Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Biol Macromol ; 187: 976-987, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34333006

RESUMEN

Coronavirus 3C-like protease (3CLpro) is a crucial target for treating coronavirus diseases including COVID-19. Our preliminary screening showed that Ampelopsis grossedentata extract (AGE) displayed potent SARS-CoV-2-3CLpro inhibitory activity, but the key constituents with SARS-CoV-2-3CLpro inhibitory effect and their mechanisms were unrevealed. Herein, a practical strategy via integrating bioactivity-guided fractionation and purification, mass spectrometry-based peptide profiling and time-dependent biochemical assay, was applied to identify the crucial constituents in AGE and to uncover their inhibitory mechanisms. The results demonstrated that the flavonoid-rich fractions (10-17.5 min) displayed strong SARS-CoV-2-3CLpro inhibitory activities, while the constituents in these fractions were isolated and their SARS-CoV-2-3CLpro inhibitory activities were investigated. Among all isolated flavonoids, dihydromyricetin, isodihydromyricetin and myricetin strongly inhibited SARS-CoV-2 3CLpro in a time-dependent manner. Further investigations demonstrated that myricetin could covalently bind on SARS-CoV-2 3CLpro at Cys300 and Cys44, while dihydromyricetin and isodihydromyricetin covalently bound at Cys300. Covalent docking coupling with molecular dynamics simulations showed the detailed interactions between the orthoquinone form of myricetin and two covalent binding sites (surrounding Cys300 and Cys44) of SARS-CoV-2 3CLpro. Collectively, the flavonoids in AGE strongly and time-dependently inhibit SARS-CoV-2 3CLpro, while the newly identified SARS-CoV-2 3CLpro inhibitors in AGE offer promising lead compounds for developing novel antiviral agents.


Asunto(s)
Proteasas Virales 3C/química , Proteasas Virales 3C/metabolismo , Ampelopsis/química , Antivirales/farmacología , Flavonoides/farmacología , SARS-CoV-2/enzimología , Antivirales/química , Sitios de Unión/efectos de los fármacos , Cisteína/metabolismo , Flavonoides/química , Flavonoles/química , Flavonoles/farmacología , Espectrometría de Masas , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Extractos Vegetales/química , Extractos Vegetales/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos
2.
Int J Antimicrob Agents ; 53(2): 128-136, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30063999

RESUMEN

Human enterovirus 71 (EV-A71) infections cause a wide array of diseases ranging from diarrhoea and rashes to hand-foot-and-mouth disease and, in rare cases, severe neurological disorders. No specific antiviral drug therapy is currently available. Extracts from 75 Chinese medicinal plants selected for antiviral activity based on the Chinese pharmacopeia and advice from traditional Chinese medicine clinicians were tested for activity against EV-A71. The aqueous extract of the rhizome of Cimicifuga heracleifolia (Sheng Ma) and Arnebia euchroma (Zi Cao) showed potent antiviral activity. The active fractions were isolated by bioassay-guided purification, and identified by a combination of high-resolution mass spectrometry and nuclear magnetic resonance. Fukinolic acid and cimicifugic acid A and J, were identified as active anti-EV-A71 compounds for C. heracleifolia, whereas for A. euchroma, two caffeic acid derivatives were tentatively deduced. Commercially available fukinolic acid analogues such as L-chicoric acid and D-chicoric also showed in vitro micromolar activity against EV-A71 lab-strain and clinical isolates.


Asunto(s)
Antivirales/farmacología , Boraginaceae/química , Ácidos Cafeicos/farmacología , Cimicifuga/química , Enterovirus Humano A/efectos de los fármacos , Fenilacetatos/farmacología , Extractos Vegetales/farmacología , Succinatos/farmacología , Proteasas Virales 3C , Cisteína Endopeptidasas , Enterovirus Humano A/aislamiento & purificación , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/virología , Humanos , Espectrometría de Masas , Medicina Tradicional China , Pruebas de Sensibilidad Microbiana , Resonancia Magnética Nuclear Biomolecular , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
3.
J Gen Virol ; 98(3): 385-395, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27902359

RESUMEN

The foot-and-mouth disease virus (FMDV) capsid precursor, P1-2A, is cleaved by FMDV 3C protease to yield VP0, VP3, VP1 and 2A. Cleavage of the VP1/2A junction is the slowest. Serotype O FMDVs with uncleaved VP1-2A (having a K210E substitution in VP1; at position P2 in cleavage site) have been described previously and acquired a second site substitution (VP1 E83K) during virus rescue. Furthermore, introduction of the VP1 E83K substitution alone generated a second site change at the VP1/2A junction (2A L2P, position P2' in cleavage site). These virus adaptations have now been analysed using next-generation sequencing to determine sub-consensus level changes in the virus; this revealed other variants within the E83K mutant virus population that changed residue VP1 K210. The construction of serotype A viruses with a blocked VP1/2A cleavage site (containing K210E) has now been achieved. A collection of alternative amino acid substitutions was made at this site, and the properties of the mutant viruses were determined. Only the presence of a positively charged residue at position P2 in the cleavage site permitted efficient cleavage of the VP1/2A junction, consistent with analyses of diverse FMDV genome sequences. Interestingly, in contrast to the serotype O virus results, no second site mutations occurred within the VP1 coding region of serotype A viruses with the blocked VP1/2A cleavage site. However, some of these viruses acquired changes in the 2C protein that is involved in enterovirus morphogenesis. These results have implications for the testing of potential antiviral agents targeting the FMDV 3C protease.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Cisteína Endopeptidasas/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Fiebre Aftosa/virología , Proteínas Virales/metabolismo , Proteasas Virales 3C , Sustitución de Aminoácidos , Animales , Antivirales/farmacología , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Evaluación Preclínica de Medicamentos , Virus de la Fiebre Aftosa/efectos de los fármacos , Virus de la Fiebre Aftosa/genética , Ácido Glutámico/genética , Lisina/genética , Mutación , Ensamble de Virus/efectos de los fármacos
4.
Eur J Med Chem ; 124: 981-991, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27776325

RESUMEN

Hand, foot and mouth disease (HFMD) is a serious, highly contagious disease. HFMD caused by Enterovirus 71 (EV71), results in severe complications and even death. The pivotal role of EV71 3Cpro in the viral life cycle makes it an attractive target for drug discovery and development to treat HFMD. In this study, we identified novel EV71 3Cpro inhibitors by docking-based virtual screening. Totally 50 compounds were selected to test their inhibitory activity against EV71 3Cpro. The best inhibitor DC07090 exhibited the inhibition potency with an IC50 value of 21.72 ± 0.95 µM without apparent toxicity (CC50 > 200 µM). To explore structure-activity relationship of DC07090, 15 new derivatives were designed, synthesized and evaluated in vitro enzyme assay accordingly. Interestingly, four compounds showed inhibitory activities against EV71 3Cpro and only DC07090 inhibited EV71 replication with an EC50 value of 22.09 ± 1.07 µM. Enzyme inhibition kinetic experiments showed that the compound was a reversible and competitive inhibitor. The Ki value was determined to be 23.29 ± 12.08 µM. Further molecular docking, MD simulation and mutagenesis studies confirmed the binding mode of DC07090 and EV71 3Cpro. Besides, DC07090 could also inhibit coxsackievirus A16 (CVA16) replication with an EC50 value of 27.76 ± 0.88 µM. Therefore, DC07090 represents a new non-peptidyl small molecule inhibitor for further development of antiviral therapy against EV71 or other picornaviruses.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Enterovirus Humano A/enzimología , Oxazoles/química , Oxazoles/farmacología , Piridinas/química , Piridinas/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/metabolismo , Sitios de Unión , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Evaluación Preclínica de Medicamentos , Enterovirus Humano A/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Oxazoles/metabolismo , Conformación Proteica , Piridinas/metabolismo , Relación Estructura-Actividad , Interfaz Usuario-Computador , Proteínas Virales/química , Proteínas Virales/metabolismo
5.
Sci Rep ; 6: 33663, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27645381

RESUMEN

Hand, Foot and Mouth Disease is a highly contagious disease caused by a range of human enteroviruses. Outbreaks occur regularly, especially in the Asia-Pacific region, putting a burden on public healthcare systems. Currently, there is no antiviral for treating this infectious disease and the only vaccines are limited to circulation in China, presenting an unmet medical need that needs to be filled urgently. The human enterovirus 3 C protease has been deemed a plausible drug target due to its essential roles in viral replication. In this study, we designed and synthesized 10 analogues of the Rhinovirus 3 C protease inhibitor, Rupintrivir, and tested their 3 C protease inhibitory activities followed by a cellular assay using human enterovirus 71 (EV71)-infected human RD cells. Our results revealed that a peptide-based compound containing a trifluoromethyl moiety to be the most potent analogue, with an EC50 of 65 nM, suggesting its potential as a lead for antiviral drug discovery.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/enzimología , Péptidos/farmacología , Inhibidores de Proteasas/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/química , Línea Celular , Cisteína Endopeptidasas , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Enterovirus/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Péptidos/química , Inhibidores de Proteasas/química , Replicación Viral/efectos de los fármacos
6.
PLoS One ; 11(2): e0148693, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26870944

RESUMEN

Luteoloside is a member of the flavonoids family that exhibits several bioactivities including anti-microbial and anti-cancer activities. However, the antiviral activity of luteoloside against enterovirus 71 (EV71) and the potential mechanism(s) responsible for this effect remain unknown. In this study, the antiviral potency of luteoloside against EV71 and its inhibitory effects on 3C protease activity were evaluated. First, we investigated the cytotoxicity of luteoloside against rhabdomyosarcoma (RD) cells, which was the cell line selected for an in vitro infection model. In a subsequent antiviral assay, the cytopathic effect of EV71 was significantly and dose-dependently relieved by the administration of luteoloside (EC50 = 0.43 mM, selection index = 5.3). Using a plaque reduction assay, we administered luteoloside at various time points and found that the compound reduced EV71 viability in RD cells rather than increasing defensive mobilization or viral absorption. Moreover, biochemical studies focused on VP1 (a key structural protein of EV71) mRNA transcript and protein levels also revealed the inhibitory effects of luteoloside on the EV71 viral yield. Finally, we performed inhibition assays using luteoloside to evaluate its effect on recombinant 3C protease activity. Our results demonstrated that luteoloside blocked 3C protease enzymatic activity in a dose-dependent manner (IC50 = 0.36 mM) that was similar to the effect of rutin, which is a well-known C3 protease inhibitor. Collectively, the results from this study indicate that luteoloside can block 3C protease activity and subsequently inhibit EV71 production in vitro.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/farmacología , Enterovirus Humano A/efectos de los fármacos , Infecciones por Enterovirus/tratamiento farmacológico , Glucósidos/farmacología , Luteolina/farmacología , Proteínas Virales/química , Proteasas Virales 3C , Antivirales/química , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/química , Evaluación Preclínica de Medicamentos , Enterovirus Humano A/enzimología , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/virología , Humanos , Concentración 50 Inhibidora , Medicina Tradicional China , Proteínas Virales/antagonistas & inhibidores
7.
PLoS One ; 9(3): e89668, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24598537

RESUMEN

Enterovirus 71 (EV71) can cause severe disease and even lead to death in children, and an effective antiviral drug is currently unavailable. The anti-EV71 effect of chrysin (5,7-dihydroxyflavone), a natural flavonoid commonly found in many plants, was tested in this report. By using the predicting program Autodock 4.0 and an in vitro protease inhibition assay, we found that chrysin could suppress viral 3Cpro activity. Replication of viral RNA and production of viral capsid protein and the infectious virion were strongly inhibited by chrysin, without noticeable cytotoxicity. Cytopathic effects on cells were also prevented. Diisopropyl chrysin-7-yl phosphate (CPI), the phosphate ester for chrysin, was generated through a simplified Atheron-Todd reaction to achieve stronger anti-viral activity. CPI was also able to bind with and inhibit viral 3Cpro activity in vitro. As expected, CPI demonstrated more potent antiviral activity against EV71.


Asunto(s)
Antivirales/farmacología , Crisenos/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Enterovirus Humano A/fisiología , Flavonoides/farmacología , Organofosfatos/farmacología , Proteasas Virales 3C , Antivirales/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Crisenos/química , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/química , Evaluación Preclínica de Medicamentos , Enterovirus Humano A/efectos de los fármacos , Flavonoides/química , Humanos , Simulación del Acoplamiento Molecular , Organofosfatos/química , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Replicación Viral/efectos de los fármacos
8.
Antiviral Res ; 103: 17-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24393668

RESUMEN

Proteolytical cleavage of the picornaviral polyprotein is essential for viral replication. Therefore, viral proteases are attractive targets for anti-viral therapy. Most assays available for testing proteolytical activity of proteases are performed in vitro, using heterologously expressed proteases and peptide substrates. To deal with the disadvantages associated with in vitro assays, we modified a cell-based protease assay for picornavirus proteases. The assay is based on the induction of expression of a firefly luciferase reporter by a chimeric transcription factor in which the viral protease and cleavage sites are inserted between the GAL4 binding domain and the VP16 activation domain. Firefly luciferase expression is dependent on cleavage of the transcription factor by the viral protease. This biosafe assay enables testing the effect of compounds on protease activity in cells while circumventing the need for infection. We designed the assay for 3C proteases (3C(pro)) of various enteroviruses as well as of viruses of several other picornavirus genera, and show that the assay is amenable for use in a high-throughput setting. Furthermore, we show that the spectrum of activity of 3C(pro) inhibitor AG7088 (rupintrivir) not only encompasses enterovirus 3C(pro) but also 3C(pro) of foot-and-mouth disease virus (FMDV), an aphthovirus. In contrary, AG7404 (compound 1), an analogue of AG7088, had no effect on FMDV 3C(pro) activity, for which we provide a structural explanation.


Asunto(s)
Antivirales/aislamiento & purificación , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Picornaviridae/efectos de los fármacos , Picornaviridae/enzimología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Animales , Línea Celular , Cisteína Endopeptidasas , Genes Reporteros , Humanos , Luciferasas de Luciérnaga/análisis , Luciferasas de Luciérnaga/genética , Inhibidores de Proteasas/aislamiento & purificación , Inhibidores de Proteasas/farmacología
9.
Antiviral Res ; 101: 82-92, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24263113

RESUMEN

Enterovirus 71 (EV71), a member of Picornaviridae, is one of the major pathogens of human hand, foot and mouth disease. EV71 mainly infects children and causes severe neurological complications and even death. The pathogenesis of EV71 infection is largely unknown, and no clinically approved vaccine or effective treatment is available to date. Here we described a novel bioluminescence imaging approach for EV71 detection. In this approach, a plasmid-based reporter was constructed to express the fusion protein AmN(Q/G)BC, a split firefly luciferase mutant, which can be specifically cleaved by EV71 protease 3C(pro). Upon cleavage, the splitting fusion protein restores luciferase activity. Our test confirmed that AmN(Q/G)BC was specifically cleaved by 3C(pro) and EV71 and restored the luciferase activity to a degree that corresponds to the 3C(pro) and virus doses in cells and mice. The anti-EV71 effect of GW5074 and U0126, two mitogen-activated protein kinase (MAPK) inhibitors, was evaluated using this approach to validate its application of screening anti-EV71 agents. We found that the AmN(Q/G)BC reporter efficiently monitored the inhibitory effect of GW5074 and U0126 on EV71 infection under in vitro and in vivo conditions. The data from AmN(Q/G)BC reporter were consistent with Western blotting and histopathology examination. Taken together, this real-time imaging approach can quantitatively monitor the efficacy of anti-EV71 agents and is valuable for anti-EV71 drug screening and evaluation, especially, under in vivo conditions.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/efectos de los fármacos , Mediciones Luminiscentes/métodos , Imagen Óptica/métodos , Proteínas Virales/metabolismo , Proteasas Virales 3C , Animales , Antivirales/aislamiento & purificación , Línea Celular , Cisteína Endopeptidasas/genética , Evaluación Preclínica de Medicamentos/métodos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Ratones , Ratones Endogámicos ICR , Proteínas Virales/genética
10.
J Virol ; 87(21): 11955-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23986593

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is associated with an outbreak of more than 90 cases of severe pneumonia with high mortality (greater than 50%). To date, there are no antiviral drugs or specific therapies to treat MERS-CoV. To rapidly identify potential inhibitors of MERS-CoV replication, we expressed the papain-like protease (PLpro) and the 3-chymotrypsin-like protease (3CLpro) from MERS-CoV and developed luciferase-based biosensors to monitor protease activity in cells. We show that the expressed MERS-CoV PLpro recognizes and processes the canonical CoV-PLpro cleavage site RLKGG in the biosensor. However, existing CoV PLpro inhibitors were unable to block MERS-CoV PLpro activity, likely due to the divergence of the amino acid sequence in the drug binding site. To investigate MERS-CoV 3CLpro activity, we expressed the protease in context with flanking nonstructural protein 4 (nsp4) and the amino-terminal portion of nsp6 and detected processing of the luciferase-based biosensors containing the canonical 3CLpro cleavage site VRLQS. Importantly, we found that a small-molecule inhibitor that blocks replication of severe acute respiratory syndrome (SARS) CoV and murine CoV also inhibits the activity of MERS-CoV 3CLpro. Overall, the protease expression and biosensor assays developed here allow for rapid evaluation of viral protease activity and the identification of protease inhibitors. These biosensor assays can now be used to screen for MERS-CoV-specific or broad-spectrum coronavirus PLpro and 3CLpro inhibitors.


Asunto(s)
Antivirales/aislamiento & purificación , Técnicas Biosensibles/métodos , Coronavirus/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Luciferasas/análisis , Inhibidores de Proteasas/aislamiento & purificación , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/metabolismo , Línea Celular , Coronavirus/enzimología , Cisteína Endopeptidasas/metabolismo , Genes Reporteros , Humanos , Luciferasas/genética , Inhibidores de Proteasas/metabolismo , Proteínas Virales/metabolismo
11.
J Virol Methods ; 182(1-2): 93-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22465253

RESUMEN

Enterovirus A71 (EV-A71) causes severe complications: encephalitis, pulmonary edema, and death. No effective drug has been approved for clinical use. This study investigated the antiviral effects of flavonoids against EV-A71. An in vitro inhibitor screening assay using recombinant EV-A71 3C protease (3Cpro) demonstrated fisetin and rutin inhibiting 3Cpro enzymatic activity in a dose-dependent manner. Cell-based fluorescence resonance energy transfer (FRET) assay with an EV-A71 3Cpro cleavage motif probe also confirmed that fisetin and rutin inhibited the replication of EV-A71 in cells. A virus replication assay indicated that fisetin and rutin reduced significantly the EV-A71-induced cytopathic effect and viral plaque titers in RD cells culture. The IC(50) values of plaque reduction against EV-A71 were 85 µM for fisetin and 110 µM for rutin. Therapeutic indices (CC50/IC50 of plaque reduction assays) of fisetin and rutin exceeded 10. The study suggests that fisetin and rutin inhibit the replication of EV-A71.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/enzimología , Flavonoides/farmacología , Inhibidores de Proteasas/farmacología , Rutina/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Línea Celular , Cisteína Endopeptidasas , Efecto Citopatogénico Viral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Flavonoles , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Carga Viral , Ensayo de Placa Viral , Replicación Viral/efectos de los fármacos
12.
Vet Immunol Immunopathol ; 121(1-2): 83-90, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18006078

RESUMEN

The expression of recombinant antigens in transgenic plants is increasingly used as an alternative method of producing experimental immunogens. In this report, we describe the production of transgenic tomato plants that express the structural polyprotein, P1-2A, and protease, 3C, from foot-and-mouth disease (FMDV). P1-2A3C was inserted into the plant binary vector, pBin438, and transformed into tomato plants using Agrobacterium tumefaciens strain, GV3101. The presence of P1-2A3C was confirmed by PCR, transcription was verified by RT-PCR, and recombinant protein expression was confirmed by sandwich-ELISA and Western blot analyses. Guinea pigs immunized intramuscularly with foliar extracts from P1-2A3C-transgenic tomato plants were found to develop a virus-specific antibody response against FMDV. Vaccinated guinea pigs were fully protected against a challenge infection, while guinea pigs injected with untransformed plant extracts failed to elicit an antibody response and were not protected against challenge. These results demonstrate that transgenic tomato plants expressing the FMDV structural polyprotein, P1-2A, and the protease, 3C, can be used as a source of recombinant antigen for vaccine production.


Asunto(s)
Cisteína Endopeptidasas/inmunología , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/inmunología , Inmunización/métodos , Plantas Modificadas Genéticamente/química , Poliproteínas/inmunología , Proteínas Virales/inmunología , Proteasas Virales 3C , Animales , Anticuerpos Antivirales/sangre , Cisteína Endopeptidasas/genética , ADN Viral/genética , Ensayo de Inmunoadsorción Enzimática , Fiebre Aftosa/prevención & control , Virus de la Fiebre Aftosa/genética , Cobayas , Solanum lycopersicum/química , Solanum lycopersicum/genética , Extractos Vegetales/inmunología , Extractos Vegetales/farmacología , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/inmunología , Reacción en Cadena de la Polimerasa , Poliproteínas/genética , Distribución Aleatoria , Transcripción Genética , Transformación Genética , Proteínas Virales/genética
13.
Anal Biochem ; 343(1): 159-65, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15935325

RESUMEN

3C-like proteinase of severe acute respiratory syndrome (SARS) coronavirus has been demonstrated to be a key target for drug design against SARS. The interaction between SARS coronavirus 3C-like (3CL) proteinase and an octapeptide interface inhibitor was studied by affinity capillary electrophoresis (ACE). The binding constants were estimated by the change of migration time of the analytes in the buffer solution containing different concentrations of SARS 3CL proteinase. The results showed that SARS 3CL proteinase was able to complex with the octapeptide competitively, with binding constants of 2.44 x 10(4) M(-1) at 20 degrees C and 2.11 x 10(4)M(-1) at 37 degrees C. In addition, the thermodynamic parameters deduced reveal that hydrophobic interaction might play major roles, along with electrostatic force, in the binding process. The ACE method used here could be developed to be an effective and simple way of applying large-scale drug screening and evaluation.


Asunto(s)
Oligopéptidos/química , Inhibidores de Proteasas/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Dimerización , Evaluación Preclínica de Medicamentos/métodos , Electroforesis Capilar/métodos , Oligopéptidos/metabolismo , Inhibidores de Proteasas/metabolismo , Unión Proteica , Proteínas Virales/química , Proteínas Virales/metabolismo
14.
Bioorg Med Chem Lett ; 13(20): 3531-6, 2003 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-14505664

RESUMEN

We describe herein the synthesis and biological evaluation of a series of tripeptidyl alpha-ketoamides as human rhinovirus (HRV) 3C protease inhibitors. The most potent inhibitor discussed in this manuscript, 4I, exhibited impressive enzyme inhibitory activity as well as antiviral activity against HRV-14.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Amidas/química , Cisteína Endopeptidasas , Evaluación Preclínica de Medicamentos , Humanos , Inhibidores de Proteasas/química , Relación Estructura-Actividad
15.
Antiviral Res ; 49(1): 1-14, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11166856

RESUMEN

The common cold is an important illness both as a result of the economic impact of this common disease and because of the morbidity associated with the complications of the illness. Recent attempts to develop antiviral treatments for the common cold represent a substantial advance over previous efforts. Formidable barriers remain to be overcome, however, before any of these new products will be proven to be clinically useful. Recent advances in our understanding of the pathogenesis of common cold symptoms have provided insights into potential new targets for the treatment of this illness.


Asunto(s)
Antivirales/uso terapéutico , Resfriado Común/tratamiento farmacológico , Infecciones por Picornaviridae/tratamiento farmacológico , Rhinovirus/efectos de los fármacos , Proteínas Virales , Proteasas Virales 3C , Ensayos Clínicos como Asunto , Terapia Combinada , Resfriado Común/virología , Cisteína Endopeptidasas/metabolismo , Suplementos Dietéticos , Quimioterapia Combinada , Echinacea/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/uso terapéutico , Isoxazoles/uso terapéutico , Oxadiazoles/uso terapéutico , Oxazoles , Fenilalanina/análogos & derivados , Fitoterapia , Infecciones por Picornaviridae/virología , Extractos Vegetales/uso terapéutico , Plantas Medicinales , Pirrolidinonas/uso terapéutico , Receptores Virales/antagonistas & inhibidores , Receptores Virales/inmunología , Valina/análogos & derivados , Zinc/uso terapéutico
16.
J Med Chem ; 42(7): 1213-24, 1999 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-10197965

RESUMEN

The structure-based design, chemical synthesis, and biological evaluation of various human rhinovirus (HRV) 3C protease (3CP) inhibitors which incorporate P1 lactam moieties in lieu of an L-glutamine residue are described. These compounds are comprised of a tripeptidyl or peptidomimetic binding determinant and an ethyl propenoate Michael acceptor moiety which forms an irreversible covalent adduct with the active site cysteine residue of the 3C enzyme. The P1-lactam-containing inhibitors display significantly increased 3CP inhibition activity along with improved antirhinoviral properties relative to corresponding L-glutamine-derived molecules. In addition, several lactam-containing compounds exhibit excellent selectivity for HRV 3CP over several other serine and cysteine proteases and are not appreciably degraded by a variety of biological agents. One of the most potent inhibitors (AG7088, mean antirhinoviral EC90 approximately 0.10 microM, n = 46 serotypes) is shown to warrant additional preclinical development to explore its potential for use as an antirhinoviral agent.


Asunto(s)
Antivirales/síntesis química , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/síntesis química , Glutamina/química , Isoxazoles/síntesis química , Lactamas/síntesis química , Oligopéptidos/síntesis química , Pirrolidinonas/síntesis química , Rhinovirus/enzimología , Proteínas Virales , Proteasas Virales 3C , Antivirales/química , Antivirales/farmacología , Línea Celular , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Isoxazoles/química , Isoxazoles/farmacología , Lactamas/química , Lactamas/farmacología , Modelos Moleculares , Imitación Molecular , Oligopéptidos/química , Oligopéptidos/farmacología , Fenilalanina/análogos & derivados , Pirrolidinonas/química , Pirrolidinonas/farmacología , Rhinovirus/efectos de los fármacos , Relación Estructura-Actividad , Valina/análogos & derivados
17.
J Med Chem ; 41(15): 2819-34, 1998 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-9667971

RESUMEN

The structure-based design, chemical synthesis, and biological evaluation of various peptide-derived human rhinovirus (HRV) 3C protease (3CP) inhibitors are described. These compounds are comprised of an ethyl propenoate Michael acceptor moiety and a tripeptidyl binding determinant. The systematic modification of each amino acid residue present in the binding determinant as well as the N-terminal functionality is described. Such modifications are shown to provide irreversible HRV-14 3CP inhibitors with anti-3CP activities (kobs/[I]) ranging from 60 to 280 000 M-1 s-1 and antiviral EC50's which approach 0.15 microM. An optimized inhibitor which incorporates several improvements identified by the structure-activity studies is also described. This molecule displays very rapid irreversible inhibition of HRV-14 3CP (kobs/[I] = 800 000 M-1 s-1) and potent antiviral activity against HRV-14 in cell culture (EC50 = 0.056 microM). A 1.9 A crystal structure of an S-alkylthiocarbamate-containing inhibitor complexed with HRV-2 3CP is also detailed.


Asunto(s)
Antivirales , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa , Diseño de Fármacos , Oligopéptidos , Rhinovirus/efectos de los fármacos , Proteínas Virales , Proteasas Virales 3C , Antivirales/síntesis química , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , Línea Celular Transformada , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Oligopéptidos/síntesis química , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Rhinovirus/enzimología , Relación Estructura-Actividad
18.
J Biol Chem ; 269(45): 28429-35, 1994 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-7961784

RESUMEN

The encephalomyocarditis virus 3C protease has been shown to be rapidly degraded in infected cells and in vitro in rabbit reticulocyte lysate. The in vitro degradation, at least, is accomplished by a virus-independent, ATP-dependent proteolytic system. Here we identify this proteolytic system as the ubiquitin-mediated system. Incubation of the 3C protease in rabbit reticulocyte or cultured mouse cell lysate preparations, alone or in the presence of added ubiquitin or methylated ubiquitin, resulted in the generation of new higher molecular weight species. These new products were shown to be 3C protease-ubiquitin conjugates by their ability to bind antibodies against both the 3C protease and ubiquitin. Supplemental ubiquitin also stimulated the degradation of the 3C protease in these preparations. Large 3C protease-polyubiquitin conjugates were observed to accumulate in reticulocyte lysate in the presence of adenosine 5'-O-(3-thiotriphosphate), an inhibitor of the 26 S multicatalytic protease. This, combined with the fact that the proteolytic activity could be removed from the lysate by sedimentation, implicates the multicatalytic protease in the degradation of the 3C protease-ubiquitin conjugates. It was also found that the slow rate of degradation of a model polyprotein, which resembles the stable viral 3CD diprotein produced in vivo, is likely due to the fact that the polyprotein is a poor substrate for the ubiquitin-conjugating system.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Virus de la Encefalomiocarditis/enzimología , Ubiquitinas/metabolismo , Proteínas Virales , Proteasas Virales 3C , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Cisteína Endopeptidasas/biosíntesis , Electroforesis en Gel de Poliacrilamida , Estabilidad de Enzimas , Escherichia coli , Cinética , Datos de Secuencia Molecular , Mutagénesis Insercional , Biosíntesis de Proteínas , Conejos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Reticulocitos/metabolismo , Especificidad por Sustrato , Transcripción Genética , Ubiquitinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA