Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Nucleic Acids Res ; 50(3): 1484-1500, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35037045

RESUMEN

The SARS-CoV-2 coronavirus is the causal agent of the current global pandemic. SARS-CoV-2 belongs to an order, Nidovirales, with very large RNA genomes. It is proposed that the fidelity of coronavirus (CoV) genome replication is aided by an RNA nuclease complex, comprising the non-structural proteins 14 and 10 (nsp14-nsp10), an attractive target for antiviral inhibition. Our results validate reports that the SARS-CoV-2 nsp14-nsp10 complex has RNase activity. Detailed functional characterization reveals nsp14-nsp10 is a versatile nuclease capable of digesting a wide variety of RNA structures, including those with a blocked 3'-terminus. Consistent with a role in maintaining viral genome integrity during replication, we find that nsp14-nsp10 activity is enhanced by the viral RNA-dependent RNA polymerase complex (RdRp) consisting of nsp12-nsp7-nsp8 (nsp12-7-8) and demonstrate that this stimulation is mediated by nsp8. We propose that the role of nsp14-nsp10 in maintaining replication fidelity goes beyond classical proofreading by purging the nascent replicating RNA strand of a range of potentially replication-terminating aberrations. Using our developed assays, we identify drug and drug-like molecules that inhibit nsp14-nsp10, including the known SARS-CoV-2 major protease (Mpro) inhibitor ebselen and the HIV integrase inhibitor raltegravir, revealing the potential for multifunctional inhibitors in COVID-19 treatment.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Exorribonucleasas/metabolismo , Genoma Viral/genética , Inestabilidad Genómica , SARS-CoV-2/enzimología , SARS-CoV-2/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Exorribonucleasas/antagonistas & inhibidores , Genoma Viral/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Inhibidores de Integrasa VIH/farmacología , Isoindoles/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/metabolismo , Compuestos de Organoselenio/farmacología , ARN Viral/biosíntesis , ARN Viral/genética , Raltegravir Potásico/farmacología , SARS-CoV-2/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
2.
PLoS Pathog ; 16(4): e1008487, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32302364

RESUMEN

Most simian immunodeficiency viruses use Nef to counteract the tetherin proteins of their nonhuman primate hosts. Nef also downmodulates cell-surface CD4 and MHC class I (MHC I) molecules and enhances viral infectivity by counteracting SERINC5. We previously demonstrated that tetherin antagonism by SIV Nef is genetically separable from CD4- and MHC I-downmodulation. Here we show that disruption of tetherin antagonism by Nef impairs virus replication during acute SIV infection of rhesus macaques. A combination of mutations was introduced into the SIVmac239 genome resulting in three amino acid substitutions in Nef that impair tetherin antagonism, but not CD3-, CD4- or MHC I-downmodulation. Further characterization of this mutant (SIVmac239AAA) revealed that these changes also result in partial sensitivity to SERINC5. Separate groups of four rhesus macaques were infected with either wild-type SIVmac239 or SIVmac239AAA, and viral RNA loads in plasma and sequence changes in the viral genome were monitored. Viral loads were significantly lower during acute infection in animals infected with SIVmac239AAA than in animals infected with wild-type SIVmac239. Sequence analysis of the virus population in plasma confirmed that the substitutions in Nef were retained during acute infection; however, changes were observed by week 24 post-infection that fully restored anti-tetherin activity and partially restored anti-SERINC5 activity. These observations reveal overlap in the residues of SIV Nef required for counteracting tetherin and SERINC5 and selective pressure to overcome these restriction factors in vivo.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/virología , Productos del Gen nef , Macaca mulatta , Proteínas de la Membrana/metabolismo , ARN Viral/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Carga Viral , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores
3.
Biochem J ; 478(13): 2445-2464, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34198326

RESUMEN

SARS-CoV-2 is a coronavirus that emerged in 2019 and rapidly spread across the world causing a deadly pandemic with tremendous social and economic costs. Healthcare systems worldwide are under great pressure, and there is an urgent need for effective antiviral treatments. The only currently approved antiviral treatment for COVID-19 is remdesivir, an inhibitor of viral genome replication. SARS-CoV-2 proliferation relies on the enzymatic activities of the non-structural proteins (nsp), which makes them interesting targets for the development of new antiviral treatments. With the aim to identify novel SARS-CoV-2 antivirals, we have purified the exoribonuclease/methyltransferase (nsp14) and its cofactor (nsp10) and developed biochemical assays compatible with high-throughput approaches to screen for exoribonuclease inhibitors. We have screened a library of over 5000 commercial compounds and identified patulin and aurintricarboxylic acid (ATA) as inhibitors of nsp14 exoribonuclease in vitro. We found that patulin and ATA inhibit replication of SARS-CoV-2 in a VERO E6 cell-culture model. These two new antiviral compounds will be valuable tools for further coronavirus research as well as potentially contributing to new therapeutic opportunities for COVID-19.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Exorribonucleasas/antagonistas & inhibidores , SARS-CoV-2/enzimología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Animales , Ácido Aurintricarboxílico/farmacología , Chlorocebus aethiops , Pruebas de Enzimas , Exorribonucleasas/metabolismo , Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Patulina/farmacología , Reproducibilidad de los Resultados , SARS-CoV-2/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Células Vero , Proteínas no Estructurales Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo
4.
Molecules ; 27(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35408684

RESUMEN

As a continuation of our earlier work against SARS-CoV-2, seven FDA-approved drugs were designated as the best SARS-CoV-2 nsp16-nsp10 2'-o-methyltransferase (2'OMTase) inhibitors through 3009 compounds. The in silico inhibitory potential of the examined compounds against SARS-CoV-2 nsp16-nsp10 2'-o-methyltransferase (PDB ID: (6W4H) was conducted through a multi-step screening approach. At the beginning, molecular fingerprints experiment with SAM (S-Adenosylmethionine), the co-crystallized ligand of the targeted enzyme, unveiled the resemblance of 147 drugs. Then, a structural similarity experiment recommended 26 compounds. Therefore, the 26 compounds were docked against 2'OMTase to reveal the potential inhibitory effect of seven promising compounds (Protirelin, (1187), Calcium folinate (1913), Raltegravir (1995), Regadenoson (2176), Ertapenem (2396), Methylergometrine (2532), and Thiamine pyrophosphate hydrochloride (2612)). Out of the docked ligands, Ertapenem (2396) showed an ideal binding mode like that of the co-crystallized ligand (SAM). It occupied all sub-pockets of the active site and bound the crucial amino acids. Accordingly, some MD simulation experiments (RMSD, RMSF, Rg, SASA, and H-bonding) have been conducted for the 2'OMTase-Ertapenem complex over 100 ns. The performed MD experiments verified the correct binding mode of Ertapenem against 2'OMTase exhibiting low energy and optimal dynamics. Finally, MM-PBSA studies indicated that Ertapenem bonded advantageously to the targeted protein with a free energy value of -43 KJ/mol. Furthermore, the binding free energy analysis revealed the essential amino acids of 2'OMTase that served positively to the binding. The achieved results bring hope to find a treatment for COVID-19 via in vitro and in vivo studies for the pointed compounds.


Asunto(s)
Metiltransferasas , SARS-CoV-2 , Proteínas no Estructurales Virales , Proteínas Reguladoras y Accesorias Virales , Ertapenem/farmacología , Ligandos , Metiltransferasas/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , S-Adenosilmetionina/química , SARS-CoV-2/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores
5.
J Infect Dis ; 223(11): 1914-1922, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33038249

RESUMEN

BIT225 is a first-in-class inhibitor of human immunodeficiency virus (HIV) type 1 Vpu. A phase II trial enrolled 36 HIV-1-infected, treatment-naive participants in Thailand to receive standard-of-care antiretroviral therapy (ART), tenofovir disoproxil fumarate/emtricitabine/efavirenz (Atripla), with 100 or 200 mg of BIT225 or placebo (daily) for 12 weeks. Combined treatment with BIT225 and ART was found to be generally safe and well tolerated, with antiviral efficacy comparable to that of ART alone. The secondary end point-soluble CD163, a marker of monocyte/macrophage inflammation-was noted to be significantly decreased in the BIT225 arm. Plasma-derived activated CD4+ and CD8+ T cells, natural killer cells, and interleukin 21 were increased in those treated with BIT225. These findings are consistent with inhibition of the known effects of HIV Vpu and may reflect clinically important modulation of inflammatory and immune function. Further clinical study is planned to both confirm and extend these important findings in treatment-naive, and treatment-experienced individuals. Clinical Trials Registration. Australian New Zealand Clinical Trials Registry (Universal Trial Number U1111-1191-2194).


Asunto(s)
Fármacos Anti-VIH , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil , Guanidinas/uso terapéutico , Infecciones por VIH , Pirazoles/uso terapéutico , Fármacos Anti-VIH/uso terapéutico , Australia , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Quimioterapia Combinada , Combinación Efavirenz, Emtricitabina y Fumarato de Tenofovir Disoproxil/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , VIH-1 , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Humanos , Inflamación/tratamiento farmacológico , Tailandia , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Viroporinas/antagonistas & inhibidores
6.
Biotechnol Appl Biochem ; 68(4): 918-926, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32860447

RESUMEN

The importance of new effective treatment methodologies for human immunodeficiency virus (HIV) is undeniable for the medical society. Viral protein U (Vpu), one of the disparaged accessory proteins of HIV, is responsible for the dissemination of viral particles, and HIV mutants lacking Vpu protein have remarkably reduced pathogenicity. Here, we explored the marine natural products to find the leading structures which can potentially inhibit the activity of Vpu in silico. To fulfill this goal, we set up a virtual screening based on molecular docking to evaluate the binding capacity of different marine products to Vpu. For validation, we used molecular dynamics simulation and monitored the root mean square deviation value and binding interactions. The results were intriguing when we realized that the hit compounds (phlorotannins) had previously been identified as reverse transcriptase and HIV protease inhibitors. This research inaugurates a new road to combat HIV by multifaceted mode of action of these marine natural products without putting the normal cells in jeopardy (with their safe toxicological profile).


Asunto(s)
Antirretrovirales/química , Organismos Acuáticos/química , Productos Biológicos/química , VIH-1/química , Proteínas del Virus de la Inmunodeficiencia Humana , Simulación del Acoplamiento Molecular , Proteínas Reguladoras y Accesorias Virales , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Proteínas del Virus de la Inmunodeficiencia Humana/química , Humanos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/química
7.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34502213

RESUMEN

Bone marrow stromal cell antigen 2 (BST-2), also known as CD317 or tetherin, has been identified as a host restriction factor that suppresses the release of enveloped viruses from host cells by physically tethering viral particles to the cell surface; however, this host defense can be subverted by multiple viruses. For example, human immunodeficiency virus (HIV)-1 encodes a specific accessory protein, viral protein U (Vpu), to counteract BST-2 by binding to it and directing its lysosomal degradation. Thus, blocking the interaction between Vpu and BST-2 will provide a promising strategy for anti-HIV therapy. Here, we report a NanoLuc Binary Technology (NanoBiT)-based high-throughput screening assay to detect inhibitors that disrupt the Vpu-BST-2 interaction. Out of more than 1000 compounds screened, four inhibitors were identified with strong activity at nontoxic concentrations. In subsequent cell-based BST-2 degradation assays, inhibitor Y-39983 HCl restored the cell-surface and total cellular level of BST-2 in the presence of Vpu. Furthermore, the Vpu-mediated enhancement of pesudotyped viral particle production was inhibited by Y-39983 HCl. Our findings indicate that our newly developed assay can be used for the discovery of potential antiviral molecules with novel mechanisms of action.


Asunto(s)
Fármacos Anti-VIH/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Antígenos CD/metabolismo , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Nanotecnología/métodos , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral
8.
Molecules ; 26(20)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34684735

RESUMEN

In continuation of our previous effort, different in silico selection methods were applied to 310 naturally isolated metabolites that exhibited antiviral potentialities before. The applied selection methods aimed to pick the most relevant inhibitor of SARS-CoV-2 nsp10. At first, a structural similarity study against the co-crystallized ligand, S-Adenosyl Methionine (SAM), of SARS-CoV-2 nonstructural protein (nsp10) (PDB ID: 6W4H) was carried out. The similarity analysis culled 30 candidates. Secondly, a fingerprint study against SAM preferred compounds 44, 48, 85, 102, 105, 182, 220, 221, 282, 284, 285, 301, and 302. The docking studies picked 48, 182, 220, 221, and 284. While the ADMET analysis expected the likeness of the five candidates to be drugs, the toxicity study preferred compounds 48 and 182. Finally, a density-functional theory (DFT) study suggested vidarabine (182) to be the most relevant SARS-Cov-2 nsp10 inhibitor.


Asunto(s)
Antivirales/química , Productos Biológicos/química , SARS-CoV-2/metabolismo , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Antivirales/metabolismo , Antivirales/uso terapéutico , Sitios de Unión , Productos Biológicos/metabolismo , Productos Biológicos/uso terapéutico , COVID-19/patología , Teoría Funcional de la Densidad , Humanos , Ligandos , Simulación del Acoplamiento Molecular , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , SARS-CoV-2/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Vidarabina/química , Vidarabina/metabolismo , Vidarabina/uso terapéutico , Proteínas Reguladoras y Accesorias Virales/metabolismo , Tratamiento Farmacológico de COVID-19
9.
RNA ; 24(4): 480-485, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29284756

RESUMEN

Inhibitory aptamers against a protein are promising as antagonistic reagents and repressive genetic components. Typically, improvement of such aptamers is achieved by acquiring higher binding affinity. Here, we report an alternative mechanism for the improvement of aptamer activity. Recently, we reported a transcriptional activator based on an inhibitory RNA aptamer against lambda cI repressor. We improved the aptamer through in vitro selection (SELEX) from a randomly mutagenized aptamer pool, followed by in vivo screening and truncation. Biochemical analyses indicated that the activity improvement was achieved by alteration of the complex formation stoichiometry, rather than by higher affinity or expression. Our results suggest an alternative strategy for improving aptamer activity.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Escherichia coli/virología , Proteínas Represoras/antagonistas & inhibidores , Técnica SELEX de Producción de Aptámeros/métodos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Aptámeros de Nucleótidos/genética , Secuencia de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , ARN/genética , Proteínas Represoras/genética , Activación Transcripcional/genética , Proteínas Reguladoras y Accesorias Virales/genética
10.
J Recept Signal Transduct Res ; 40(6): 605-612, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32476594

RESUMEN

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


Asunto(s)
Antivirales/química , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Proteínas no Estructurales Virales/química , Proteínas Reguladoras y Accesorias Virales/química , Aciclovir/análogos & derivados , Aciclovir/química , Aciclovir/uso terapéutico , Ancitabina/química , Ancitabina/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/virología , Evaluación Preclínica de Medicamentos , Guanina , Humanos , Meropenem/química , Meropenem/uso terapéutico , Metiltransferasas , Modelos Moleculares , Simulación del Acoplamiento Molecular , Pandemias , Neumonía Viral/virología , Conformación Proteica/efectos de los fármacos , Ribitol/química , Ribitol/uso terapéutico , SARS-CoV-2 , Trifluridina/química , Trifluridina/uso terapéutico , Interfaz Usuario-Computador , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/ultraestructura , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/ultraestructura
11.
Biochemistry ; 57(44): 6367-6378, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30298725

RESUMEN

Ebola virus (EBOV) is a filovirus that causes a severe and rapidly progressing hemorrhagic syndrome; a recent epidemic illustrated the urgent need for novel therapeutic agents because no drugs have been approved for treatment of Ebola virus. A key contribution to the high lethality observed during EBOV outbreaks comes from viral evasion of the host antiviral innate immune response in which viral protein VP35 plays a crucial role, blocking interferon type I production, first by masking the viral double-stranded RNA (dsRNA) and preventing its detection by the pattern recognition receptor RIG-I. Aiming to identify inhibitors of the interaction of VP35 with the viral dsRNA, counteracting the VP35 viral innate immune evasion, we established a new methodology for high-yield recombinant VP35 (rVP35) expression and purification and a novel and robust fluorescence-based rVP35-RNA interaction assay ( Z' factor of 0.69). Taking advantage of such newly established methods, we screened a small library of Sardinian natural extracts, identifying Limonium morisianum as the most potent inhibitor extract. A bioguided fractionation led to the identification of myricetin as the component that can inhibit rVP35-dsRNA interaction with an IC50 value of 2.7 µM. Molecular docking studies showed that myricetin interacts with the highly conserved region of the VP35 RNA binding domain, laying the basis for further structural optimization of potent inhibitors of VP35-dsRNA interaction.


Asunto(s)
Antivirales/farmacología , Flavonoides/farmacología , Fluorescencia , Extractos Vegetales/farmacología , ARN Bicatenario/antagonistas & inhibidores , ARN Viral/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Humanos , Simulación del Acoplamiento Molecular , Plumbaginaceae/química , Conformación Proteica , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
12.
Int J Mol Sci ; 17(11)2016 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-27792169

RESUMEN

The Ebola virus (EBOV) has been recognised for nearly 40 years, with the most recent EBOV outbreak being in West Africa, where it created a humanitarian crisis. Mortalities reported up to 30 March 2016 totalled 11,307. However, up until now, EBOV drugs have been far from achieving regulatory (FDA) approval. It is therefore essential to identify parent compounds that have the potential to be developed into effective drugs. Studies on Ebola viral proteins have shown that some can elicit an immunological response in mice, and these are now considered essential components of a vaccine designed to protect against Ebola haemorrhagic fever. The current study focuses on chemoinformatic approaches to identify virtual hits against Ebola viral proteins (VP35 and VP40), including protein binding site prediction, drug-likeness, pharmacokinetic and pharmacodynamic properties, metabolic site prediction, and molecular docking. Retrospective validation was performed using a database of non-active compounds, and early enrichment of EBOV actives at different false positive rates was calculated. Homology modelling and subsequent superimposition of binding site residues on other strains of EBOV were carried out to check residual conformations, and hence to confirm the efficacy of potential compounds. As a mechanism for artefactual inhibition of proteins through non-specific compounds, virtual hits were assessed for their aggregator potential compared with previously reported aggregators. These systematic studies have indicated that a few compounds may be effective inhibitors of EBOV replication and therefore might have the potential to be developed as anti-EBOV drugs after subsequent testing and validation in experiments in vivo.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Diseño Asistido por Computadora , Diseño de Fármacos , Ebolavirus/efectos de los fármacos , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Secuencia de Aminoácidos , Ebolavirus/metabolismo , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Humanos , Simulación del Acoplamiento Molecular , Alineación de Secuencia , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo
13.
Proteins ; 83(12): 2263-78, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26466951

RESUMEN

Ebola viruses (EBOVs) cause an acute and serious illness which is often fatal if untreated, and there is no effective vaccine until now. Multifunctional VP35 is critical for viral replication, RNA silencing suppression and nucleocapsid formation, and it is considered as a future target for the molecular biology technique. In the present work, the binding of inhibitor pyrrole-based compounds (GA017) to wild-type (WT), single (K248A, K251A, and I295A), and double (K248A/I295A) mutant VP35 were investigated by all-atom molecular dynamic (MD) simulations and Molecular Mechanics Generalized Born surface area (MM/GBSA) energy calculation. The calculated results indicate that the binding with GA017 makes the binding pocket more stable and reduces the space of the binding pocket. Moreover, the electrostatic interactions (ΔEele) and VDW energy (ΔEvdw) provide the major forces for affinity binding, and single mutation I295A and double mutation K248A/I295A have great influence on the conformation of the VP35 binding pocket. Interestingly, the residues R300-G301-D302 of I295A form a new helix and the sheet formed by the residues V294-I295-H296-I297 disappears in the double mutation K248A/I295A as compared with WT. Moreover, the binding free energy calculations show that I295A and K248A/I295A mutations decrease of absolute binding free energies while K248A and K251A mutations increase absolute binding free energy. Our calculated results are in good agreement with the experimental results that K248A/I295A double mutant results in near-complete loss of compound binding. The obtained information will be useful for design effective inhibitors for treating Ebola virus.


Asunto(s)
Antivirales/metabolismo , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/química , Antivirales/química , Sitios de Unión , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Mutación , Conformación Proteica , Pirroles/química , Pirroles/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
14.
J Theor Biol ; 386: 89-104, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26385832

RESUMEN

Human immunodeficiency virus-1 (HIV-1) employs accessory proteins to evade innate immune responses by neutralizing the anti-viral activity of host restriction factors. Apolipoprotein B mRNA-editing enzyme 3G (APOBEC3G, A3G) and bone marrow stromal cell antigen 2 (BST2) are host resistance factors that potentially inhibit HIV-1 infection. BST2 reduces viral production by tethering budding HIV-1 particles to virus producing cells, while A3G inhibits the reverse transcription (RT) process and induces viral genome hypermutation through cytidine deamination, generating fewer replication competent progeny virus. Two HIV-1 proteins counter these cellular restriction factors: Vpu, which reduces surface BST2, and Vif, which degrades cellular A3G. The contest between these host and viral proteins influences whether HIV-1 infection is established and progresses towards AIDS. In this work, we present an age-structured multi-scale viral dynamics model of in vivo HIV-1 infection. We integrated the intracellular dynamics of anti-viral activity of the host factors and their neutralization by HIV-1 accessory proteins into the virus/cell population dynamics model. We calculate the basic reproductive ratio (Ro) as a function of host-viral protein interaction coefficients, and numerically simulated the multi-scale model to understand HIV-1 dynamics following host factor-induced perturbations. We found that reducing the influence of Vpu triggers a drop in Ro, revealing the impact of BST2 on viral infection control. Reducing Vif׳s effect reveals the restrictive efficacy of A3G in blocking RT and in inducing lethal hypermutations, however, neither of these factors alone is sufficient to fully restrict HIV-1 infection. Interestingly, our model further predicts that BST2 and A3G function synergistically, and delineates their relative contribution in limiting HIV-1 infection and disease progression. We provide a robust modeling framework for devising novel combination therapies that target HIV-1 accessory proteins and boost antiviral activity of host factors.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Sinergismo Farmacológico , Quimioterapia Combinada , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/química , VIH-1/fisiología , Humanos , Carga Viral/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/análisis , Replicación Viral/efectos de los fármacos
15.
Nature ; 451(7177): 425-30, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18200009

RESUMEN

Human cells possess an antiviral activity that inhibits the release of retrovirus particles, and other enveloped virus particles, and is antagonized by the HIV-1 accessory protein, Vpu. This antiviral activity can be constitutively expressed or induced by interferon-alpha, and it consists of protein-based tethers, which we term 'tetherins', that cause retention of fully formed virions on infected cell surfaces. Using deductive constraints and gene expression analyses, we identify CD317 (also called BST2 or HM1.24), a membrane protein of previously unknown function, as a tetherin. Specifically, CD317 expression correlated with, and induced, a requirement for Vpu during HIV-1 and murine leukaemia virus particle release. Furthermore, in cells where HIV-1 virion release requires Vpu expression, depletion of CD317 abolished this requirement. CD317 caused retention of virions on cell surfaces and, after endocytosis, in CD317-positive compartments. Vpu co-localized with CD317 and inhibited these effects. Inhibition of Vpu function and consequent mobilization of tetherin's antiviral activity is a potential therapeutic strategy in HIV/AIDS.


Asunto(s)
Antígenos CD/metabolismo , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Antígenos CD/genética , Línea Celular , Membrana Celular/virología , Endocitosis , Proteínas Ligadas a GPI , Perfilación de la Expresión Génica , Infecciones por VIH/metabolismo , Infecciones por VIH/terapia , Infecciones por VIH/virología , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Humanos , Interferón-alfa/farmacología , Virus de la Leucemia Murina/metabolismo , Glicoproteínas de Membrana/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transporte de Proteínas , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Virión/metabolismo , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
16.
Planta Med ; 80(8-9): 682-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24963618

RESUMEN

Various drugs found in Chinese herbs are well known for their antiviral potency. We have tested several flavonoids with respect to their potency to block the viral protein U of the human immunodeficiency type 1 virus, which is believed to form a cation-permeable ion channel in the infected cell. We used Xenopus oocytes with heterologously expressed viral protein U as model system to test the efficacy of the drugs in voltage-clamp experiments. This method had been demonstrated in the past as a useful tool to screen drugs for their potency in inhibition of ion channel activity. The viral protein U-mediated current could be inhibited by Ba(2+) with a K1/2 value of 1.6 mM. Therefore, we determined viral protein U-mediated current as current component blocked by 10 mM Ba(2+). We screened several flavonoids with respect to their effects on this current. The flavonols quercetin and kaempferol, and the flavanols (-)epigallochatechin and (-)epichatechin were ineffective. The flavanone naringenin showed at 20 µM slight (about 10%) inhibition. The most potent drug was the isoflavon genistein which exhibited at 20 µM significant inhibition of about 40% with a K1/2 value of 81 ± 4 µM. We suggest that viral ion channels, in general, may be a good target for development of antiviral agents, and that, in particular, isoflavons may be candidates for development of drugs targeting viral protein U.


Asunto(s)
Antivirales/farmacología , Flavanonas/farmacología , Flavonoides/farmacología , Genisteína/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Animales , Antivirales/química , Femenino , Flavanonas/química , Flavonoides/química , Expresión Génica , Genisteína/química , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Humanos , Canales Iónicos/efectos de los fármacos , Técnicas de Placa-Clamp , ARN Complementario/genética , Transgenes , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Liberación del Virus/efectos de los fármacos , Xenopus laevis
17.
Biomolecules ; 14(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38927063

RESUMEN

The Ebola virus (EBOV) is a lethal pathogen causing hemorrhagic fever syndrome which remains a global health challenge. In the EBOV, two multifunctional proteins, VP35 and VP40, have significant roles in replication, virion assembly, and budding from the cell and have been identified as druggable targets. In this study, we employed in silico methods comprising molecular docking, molecular dynamic simulations, and pharmacological properties to identify prospective drugs for inhibiting VP35 and VP40 proteins from the myxobacterial bioactive natural product repertoire. Cystobactamid 934-2, Cystobactamid 919-1, and Cittilin A bound firmly to VP35. Meanwhile, 2-Hydroxysorangiadenosine, Enhypyrazinone B, and Sorangiadenosine showed strong binding to the matrix protein VP40. Molecular dynamic simulations revealed that, among these compounds, Cystobactamid 919-1 and 2-Hydroxysorangiadenosine had stable interactions with their respective targets. Similarly, molecular mechanics Poisson-Boltzmann surface area (MMPBSA) calculations indicated close-fitting receptor binding with VP35 or VP40. These two compounds also exhibited good pharmacological properties. In conclusion, we identified Cystobactamid 919-1 and 2-Hydroxysorangiadenosine as potential ligands for EBOV that target VP35 and VP40 proteins. These findings signify an essential step in vitro and in vivo to validate their potential for EBOV inhibition.


Asunto(s)
Antivirales , Productos Biológicos , Ebolavirus , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Ebolavirus/efectos de los fármacos , Productos Biológicos/farmacología , Productos Biológicos/química , Antivirales/farmacología , Antivirales/química , Myxococcales/química , Humanos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/metabolismo , Proteínas de la Matriz Viral/química , Proteínas de la Nucleocápside
18.
Biochemistry ; 52(47): 8406-19, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24067086

RESUMEN

Viral protein 35 (VP35), encoded by filoviruses, is a multifunctional dsRNA binding protein that plays important roles in viral replication, innate immune evasion, and pathogenesis. The multifunctional nature of these proteins also presents opportunities to develop countermeasures that target distinct functional regions. However, functional validation and the establishment of therapeutic approaches toward such multifunctional proteins, particularly for nonenzymatic targets, are often challenging. Our previous work on filoviral VP35 proteins defined conserved basic residues located within its C-terminal dsRNA binding interferon (IFN) inhibitory domain (IID) as important for VP35 mediated IFN antagonism and viral polymerase cofactor functions. In the current study, we used a combination of structural and functional data to determine regions of Ebola virus (EBOV) VP35 (eVP35) to target for aptamer selection using SELEX. Select aptamers, representing, two distinct classes, were further characterized based on their interaction properties to eVP35 IID. These results revealed that these aptamers bind to distinct regions of eVP35 IID with high affinity (10-50 nM) and specificity. These aptamers can compete with dsRNA for binding to eVP35 and disrupt the eVP35-nucleoprotein (NP) interaction. Consistent with the ability to antagonize the eVP35-NP interaction, select aptamers can inhibit the function of the EBOV polymerase complex reconstituted by the expression of select viral proteins. Taken together, our results support the identification of two aptamers that bind filoviral VP35 proteins with high affinity and specificity and have the capacity to potentially function as filoviral VP35 protein inhibitors.


Asunto(s)
Antivirales/química , Aptámeros de Nucleótidos/química , ARN/química , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Secuencia de Aminoácidos , Antivirales/metabolismo , Antivirales/farmacología , Aptámeros de Nucleótidos/metabolismo , Aptámeros de Nucleótidos/farmacología , Unión Competitiva , Secuencia Conservada , Ebolavirus/efectos de los fármacos , Ebolavirus/metabolismo , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/metabolismo , Cinética , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformación de Ácido Nucleico , Nucleoproteínas/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN/metabolismo , ARN/farmacología , ARN Bicatenario/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Técnica SELEX de Producción de Aptámeros , Especificidad de la Especie , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
19.
Biochemistry ; 51(6): 1288-96, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22264038

RESUMEN

Bone marrow stromal cell antigen 2 (BST-2) inhibits the release of enveloped viruses from the cell surface. Various viral counter measures have been discovered, which allow viruses to escape BST-2 restriction. Human immunodeficiency virus type 1 (HIV-1) encodes viral protein U (Vpu) that interacts with BST-2 through their transmembrane domains and causes the downregulation of cell surface BST-2. In this study, we used a computer modeling method to establish a molecular model to investigate the binding interface of the transmembrane domains of BST-2 and Vpu. The model predicts that the interface is composed of Vpu residues I6, A10, A14, A18, V25, and W22 and BST-2 residues L23, I26, V30, I34, V35, L41, I42, and T45. Introduction of mutations that have been previously reported to disrupt the Vpu-BST-2 interaction led to a calculated higher binding free energy (MMGBSA), which supports our molecular model. A pharmacophore was also generated on the basis of this model. Our results provide a precise model that predicts the detailed interaction occurring between the transmembrane domains of Vpu and BST-2 and should facilitate the design of anti-HIV agents that are able to disrupt this interaction.


Asunto(s)
Antígenos CD/química , Células de la Médula Ósea/química , Técnicas Químicas Combinatorias/métodos , VIH-1/química , Proteínas del Virus de la Inmunodeficiencia Humana/química , Modelos Moleculares , Complejos Multiproteicos/química , Proteínas Reguladoras y Accesorias Virales/química , Antígenos CD/genética , Membrana Celular/química , Membrana Celular/genética , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Simulación de Dinámica Molecular , Complejos Multiproteicos/genética , Valor Predictivo de las Pruebas , Unión Proteica/genética , Mapeo de Interacción de Proteínas , Multimerización de Proteína/genética , Estructura Secundaria de Proteína/genética , Células del Estroma/química , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Proteínas Reguladoras y Accesorias Virales/genética
20.
J Cell Physiol ; 227(8): 3119-27, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22034072

RESUMEN

An indispensable role for oligodendrocytes in the protection of axon function and promotion of neuronal survival is strongly supported by the finding of progressive neuron/axon degeneration in human neurological diseases that affect oligodendrocytes. Imaging and pathological studies of the CNS have shown the presence of neuroaxonal injury in progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the CNS, resulting from destruction of oligodendrocytes upon productive replication of the pathogenic neurotropic polyomavirus JC. Here, we examined the extracellular factors involved in communication between oligodendrocytes and neurons. Culturing cortical neurons with conditioned medium (CM) from rat CG4 oligodendrocytic cells that express the JCV agnoprotein showed that CXCL5/LIX, which is a chemokine closely related to the human CXCL5/ENA78 and CXCL6/GCP-2 chemokines, is essential for neuronal cell survival. We found that in CM from agnoprotein-producing CG-4 cells level of CXC5/LIX is decreased compared to control cells. We also demonstrated that a reduced expression of CXCL5/LIX by CG4 GFP-Agno cells triggered a cascade of signaling events in cortical neurons. Analysis of mitogen-activated protein kinases (MAPK) and glycogen synthase kinase (GSK3) pathways showed that they are involved in mechanisms of neuronal apoptosis in response to the depletion of CXCL5/LIX signaling. These data suggest that agnoprotein-induced dysregulation of chemokine production by oligodendrocytes may contribute to neuronal/axonal injury in the pathogenesis of PML lesions.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL5/metabolismo , Leucoencefalopatía Multifocal Progresiva/metabolismo , Neuronas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Células Cultivadas , Medios de Cultivo Condicionados , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Virus JC/metabolismo , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/patología , Leucoencefalopatía Multifocal Progresiva/virología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neuronas/citología , Oligodendroglía/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda