminoacil-ARN de Transferencia/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Aspartato-ARNt Ligasa/química , Aspartato-ARNt Ligasa/genética , Aspartato-ARNt Ligasa/aislamiento & purificación , Western Blotting , Cromatografía en Gel , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/genética , Aminoacil-ARN de Transferencia/aislamiento & purificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
19.
Biochim Biophys Acta Gen Subj ; 1862(6): 1306-1316, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29526505

RESUMEN

The thiol-disulfide redox metabolism in platyhelminth parasites depends entirely on a single selenocysteine (Sec) containing flavoenzyme, thioredoxin glutathione reductase (TGR) that links the classical thioredoxin (Trx) and glutathione (GSH) systems. In the present study, we investigated the catalytic and structural properties of different variants of Fasciola gigantica TGR to understand the role of Sec. The recombinant full-length Sec containing TGR (FgTGRsec), TGR without Sec (FgTGR) and TGRsec without the N-terminal glutaredoxin (Grx) domain (∆NTD-FgTGRsec) were purified to homogeneity. Biochemical studies revealed that Sec597 is responsible for higher thioredoxin reductase (TrxR) and glutathione reductase (GR) activity of FgTGRsec. The N-terminal Grx domain was found to positively regulate the DTNB-based TrxR activity of FgTGRsec. The FgTGRsec was highly sensitive to inhibition by auranofin (AF). The structure of FgTGR was modeled, and the inhibitor AF was docked, and binding sites were identified. Unfolding studies suggest that all three proteins are highly cooperative molecules since during GdnHCl-induced denaturation, a monophasic unfolding of the proteins without stabilization of any intermediate is observed. The Cm for GdnHCl induced unfolding of FgTGR was higher than FgTGRsec and ∆NTD-FgTGRsec suggesting that FgTGR without Sec was more stable in solution than the other protein variants. The free energy of stabilization for the proteins was also determined. To our knowledge, this is also the first report on unfolding and stability analysis of any TGR.


Asunto(s)
Fasciola/enzimología , Hígado/parasitología , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/metabolismo , Selenocisteína/química , Selenocisteína/metabolismo , Animales , Sitios de Unión , Bovinos , Glutatión/metabolismo , Hígado/enzimología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Complejos Multienzimáticos/aislamiento & purificación , NADH NADPH Oxidorreductasas/aislamiento & purificación , Oxidación-Reducción , Unión Proteica , Termodinámica
20.
Biochem Biophys Res Commun ; 490(2): 276-282, 2017 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-28610921

RESUMEN

Mycobacterium tuberculosis isocitrate lyase (MtbICL) is a key enzyme of the glyoxylate cycle that catalyzes the cleavage of isocitrate to succinate and glyoxylate and is a potential antituberculosis drug target. The aim of this research was to explore the structural alterations induced by L418A point mutation that caused the loss of enzyme activity. In-depth structural analyses were carried out for understanding the influence of L418A mutation using techniques, viz. molecular dynamics, principal component analysis, time-dependent secondary structure, residue interaction network and molecular docking. Since L418A mutation site is structurally far from the active site, it cannot influence the binding of the substrate directly. Our results showed that collective motions, residual mobility, and flexibility of the enzyme increased upon mutation. The mutated residue changed the global conformational dynamics of the system along with the residue-residue interaction network, leading to a loss of the enzyme activity. The docking results suggest that L418A mutation influenced the binding interactions of the substrate with several residues in the active site of MtbICL. This study provides information on the structural dynamics of MtbICL and highlights the importance of residue level interactions in the protein. Thus, our results may provide significant guidance to the scientific community engaged in designing potent inhibitors targeting MtbICL.


Asunto(s)
Alanina/genética , Isocitratoliasa/metabolismo , Lisina/genética , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/enzimología , Mutación Puntual , Mycobacterium tuberculosis/metabolismo , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA