Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
BMC Vet Res ; 19(1): 26, 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: covidwho-2287307

RESUMEN

BACKGROUND: Porcine circovirus type 2 (PCV2) is one of the major pathogens commonly found in pigs, which causes immunosuppression and apoptosis. Vaccination and a single drug cannot totally prevent and treat PCV2 infection. Our previous in vitro study reported that the synergistic anti-PCV2 effect of Matrine and Osthole was better than that of Matrine or Osthole alone, This study was aimed to evaluate the synergistic anti-PCV2 effect as well as the underline molecular mechanism of Matrine and Osthole in Kunming (KM) mice model infected with PCV2. KM mice were randomly divided into 8 groups namely control group, PCV2 infected, Matrine combined with Osthole high dose treatment (40 mg/kg + 12 mg/kg), medium dose treatment (20 mg/kg + 6 mg/kg), low dose treatment (10 mg/kg + 3 mg/kg), Matrine treatment (40 mg/kg), Osthole treatment (12 mg/kg) and Ribavirin positive control (40 mg/kg) groups. PCV2 was intraperitoneally (i.p.) injected in all mice except the control group. 5 days of post-infection (dpi), mice in different treatment groups were injected i.p. with various doses of Matrine, Osthole and Ribavirin once daily for the next 5 consecutive days. RESULTS: The synergistic inhibitory effect of Matrine and Osthole on PCV2 replication in mouse liver was significantly heigher than that of Matrine and Osthole alone. The expression of GRP78, p-PERK, p-eIF2α, ATF4, CHOP, cleaved caspase-3 and Bax proteins were significantly reduced, while that of Bcl-2 was significantly increased in Matrine combined with Osthole groups, which alleviated the pathological changes caused by PCV2, such as interstitial pneumonia, loss of spleen lymphocytes, infiltration of macrophages and eosinophils. CONCLUSIONS: The synergistic anti-apoptotic effect of Matrine and Osthole was better than their alone effect, Both Matrine and Osthole had directly inhibited the expression of PCV2 Cap and the apoptosis of spleen cells induced by PCV2 Cap through the PERK pathway activated by endoplasmic reticulum (ER) GRP78. These results provided a new insight to control PCV2 infection and provide good component prescription candidate for the development of novel anti-PCV2 drugs.


Asunto(s)
Infecciones por Circoviridae , Circovirus , Matrinas , Animales , Ratones , Apoptosis , Infecciones por Circoviridae/tratamiento farmacológico , Infecciones por Circoviridae/patología , Chaperón BiP del Retículo Endoplásmico , Matrinas/farmacología , Ribavirina/farmacología , Bazo
2.
Antimicrob Agents Chemother ; 67(1): e0131522, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: covidwho-2245366

RESUMEN

We report that ribavirin exerts an inhibitory and mutagenic activity on SARS-CoV-2-infecting Vero cells, with a therapeutic index higher than 10. Deep sequencing analysis of the mutant spectrum of SARS-CoV-2 replicating in the absence or presence of ribavirin indicated an increase in the number of mutations, but not in deletions, and modification of diversity indices, expected from a mutagenic activity. Notably, the major mutation types enhanced by replication in the presence of ribavirin were A→G and U→C transitions, a pattern which is opposite to the dominance of G→A and C→U transitions previously described for most RNA viruses. Implications of the inhibitory activity of ribavirin, and the atypical mutational bias produced on SARS-CoV-2, for the search for synergistic anti-COVID-19 lethal mutagen combinations are discussed.


Asunto(s)
COVID-19 , Ribavirina , Animales , Chlorocebus aethiops , Ribavirina/farmacología , Ribavirina/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , SARS-CoV-2/genética , Células Vero , Mutación , Mutágenos/farmacología
3.
J Biol Chem ; 298(8): 102169, 2022 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1895142

RESUMEN

Remdesivir and molnupiravir have gained considerable interest because of their demonstrated activity against SARS-CoV-2. These antivirals are converted intracellularly to their active triphosphate forms remdesivir-TP and molnupiravir-TP. Cellular hydrolysis of these active metabolites would consequently decrease the efficiency of these drugs; however, whether endogenous enzymes that can catalyze this hydrolysis exist is unknown. Here, we tested remdesivir-TP as a substrate against a panel of human hydrolases and found that only Nudix hydrolase (NUDT) 18 catalyzed the hydrolysis of remdesivir-TP with notable activity. The kcat/Km value of NUDT18 for remdesivir-TP was determined to be 17,700 s-1M-1, suggesting that NUDT18-catalyzed hydrolysis of remdesivir-TP may occur in cells. Moreover, we demonstrate that the triphosphates of the antivirals ribavirin and molnupiravir are also hydrolyzed by NUDT18, albeit with lower efficiency than Remdesivir-TP. Low activity was also observed with the triphosphate forms of sofosbuvir and aciclovir. This is the first report showing that NUDT18 hydrolyzes triphosphates of nucleoside analogs of exogenous origin, suggesting that NUDT18 can act as a cellular sanitizer of modified nucleotides and may influence the antiviral efficacy of remdesivir, molnupiravir, and ribavirin. As NUDT18 is expressed in respiratory epithelial cells, it may limit the antiviral efficacy of remdesivir and molnupiravir against SARS-CoV-2 replication by decreasing the intracellular concentration of their active metabolites at their intended site of action.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/metabolismo , Antivirales/farmacología , Citidina/análogos & derivados , Humanos , Hidrólisis , Hidroxilaminas , Polifosfatos , Pirofosfatasas , Ribavirina/farmacología , Ribavirina/uso terapéutico , SARS-CoV-2
4.
Sci Rep ; 11(1): 19998, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1462031

RESUMEN

Understanding the effects of metabolism on the rational design of novel and more effective drugs is still a considerable challenge. To the best of our knowledge, there are no entirely computational strategies that make it possible to predict these effects. From this perspective, the development of such methodologies could contribute to significantly reduce the side effects of medicines, leading to the emergence of more effective and safer drugs. Thereby, in this study, our strategy is based on simulating the electron ionization mass spectrometry (EI-MS) fragmentation of the drug molecules and combined with molecular docking and ADMET models in two different situations. In the first model, the drug is docked without considering the possible metabolic effects. In the second model, each of the intermediates from the EI-MS results is docked, and metabolism occurs before the drug accesses the biological target. As a proof of concept, in this work, we investigate the main antiviral drugs used in clinical research to treat COVID-19. As a result, our strategy made it possible to assess the biological activity and toxicity of all potential by-products. We believed that our findings provide new chemical insights that can benefit the rational development of novel drugs in the future.


Asunto(s)
Antivirales/metabolismo , Tratamiento Farmacológico de COVID-19 , Descubrimiento de Drogas , SARS-CoV-2/efectos de los fármacos , Adenina/efectos adversos , Adenina/análogos & derivados , Adenina/metabolismo , Adenina/farmacología , Adenosina/efectos adversos , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Adenosina Monofosfato/efectos adversos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Alanina/efectos adversos , Alanina/análogos & derivados , Alanina/metabolismo , Alanina/farmacología , Amidas/efectos adversos , Amidas/metabolismo , Amidas/farmacología , Antivirales/efectos adversos , Antivirales/farmacología , COVID-19/metabolismo , Cloroquina/efectos adversos , Cloroquina/análogos & derivados , Cloroquina/metabolismo , Cloroquina/farmacología , Diseño de Fármacos , Humanos , Redes y Vías Metabólicas , Simulación del Acoplamiento Molecular , Nitrocompuestos/efectos adversos , Nitrocompuestos/metabolismo , Nitrocompuestos/farmacología , Pirazinas/efectos adversos , Pirazinas/metabolismo , Pirazinas/farmacología , Pirrolidinas/efectos adversos , Pirrolidinas/metabolismo , Pirrolidinas/farmacología , Ribavirina/efectos adversos , Ribavirina/metabolismo , Ribavirina/farmacología , SARS-CoV-2/metabolismo , Tiazoles/efectos adversos , Tiazoles/metabolismo , Tiazoles/farmacología
5.
Viruses ; 13(4)2021 04 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1389536

RESUMEN

Replication of RNA viruses is characterized by exploration of sequence space which facilitates their adaptation to changing environments. It is generally accepted that such exploration takes place mainly in response to positive selection, and that further diversification is boosted by modifications of virus population size, particularly bottleneck events. Our recent results with hepatitis C virus (HCV) have shown that the expansion in sequence space of a viral clone continues despite prolonged replication in a stable cell culture environment. Diagnosis of the expansion was based on the quantification of diversity indices, the occurrence of intra-population mutational waves (variations in mutant frequencies), and greater individual residue variations in mutant spectra than those anticipated from sequence alignments in data banks. In the present report, we review our previous results, and show additionally that mutational waves in amplicons from the NS5A-NS5B-coding region are equally prominent during HCV passage in the absence or presence of the mutagenic nucleotide analogues favipiravir or ribavirin. In addition, by extending our previous analysis to amplicons of the NS3- and NS5A-coding region, we provide further evidence of the incongruence between amino acid conservation scores in mutant spectra from infected patients and in the Los Alamos National Laboratory HCV data banks. We hypothesize that these observations have as a common origin a permanent state of HCV population disequilibrium even upon extensive viral replication in the absence of external selective constraints or changes in population size. Such a persistent disequilibrium-revealed by the changing composition of the mutant spectrum-may facilitate finding alternative mutational pathways for HCV antiviral resistance. The possible significance of our model for other genetically variable viruses is discussed.


Asunto(s)
Hepacivirus/genética , Hepacivirus/fisiología , Hepatitis C/virología , Antivirales/farmacología , COVID-19 , Línea Celular , Farmacorresistencia Viral/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Humanos , Mutación , ARN Viral , Ribavirina/farmacología , Análisis de Secuencia , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos
6.
Mol Divers ; 26(1): 279-292, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: covidwho-1148916

RESUMEN

Since the outbreak emerged in November 2019, no effective drug has yet been found against SARS-CoV-2. Repositioning studies of existing drug molecules or candidates are gaining in overcoming COVID-19. Antiviral drugs such as remdesivir, favipiravir, ribavirin, and galidesivir act by inhibiting the vital RNA polymerase of SARS-CoV-2. The importance of in silico studies in repurposing drug research is gradually increasing during the COVID-19 process. The present study found that especially ribavirin triphosphate and galidesivir triphosphate active metabolites had a higher affinity for SARS-CoV-2 RNA polymerase than ATP by molecular docking. With the Molecular Dynamics simulation, we have observed that these compounds increase the complex's stability and validate the molecular docking results. We also explained that the interaction of RNA polymerase inhibitors with Mg++, which is in the structure of NSP12, is essential and necessary to interact with the RNA strand. In vitro and clinical studies on these two molecules need to be increased.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Ribavirina , Adenina/análogos & derivados , Adenosina/análogos & derivados , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Amidas , Antivirales/química , Reposicionamiento de Medicamentos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pirazinas , Pirrolidinas , ARN Viral , Ribavirina/farmacología , SARS-CoV-2
7.
Can J Physiol Pharmacol ; 99(5): 449-460, 2021 May.
Artículo en Inglés | MEDLINE | ID: covidwho-1125455

RESUMEN

Ribavirin is a guanosine analog with broad-spectrum antiviral activity against RNA viruses. Based on this, we aimed to show the anti-SARS-CoV-2 activity of this drug molecule via in vitro, in silico, and molecular techniques. Ribavirin showed antiviral activity in Vero E6 cells following SARS-CoV-2 infection, whereas the drug itself did not show any toxic effect over the concentration range tested. In silico analysis suggested that ribavirin has a broad-spectrum impact on SARS-CoV-2, acting at different viral proteins. According to the detailed molecular techniques, ribavirin was shown to decrease the expression of TMPRSS2 at both mRNA and protein levels 48 h after treatment. The suppressive effect of ribavirin in ACE2 protein expression was shown to be dependent on cell types. Finally, proteolytic activity assays showed that ribavirin also showed an inhibitory effect on the TMPRSS2 enzyme. Based on these results, we hypothesized that ribavirin may inhibit the expression of TMPRSS2 by modulating the formation of inhibitory G-quadruplex structures at the TMPRSS2 promoter. As a conclusion, ribavirin is a potential antiviral drug for the treatment against SARS-CoV-2, and it interferes with the effects of TMPRSS2 and ACE2 expression.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , Regulación hacia Abajo/efectos de los fármacos , Ribavirina/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Animales , Células CACO-2 , Chlorocebus aethiops , G-Cuádruplex/efectos de los fármacos , Humanos , Regiones Promotoras Genéticas/genética , SARS-CoV-2/fisiología , Serina Endopeptidasas/genética , Células Vero
8.
Viruses ; 13(2)2021 02 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1060766

RESUMEN

The long-term control strategy of SARS-CoV-2 and other major respiratory viruses needs to include antivirals to treat acute infections, in addition to the judicious use of effective vaccines. Whilst COVID-19 vaccines are being rolled out for mass vaccination, the modest number of antivirals in use or development for any disease bears testament to the challenges of antiviral development. We recently showed that non-cytotoxic levels of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, induces a potent host innate immune antiviral response that blocks influenza A virus replication. Here we show that TG is also highly effective in blocking the replication of respiratory syncytial virus (RSV), common cold coronavirus OC43, SARS-CoV-2 and influenza A virus in immortalized or primary human cells. TG's antiviral performance was significantly better than remdesivir and ribavirin in their respective inhibition of OC43 and RSV. Notably, TG was just as inhibitory to coronaviruses (OC43 and SARS-CoV-2) and influenza viruses (USSR H1N1 and pdm 2009 H1N1) in separate infections as in co-infections. Post-infection oral gavage of acid-stable TG protected mice against a lethal influenza virus challenge. Together with its ability to inhibit the different viruses before or during active infection, and with an antiviral duration of at least 48 h post-TG exposure, we propose that TG (or its derivatives) is a promising broad-spectrum inhibitor against SARS-CoV-2, OC43, RSV and influenza virus.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Coronavirus Humano OC43/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Virus Sincitial Respiratorio Humano/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Tapsigargina/farmacología , Animales , Antivirales/uso terapéutico , Betacoronavirus/fisiología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Coronavirus Humano OC43/fisiología , Estrés del Retículo Endoplásmico , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Virus Sincitial Respiratorio Humano/fisiología , Ribavirina/farmacología , SARS-CoV-2/fisiología , Tapsigargina/uso terapéutico , Replicación Viral/efectos de los fármacos
9.
Ann Palliat Med ; 10(1): 707-720, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: covidwho-1030457

RESUMEN

The whole world is battling through coronavirus disease 2019 (COVID-19) which is a fatal pandemic. In the early 2020, the World Health Organization (WHO) declared it as a global health emergency without definitive treatments and preventive approaches. In the absence of definitive therapeutic agents, this thorough review summarizes and outlines the potency and safety of all molecules and therapeutics which may have potential antiviral effects. A number of molecules and therapeutics licensed or being tested for some other conditions were found effective in different in vitro studies as well as in many small sample-sized clinical trials and independent case studies. However, in those clinical trials, there were some limitations which need to be overcome to find the most promising antiviral against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In conclusion, many of above-mentioned antivirals seems to have some therapeutic effects but none of them have been shown to have a strong evidence for their proper recommendation and approval in the treatment of COVID-19. Constantly evolving new evidences, exclusive adult data, language barrier, and type of study (observational, retrospective, small-sized clinical trials, or independent case series) resulted to the several limitations of this review. The need for multicentered, large sample-sized, randomized, placebo-controlled trials on COVID-19 patients to reach a proper conclusion on the most promising antiviral agent is warranted.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/terapia , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Amidas/farmacología , Amidas/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Azetidinas/farmacología , Azetidinas/uso terapéutico , Cloroquina/farmacología , Cloroquina/uso terapéutico , Combinación de Medicamentos , Humanos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Inmunización Pasiva , Indoles/farmacología , Indoles/uso terapéutico , Interferones/farmacología , Interferones/uso terapéutico , Ivermectina/farmacología , Ivermectina/uso terapéutico , Lopinavir/farmacología , Lopinavir/uso terapéutico , Nitrocompuestos , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , Purinas/farmacología , Purinas/uso terapéutico , Pirazinas/farmacología , Pirazinas/uso terapéutico , Pirazoles/farmacología , Pirazoles/uso terapéutico , Ribavirina/farmacología , Ribavirina/uso terapéutico , Ritonavir/farmacología , Ritonavir/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Tiazoles/farmacología , Tiazoles/uso terapéutico , Sueroterapia para COVID-19
10.
Viruses ; 13(1)2020 12 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1004758

RESUMEN

RNA viruses have gained plenty of attention during recent outbreaks of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Zika virus (ZIKV), and Ebola virus. ZIKV is a vector borne Flavivirus that is spread by mosquitoes and it mainly infects neuronal progenitor cells. One hallmark of congenital ZIKV disease is a reduced brain size in fetuses, leading to severe neurological defects. The World Health Organization (WHO) is urging the development of new antiviral treatments against ZIKV, as there are no efficient countermeasures against ZIKV disease. Previously, we presented a new class of host-targeting antivirals active against a number of pathogenic RNA viruses, such as SARS-CoV-2. Here, we show the transfer of the image-based phenotypic antiviral assay to ZIKV-infected brain cells, followed by mechanism-of-action studies and a proof-of-concept study in a three-dimensional (3D) organoid model. The novel antiviral compounds showed a therapeutic window against ZIKV in several cell models and rescued ZIKV-induced neurotoxicity in brain organoids. The compound's mechanism-of-action was pinpointed to late steps in the virus life cycle, impairing the formation of new virus particles. Collectively, in this study, we expand the antiviral activity of new small molecule inhibitors to a new virus class of Flaviviruses, but also uncover compounds' mechanism of action, which are important for the further development of antivirals.


Asunto(s)
Antivirales/farmacología , Encéfalo/metabolismo , Organoides/metabolismo , Infección por el Virus Zika/metabolismo , Virus Zika/efectos de los fármacos , Animales , Encéfalo/patología , COVID-19 , Supervivencia Celular/efectos de los fármacos , Humanos , Organoides/patología , Virus ARN , Ribavirina/farmacología , SARS-CoV-2 , Virus Zika/fisiología , Infección por el Virus Zika/virología
11.
EMBO Mol Med ; 13(1): e13105, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: covidwho-814824

RESUMEN

The ongoing SARS-CoV-2 pandemic stresses the need for effective antiviral drugs that can quickly be applied in order to reduce morbidity, mortality, and ideally viral transmission. By repurposing of broadly active antiviral drugs and compounds that are known to inhibit viral replication of related viruses, several advances could be made in the development of treatment strategies against COVID-19. The nucleoside analog remdesivir, which is known for its potent in vitro activity against Ebolavirus and other RNA viruses, was recently shown to reduce the time to recovery in patients with severe COVID-19. It is to date the only approved antiviral for treating COVID-19. Here, we provide a mechanism and evidence-based comparative review of remdesivir and other repurposed drugs with proven in vitro activity against SARS-CoV-2.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , Amidas/farmacología , Amidas/uso terapéutico , Antivirales/farmacología , Benzamidinas , Reposicionamiento de Medicamentos/métodos , Ésteres/farmacología , Ésteres/uso terapéutico , Guanidinas/farmacología , Guanidinas/uso terapéutico , Guanina/farmacología , Guanina/uso terapéutico , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Lopinavir/farmacología , Lopinavir/uso terapéutico , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Pirazinas/farmacología , Pirazinas/uso terapéutico , Ribavirina/farmacología , Ribavirina/uso terapéutico , Ritonavir/farmacología , Ritonavir/uso terapéutico , SARS-CoV-2/fisiología , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
12.
J Nanosci Nanotechnol ; 20(12): 7311-7323, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: covidwho-680345

RESUMEN

We started a study on the molecular docking of six potential pharmacologically active inhibitors compounds that can be used clinically against the COVID-19 virus, in this case, remdesivir, ribavirin, favipiravir, galidesivir, hydroxychloroquine and chloroquine interacting with the main COVID-19 protease in complex with a COVID-19 N3 protease inhibitor. The highest values of affinity energy found in order from highest to lowest were chloroquine (CHL), hydroxychloroquine (HYC), favipiravir (FAV), galidesivir (GAL), remdesivir (REM) and ribavirin (RIB). The possible formation of hydrogen bonds, associations through London forces and permanent electric dipole were analyzed. The values of affinity energy obtained for the hydroxychloroquine ligands was -9.9 kcal/mol and for the chloroquine of -10.8 kcal/mol which indicate that the coupling contributes to an effective improvement of the affinity energies with the protease. Indicating that, the position chosen to make the substitutions may be a pharmacophoric group, and cause changes in the protease.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Betacoronavirus/enzimología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Cisteína Endopeptidasas/química , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Adenina/administración & dosificación , Adenina/análogos & derivados , Adenina/química , Adenina/farmacología , Adenosina/análogos & derivados , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Alanina/administración & dosificación , Alanina/análogos & derivados , Alanina/química , Alanina/farmacología , Amidas/administración & dosificación , Amidas/química , Amidas/farmacología , Antivirales/administración & dosificación , Sitios de Unión , COVID-19 , Cloroquina/administración & dosificación , Cloroquina/química , Cloroquina/farmacología , Proteasas 3C de Coronavirus , Interacciones Farmacológicas , Humanos , Enlace de Hidrógeno , Hidroxicloroquina/administración & dosificación , Hidroxicloroquina/química , Hidroxicloroquina/farmacología , Ligandos , Simulación del Acoplamiento Molecular , Nanotecnología , Pandemias , Inhibidores de Proteasas/administración & dosificación , Pirazinas/administración & dosificación , Pirazinas/química , Pirazinas/farmacología , Pirrolidinas/administración & dosificación , Pirrolidinas/química , Pirrolidinas/farmacología , Ribavirina/administración & dosificación , Ribavirina/química , Ribavirina/farmacología , SARS-CoV-2 , Electricidad Estática , Tratamiento Farmacológico de COVID-19
13.
Pathog Dis ; 78(4)2020 06 01.
Artículo en Inglés | MEDLINE | ID: covidwho-646518

RESUMEN

The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) around the world has led to a pandemic with high morbidity and mortality. However, there are no effective drugs to prevent and treat the disease. Transcriptome-based drug repositioning, identifying new indications for old drugs, is a powerful tool for drug development. Using bronchoalveolar lavage fluid transcriptome data of COVID-19 patients, we found that the endocytosis and lysosome pathways are highly involved in the disease and that the regulation of genes involved in neutrophil degranulation was disrupted, suggesting an intense battle between SARS-CoV-2 and humans. Furthermore, we implemented a coexpression drug repositioning analysis, cogena, and identified two antiviral drugs (saquinavir and ribavirin) and several other candidate drugs (such as dinoprost, dipivefrine, dexamethasone and (-)-isoprenaline). Notably, the two antiviral drugs have also previously been identified using molecular docking methods, and ribavirin is a recommended drug in the diagnosis and treatment protocol for COVID pneumonia (trial version 5-7) published by the National Health Commission of the P.R. of China. Our study demonstrates the value of the cogena-based drug repositioning method for emerging infectious diseases, improves our understanding of SARS-CoV-2-induced disease, and provides potential drugs for the prevention and treatment of COVID-19 pneumonia.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Reposicionamiento de Medicamentos , Neumonía Viral/tratamiento farmacológico , Ribavirina/farmacología , Saquinavir/farmacología , Líquido del Lavado Bronquioalveolar/química , COVID-19 , Degranulación de la Célula/inmunología , Endocitosis/inmunología , Perfilación de la Expresión Génica , Humanos , Lisosomas/inmunología , Simulación del Acoplamiento Molecular , Activación Neutrófila/inmunología , Pandemias , SARS-CoV-2 , Transcriptoma
14.
Int J Mol Sci ; 21(11)2020 May 30.
Artículo en Inglés | MEDLINE | ID: covidwho-437471

RESUMEN

The novel coronavirus, COVID-19, caused by SARS-CoV-2, is a global health pandemic that started in December 2019. The effective drug target among coronaviruses is the main protease Mpro, because of its essential role in processing the polyproteins that are translated from the viral RNA. In this study, the bioactivity of some selected heterocyclic drugs named Favipiravir (1), Amodiaquine (2), 2'-Fluoro-2'-deoxycytidine (3), and Ribavirin (4) was evaluated as inhibitors and nucleotide analogues for COVID-19 using computational modeling strategies. The density functional theory (DFT) calculations were performed to estimate the thermal parameters, dipole moment, polarizability, and molecular electrostatic potential of the present drugs; additionally, Mulliken atomic charges of the drugs as well as the chemical reactivity descriptors were investigated. The nominated drugs were docked on SARS-CoV-2 main protease (PDB: 6LU7) to evaluate the binding affinity of these drugs. Besides, the computations data of DFT the docking simulation studies was predicted that the Amodiaquine (2) has the least binding energy (-7.77 Kcal/mol) and might serve as a good inhibitor to SARS-CoV-2 comparable with the approved medicines, hydroxychloroquine, and remdesivir which have binding affinity -6.06 and -4.96 Kcal/mol, respectively. The high binding affinity of 2 was attributed to the presence of three hydrogen bonds along with different hydrophobic interactions between the drug and the critical amino acids residues of the receptor. Finally, the estimated molecular electrostatic potential results by DFT were used to illustrate the molecular docking findings. The DFT calculations showed that drug 2 has the highest of lying HOMO, electrophilicity index, basicity, and dipole moment. All these parameters could share with different extent to significantly affect the binding affinity of these drugs with the active protein sites.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/química , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/química , Amidas/química , Amidas/farmacología , Amodiaquina/química , Amodiaquina/farmacología , Antivirales/química , Sitios de Unión , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/metabolismo , Inhibidores de Proteasas/química , Unión Proteica , Pirazinas/química , Pirazinas/farmacología , Ribavirina/química , Ribavirina/farmacología , Proteínas no Estructurales Virales/metabolismo
15.
Antiviral Res ; 178: 104786, 2020 06.
Artículo en Inglés | MEDLINE | ID: covidwho-30820

RESUMEN

An escalating pandemic by the novel SARS-CoV-2 virus is impacting global health and effective therapeutic options are urgently needed. We evaluated the in vitro antiviral effect of compounds that were previously reported to inhibit coronavirus replication and compounds that are currently under evaluation in clinical trials for SARS-CoV-2 patients. We report the antiviral effect of remdesivir, lopinavir, homorringtonine, and emetine against SARS-CoV-2 virus in Vero E6 cells with the estimated 50% effective concentration at 23.15 µM, 26.63 µM, 2.55 µM and 0.46 µM, respectively. Ribavirin or favipiravir that are currently evaluated under clinical trials showed no inhibition at 100 µM. Synergy between remdesivir and emetine was observed, and remdesivir at 6.25 µM in combination with emetine at 0.195 µM may achieve 64.9% inhibition in viral yield. Combinational therapy may help to reduce the effective concentration of compounds below the therapeutic plasma concentrations and provide better clinical benefits.


Asunto(s)
Antimetabolitos/farmacología , Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Emetina/farmacología , Homoharringtonina/farmacología , Lopinavir/farmacología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Replicación Viral/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Amidas/farmacología , Animales , Betacoronavirus/fisiología , COVID-19 , Chlorocebus aethiops , Combinación de Medicamentos , Células Epiteliales , Humanos , Pandemias , Pirazinas/farmacología , Ribavirina/farmacología , SARS-CoV-2 , Células Vero , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA