Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Virol ; 91(12)2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28381578

RESUMO

The NS2A protein of dengue virus (DENV) has eight predicted transmembrane segments (pTMS1 to -8) and participates in RNA replication, virion assembly, and host antiviral response. However, the roles of specific amino acid residues within the pTMS regions of NS2A during the viral life cycle are not clear. Here, we explore the function of DENV NS2A by introducing a series of alanine substitutions into the N-terminal half (pTMS1 to -4) of the protein in the context of a DENV infectious clone or subgenomic replicon. Six NS2A mutants (NM5, -7, -9, and -17 to -19) around pTMS1 and -2 displayed a novel phenotype showing a >1,000-fold reduction in virus yield, an absence of plaque formation despite wild-type-like replicon activity, and infectious-virus-like particle yields. HEK-293 cells infected with the six NS2A mutant viruses failed to cause a virus-induced cytopathic effect (CPE) by MitoCapture staining, cell proliferation, and lactate dehydrogenase release assays. Sequencing analyses of pseudorevertant viruses derived from lethal-mutant viruses revealed two consensus reversion mutations, leucine to phenylalanine at codon 181 (L181F) within pTMS7 of NS2A and isoleucine to threonine at codon 114 (I114T) within NS2B. The introduction of an NS2A-L181F mutation into the lethal (NM15, -16, -25, and -33) and CPE-defective (NM7, -9, and -19) mutants substantially rescued virus infectivity and virus-induced CPE, respectively, whereas the NS2B-L114T mutation rescued the NM16, -25, and -33 mutants. In conclusion, the results revealed the essential roles of the N-terminal half of NS2A in RNA replication and virus-induced CPE. Intramolecular interactions between pTMSs of NS2A and intermolecular interactions between the NS2A and NS2B proteins were also implicated.IMPORTANCE The characterization of the N-terminal (current study) and C-terminal halves of DENV NS2A is the most comprehensive mutagenesis study to date to investigate the function of NS2A during the flaviviral life cycle. A novel region responsible for virus-induced cytopathic effect (CPE) within pTMS1 and -2 of DENV NS2A was identified. Revertant genetics studies implied unexpected relationships between various pTMSs of DENV NS2A and NS2B. These results provide comprehensive information regarding the functions of DENV NS2A and the specific amino acids and transmembrane segments responsible for these functions. The positions and properties of the rescuing mutations were also revealed, providing important clues regarding the manner in which intramolecular or intermolecular interactions between the pTMSs of NS2A and NS2B regulate virus replication, assembly/secretion, and virus-induced CPE. These results expand the understanding of flavivirus replication. The knowledge may also facilitate studies of pathogenesis and novel vaccine and antiflaviviral drug development.


Assuntos
Efeito Citopatogênico Viral , Vírus da Dengue/genética , Mutagênese , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Alanina/metabolismo , Substituição de Aminoácidos , Proliferação de Células/genética , Vírus da Dengue/química , Vírus da Dengue/fisiologia , Células HEK293 , Humanos , L-Lactato Desidrogenase/metabolismo , Leucina/genética , Mutação , Fenilalanina/genética , RNA Viral/metabolismo , Análise de Sequência , Proteínas não Estruturais Virais/química , Montagem de Vírus , Replicação Viral/genética
2.
J Cell Physiol ; 232(12): 3409-3421, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28075010

RESUMO

In this study, we examine whether an anti-inflammatory thiourea derivative, compound #326, actions on ion channels. The effects of compound #326 on Ca2+ -activated K+ channels were evaluated by patch-clamp recordings obtained in cell-attached, inside-out or whole-cell configuration. In pituitary GH3 cells, compound #326 increased the amplitude of Ca2+ -activated K+ currents (IK(Ca) ) with an EC50 value of 11.6 µM, which was reversed by verruculogen, but not tolbutamide or TRAM-34. Under inside-out configuration, a bath application of compound #326 raised the probability of large-conductance Ca2+ -activated K+ (BKCa ) channels. The activation curve of BKCa channels was shifted to less depolarised potential with no modification of the gating charge of the curve; consequently, the difference of free energy was reduced in the presence of this compound. Compound #326-stimulated activity of BKCa channels is explained by a shortening of mean closed time, despite its inability to alter single-channel conductance. Neither delayed-rectifier nor erg-mediated K+ currents was modified. Compound #326 decreased the peak amplitude of voltage-gated Na+ current with no clear change in the overall current-voltage relationship of this current. In HEK293T cells expressing α-hSlo, compound #326 enhanced BKCa channels effectively. Intriguingly, the inhibitory actions of compound #326 on interleukin 1ß in lipopolysaccharide-activated microglia were significantly reversed by verruculogen, whereas BKCa channel inhibitors suppressed the expressions of inducible nitric oxide synthase. The BKCa channels could be an important target for compound #326 if similar in vivo results occur, and the multi-functionality of BKCa channels in modulating microglial immunity merit further investigation.


Assuntos
Anti-Inflamatórios/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/agonistas , Tioureia/farmacologia , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Lipopolissacarídeos/farmacologia , Potenciais da Membrana , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias Hipofisárias/metabolismo , Ratos , Tioureia/análogos & derivados , Transfecção
3.
J Antimicrob Chemother ; 71(7): 1922-32, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27098012

RESUMO

OBJECTIVES: Enterovirus 71 (EV-A71) is an important pathogen that can cause severe neurological symptoms and even death. Our aim was to identify potent anti-EV-A71 compounds and study their underlying mechanisms and in vivo activity. METHODS: We identified a potent imidazolidinone derivative (abbreviated to PR66) as an inhibitor of EV-A71 infection from the screening of compounds and subsequent structure-based modification. Time-course treatments and resistant virus selection of PR66 were employed to study the mode of mechanism of PR66. In vivo activity of PR66 was tested in the ICR strain of new-born mice challenged with EV-A71/4643/MP4. RESULTS: PR66 could impede the uncoating process during viral infection via interaction with capsid protein VP1, as shown by a resistant virus selection assay. Using site-directed mutagenesis, we confirmed that a change from valine to phenylalanine in the 179th amino acid residue of the cDNA-derived resistant virus resulted in resistance to PR66. PR66 increased the virion stability of WT viruses, but not the PR66-resistant mutant, in a particle stability thermal release assay. We further showed that PR66 had excellent anti-EV-A71 activity in an in vivo mouse model of disease, with a dose-dependent increase in survival rate and in protection against virus-induced hind-limb paralysis following oral or intraperitoneal administration. This was associated with reductions of viral titres in brain and muscle tissues. CONCLUSIONS: We demonstrated here for the first time that an imidazolidinone derivative (PR66) could protect against EV-A71-induced neurological symptoms in vivo by suppressing EV-A71 replication. This involved binding to and restricting viral uncoating.


Assuntos
Antivirais/metabolismo , Antivirais/farmacologia , Capsídeo/efeitos dos fármacos , Enterovirus Humano A/efeitos dos fármacos , Animais , Antivirais/isolamento & purificação , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Infecções por Enterovirus/tratamento farmacológico , Infecções por Enterovirus/virologia , Humanos , Concentração Inibidora 50 , Camundongos Endogâmicos ICR , Análise de Sobrevida
4.
Antimicrob Agents Chemother ; 58(1): 110-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24145533

RESUMO

Dengue virus (DENV) causes disease globally, resulting in an estimated 25 to 100 million new infections per year. No effective DENV vaccine is available, and the current treatment is only supportive. Thus, there is an urgent need to develop therapeutic agents to cure this epidemic disease. In the present study, we identified a potential small-molecule inhibitor, BP13944, via high-throughput screening (HTS) of 60,000 compounds using a stable cell line harboring an efficient luciferase replicon of DENV serotype 2 (DENV-2). BP13944 reduced the expression of the DENV replicon reporter in cells, showing a 50% effective concentration (EC50) of 1.03 ± 0.09 µM. Without detectable cytotoxicity, the compound inhibited replication or viral RNA synthesis in all four serotypes of DENV but not in Japanese encephalitis virus (JEV). Sequencing analyses of several individual clones derived from BP13944-resistant RNAs purified from cells harboring the DENV-2 replicon revealed a consensus amino acid substitution (E66G) in the region of the NS3 protease domain. Introduction of E66G into the DENV replicon, an infectious DENV cDNA clone, and recombinant NS2B/NS3 protease constructs conferred 15.2-, 17.2-, and 3.1-fold resistance to BP13944, respectively. Our results identify an effective small-molecule inhibitor, BP13944, which likely targets the DENV NS3 protease. BP13944 could be considered part of a more effective treatment regime for inhibiting DENV in the future.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Replicon/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Cricetinae , Vírus da Dengue/enzimologia , Farmacorresistência Viral , Serina Endopeptidases/metabolismo , Bibliotecas de Moléculas Pequenas
5.
Antimicrob Agents Chemother ; 57(2): 723-33, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23165461

RESUMO

Hepatitis C virus (HCV), a member of the Flaviviridae family, affects approximately 3% of the world's population and is becoming the leading cause of liver disease in the world. Therefore, the development of novel or more effective treatment strategies to treat chronic HCV infection is urgently needed. In our previous study, we identified a potential HCV NS5A inhibitor, BP008. After further systemic optimization, we discovered a more potent HCV inhibitor, DBPR110. DBPR110 reduced the reporter expression of the HCV1b replicon with a 50% effective concentration (EC(50)) and a selective index value of 3.9 ± 0.9 pM and >12,800,000, respectively. DBPR110 reduced HCV2a replicon activity with an EC(50) and a selective index value of 228.8 ± 98.4 pM and >173,130, respectively. Sequencing analyses of several individual clones derived from the DBPR110-resistant RNAs purified from cells harboring genotype 1b and 2a HCV replicons revealed that amino acid substitutions mainly within the N-terminal region (domain I) of NS5A were associated with decreased inhibitor susceptibility. P58L/T and Y93H/N in genotype 1b and T24A, P58L, and Y93H in the genotype 2a replicon were the key substitutions for resistance selection. In the 1b replicon, V153M, M202L, and M265V play a compensatory role in replication and drug resistance. Moreover, DBPR110 displayed synergistic effects with alpha interferon (IFN-α), an NS3 protease inhibitor, and an NS5B polymerase inhibitor. In summary, our results present an effective small-molecule inhibitor, DBPR110, that potentially targets HCV NS5A. DBPR110 could be part of a more effective therapeutic strategy for HCV in the future.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/genética , Hepatite C Crônica/tratamento farmacológico , Pirrolidinas/farmacologia , Tiazóis/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Substituição de Aminoácidos , Antivirais/química , Linhagem Celular Tumoral , Genótipo , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepatite C Crônica/virologia , Humanos , Interferon-alfa/farmacologia , Mutação , Ligação Proteica , RNA Viral/análise , Replicon , Análise de Sequência de RNA , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
6.
Antimicrob Agents Chemother ; 56(1): 44-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22006008

RESUMO

Hepatitis C virus (HCV) is a global health problem, affecting approximately 3% of the world's population. The standard treatment for HCV infection is often poorly tolerated and ineffective. Therefore, the development of novel or more effective treatment strategies to treat chronic HCV infection is urgently needed. In this report, BP008, a potent small-molecule inhibitor of HCV replication, was developed from a class of compounds with thiazol core structures by means of utilizing a cell-based HCV replicon system. The compound reduced the reporter expression of the HCV1b replicon with a 50% effective concentration (EC(50)) and selective index value of 4.1 ± 0.7 nM and >12,195, respectively. Sequencing analyses of several individual clones derived from BP008-resistant RNAs purified from cells harboring HCV1b replicon revealed that amino acid substitutions mainly within the N-terminal region (domain I) of NS5A were associated with decreased inhibitor susceptibility. Q24L, P58S, and Y93H are the key substitutions for resistance selection; F149L and V153M play the compensatory role in the replication and drug resistance processes. Moreover, BP008 displayed synergistic effects with alpha interferon (IFN-α), NS3 protease inhibitor, and NS5B polymerase inhibitor, as well as good oral bioavailability in SD rats and favorable exposure in rat liver. In summary, our results pointed to an effective small-molecule inhibitor, BP008, that potentially targets HCV NS5A. BP008 can be considered a part of a more effective therapeutic strategy for HCV in the future.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Tiazóis/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Genes Reporter , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatite C Crônica/virologia , Hepatócitos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Interferon-alfa/farmacologia , Masculino , Ratos , Replicon , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
7.
J Microbiol Immunol Infect ; 55(4): 598-610, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35650006

RESUMO

BACKGROUND: The purpose of this study was to examine the in vivo activity of rosmarinic acid (RA) - a phytochemical with antioxidant, anti-inflammatory, and antiviral properties - against influenza virus (IAV). An antibody-based kinase array and different in vitro functional assays were also applied to identify the mechanistic underpinnings by which RA may exert its anti-IAV activity. METHODS: We initially examined the potential efficacy of RA using an in vivo mouse model. A time-of-addition assay and an antibody-based kinase array were subsequently applied to investigate mechanism-of-action targets for RA. The hemagglutination inhibition assay, neuraminidase inhibition assay, and cellular entry assay were also performed. RESULTS: RA increased survival and prevented body weight loss in IAV-infected mice. In vitro experiments revealed that RA inhibited different IAV viruses - including oseltamivir-resistant strains. From a mechanistic point of view, RA downregulated the GSK3ß and Akt signaling pathways - which are known to facilitate IAV entry and replication into host cells. CONCLUSIONS: RA has promising preclinical efficacy against IAV, primarily by interfering with the GSK3ß and Akt signaling pathways.


Assuntos
Vírus da Influenza A , Influenza Humana , Animais , Antivirais , Cinamatos , Depsídeos , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Oseltamivir , Proteínas Proto-Oncogênicas c-akt , Replicação Viral , Ácido Rosmarínico
8.
Cancer Sci ; 102(1): 182-91, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21040217

RESUMO

BPR0C261 is a synthetic small molecule compound cytotoxic against human cancer cells and active prolonging the lifespan of leukemia mice. In the present study, we further investigated the mechanisms of its anticancer action and found that BPR0C261 inhibited microtubule polymerization through interacting with the colchicine binding sites on tubulins, disrupted microtubule arrangement and caused cell cycle arrest at G(2)/M phase in cancer cells. BPR0C261 also inhibited the clonogenic growths of cancer cells and showed cytotoxicity against human cervical cancer cells of multidrug-resistant phenotype. In addition, BPR0C261 concentration-dependently inhibited the proliferation and migration of HUVECs and disrupted the endothelial capillary-like tube formations in HUVEC and rat aorta ring cultures. Given orally, BPR0C261 inhibited angiogenesis in s.c. implanted Matrigel plugs in mice. Notably, its IC(50) values against the endothelial cell growths were approximately 10-fold lower than those against the cancer cells. It was found orally absorbable in mice and showed a good oral bioavailability (43%) in dogs. BPR0C261 permeated through the human intestinal Caco-2 cell monolayer, suggesting oral availability in humans. Orally absorbed BPR0C261 distributed readily into the s.c. xenografted tumors in nude mice in which the tumor tissue levels of BPR0C261 were found oral dose-dependent. BPR0C261 showed in vivo activities against human colorectal, gastric, and nasopharyngeal tumors in nude mice. Most interestingly, the combination of BPR0C261 plus cisplatin synergistically prolonged the lifespans of mice inoculated with murine leukemia cells. Thus, BPR0C261 is a novel orally active tubulin-binding antitumor agent with antimitotic, apoptosis-inducing, and vasculature disrupting activities.


Assuntos
Inibidores da Angiogênese/farmacologia , Antimitóticos/farmacologia , Antineoplásicos/farmacologia , Indóis/farmacologia , Tiazóis/farmacologia , Administração Oral , Animais , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cães , Humanos , Leucemia Experimental/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos , Microtúbulos/química , Microtúbulos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
9.
Antimicrob Agents Chemother ; 55(1): 229-38, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20937790

RESUMO

Dengue virus (DENV) causes disease globally, with an estimated 25 to 100 million new infections per year. At present, no effective vaccine is available, and treatment is supportive. In this study, we identified BP2109, a potent and selective small-molecule inhibitor of the DENV NS2B/NS3 protease, by a high-throughput screening assay using a recombinant protease complex consisting of the central hydrophilic portion of NS2B and the N terminus of the protease domain. BP2109 inhibited DENV (serotypes 1 to 4), but not Japanese encephalitis virus (JEV), replication and viral RNA synthesis without detectable cytotoxicity. The compound inhibited recombinant DENV-2 NS2B/NS3 protease with a 50% inhibitory concentration (IC(50)) of 15.43 ± 2.12 µM and reduced the reporter expression of the DENV-2 replicon with a 50% effective concentration (EC(50)) of 0.17 ± 0.01 µM. Sequencing analyses of several individual clones derived from BP2109-resistant DENV-2 RNAs revealed that two amino acid substitutions (R55K and E80K) are found in the region of NS2B, a cofactor of the NS2B/NS3 protease complex. The introduction of R55K and E80K double mutations into the dengue virus NS2B/NS3 protease and a dengue virus replicon construct conferred 10.3- and 73.8-fold resistance to BP2109, respectively. The E80K mutation was further determined to be the key mutation conferring dengue virus replicon resistance (61.3-fold) to BP2109, whereas the R55K mutation alone did not affect resistance to BP2109. Both the R55K and E80K mutations are located in the central hydrophilic portion of the NS2B cofactor, where extensive interactions with the NS3pro domain exist. Thus, our data provide evidence that BP2109 likely inhibits DENV by a novel mechanism.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Inibidores de Proteases/farmacologia , Animais , Cricetinae , Vírus da Dengue/crescimento & desenvolvimento , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Invest New Drugs ; 29(2): 195-206, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19960226

RESUMO

In our previous study, a series of novel cyclic cyanoguanidine compounds, eg. 5-substituted 2-cyanoimino-4-imidazodinone and 2-cyanoimino-4- pyrimidinone derivatives have been successfully synthesized and showed remarkable cytotoxicity in several cancer cell lines. In this present study, it is our aim to screen more potential candidates among the cyclic pyridyl cyanoguanidine compounds (BPR-DC-1, 2, 3) by in vitro and in vivo studies for the therapy of lung cancer, alternatively. Our results showed that BPR-DC-2 significantly inhibited proliferation of tumor cells with an IC50 of 3.60 ± 1.27 and 14.81 ± 4.23 µM in human lung carcinoma cells, H69 and A549, respectively by the MTT assay at 48 hr; BPR-DC-2 also obviously suppressed the tumor proliferation and MDR-1 gene expression, even induced cell apoptosis in the ex vivo histocultured lung tumor. We further demonstrated that, in the nude mouse model of metastatic lung cancer, BPR-DC-2 could diminish the tumor mass, retard the progression of metastasis, and prolong the survival time. In addition, it was found that BPR-DC-2 exerted its anti-tumor effects through the inhibition of MDR-1 gene expression and down-regulation of tumor anti-apoptosis signals (activated p-AKT and over-expression of PARP-1) by western blotting analysis. In conclusion, in this present study we have demonstrated that BPR-DC-2, derived from a series of novel synthetic cyclic cyanoguanidine compounds, has proved its potential as an anti-tumor drug candidate in treating lung cancer.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Regulação para Baixo , Guanidinas/uso terapêutico , Neoplasias Pulmonares/enzimologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Antígeno Carcinoembrionário/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Guanidinas/química , Guanidinas/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Análise de Sobrevida
11.
Bioorg Med Chem Lett ; 21(7): 1948-52, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21356589

RESUMO

A series of isatin-ß-thiosemicarbazones have been designed and evaluated for antiviral activity against herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) in a plaque reduction assay. Their cytotoxicity was examined using human rhabdomyosarcoma cells (RD cells). Several derivatives of isatin-ß-thiosemicarbazone exhibited significant and selective antiviral activity with low cytotoxicity. It was found that the thiourea group at thiosemicarbazone and the NH functionality at isatin were essential for their antiherpetic activity. The synthesis and structure-activity relationship studies are presented.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Isatina/análogos & derivados , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Isatina/farmacologia
12.
Bioorg Med Chem ; 18(17): 6414-21, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675142

RESUMO

A series of novel conformationally-restricted thiourea analogs were designed, synthesized, and evaluated for their anti-HCV activity. Herein we report the synthesis, structure-activity relationships (SARs), and pharmacokinetic properties of this new class of thiourea compounds that showed potent inhibitory activities against HCV in the cell-based subgenomic HCV replicon assay. Among compounds tested, the fluorene compound 4b was found to possess the most potent activity (EC(50)=0.3 microM), lower cytotoxicity (CC(50)>50 microM), and significantly better pharmacokinetic properties compared to its corresponding fluorenone compound 4c.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hepacivirus/fisiologia , Tioureia/análogos & derivados , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/farmacocinética , Linhagem Celular Tumoral , Fluorenos/química , Fluorenos/farmacocinética , Fluorenos/farmacologia , Humanos , Conformação Molecular , Ratos , Relação Estrutura-Atividade , Tioureia/síntese química , Tioureia/farmacocinética , Tioureia/farmacologia
13.
Emerg Microbes Infect ; 9(1): 1194-1205, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32397909

RESUMO

Enterovirus A71 (EV-A71), a positive-stranded RNA virus of the Picornaviridae family, may cause neurological complications or fatality in children. We examined specific factors responsible for this virulence using a chemical genetics approach. Known compounds from an anti-EV-A71 herbal medicine, Salvia miltiorrhiza (Danshen), were screened for anti-EV-A71. We identified a natural product, rosmarinic acid (RA), as a potential inhibitor of EV-A71 by cell-based antiviral assay and in vivo mouse model. Results also show that RA may affect the early stage of viral infection and may target viral particles directly, thereby interfering with virus-P-selectin glycoprotein ligand-1 (PSGL1) and virus-heparan sulfate interactions without abolishing the interaction between the virus and scavenger receptor B2 (SCARB2). Sequencing of the plaque-purified RA-resistant viruses revealed a N104K mutation in the five-fold axis of the structural protein VP1, which contains positively charged amino acids reportedly associated with virus-PSGL1 and virus-heparan sulfate interactions via electrostatic attraction. The plasmid-derived recombinant virus harbouring this mutation was confirmed to be refractory to RA inhibition. Receptor pull-down showed that this non-positively charged VP1-N104 is critical for virus binding to heparan sulfate. As the VP1-N104 residue is conserved among different EV-A71 strains, RA may be useful for inhibiting EV-A71 infection, even for emergent virus variants. Our study provides insight into the molecular mechanism of virus-host interactions and identifies a promising new class of inhibitors based on its antiviral activity and broad spectrum effects against a range of EV-A71.


Assuntos
Antivirais/administração & dosagem , Proteínas do Capsídeo/genética , Cinamatos/administração & dosagem , Depsídeos/administração & dosagem , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/tratamento farmacológico , Salvia miltiorrhiza/química , Animais , Antivirais/farmacologia , Proteínas do Capsídeo/antagonistas & inibidores , Proteínas do Capsídeo/química , Linhagem Celular , Cinamatos/farmacologia , Depsídeos/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/metabolismo , Infecções por Enterovirus/virologia , Heparitina Sulfato/metabolismo , Humanos , Células Jurkat , Glicoproteínas de Membrana/metabolismo , Camundongos , Mutação , Extratos Vegetais/administração & dosagem , Extratos Vegetais/farmacologia , Ligação Proteica/efeitos dos fármacos , Eletricidade Estática , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/química , Fatores de Virulência/genética , Ácido Rosmarínico
14.
Antimicrob Agents Chemother ; 53(7): 2740-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19414569

RESUMO

Enterovirus 71 (EV71) has emerged as an important virulent neurotropic enterovirus in young children. DTriP-22 (4{4-[(2-bromo-phenyl)-(3-methyl-thiophen-2-yl)-methyl]-piperazin-1-yl}-1-pheny-1H-pyrazolo[3,4-d]pyrimidine) was found to be a novel and potent inhibitor of EV71. The molecular target of this compound was identified by analyzing DTriP-22-resistant viruses. A substitution of lysine for Arg163 in EV71 3D polymerase rendered the virus drug resistant. DTriP-22 exhibited the ability to inhibit viral replication by reducing viral RNA accumulation. The compound suppressed the accumulated levels of both positive- and negative-stranded viral RNA during virus infection. An in vitro polymerase assay indicated that DTriP-22 inhibited the poly(U) elongation activity, but not the VPg uridylylation activity, of EV71 polymerase. These findings demonstrate that the nonnucleoside analogue DTriP-22 acts as a novel inhibitor of EV71 polymerase. DTriP-22 also exhibited a broad spectrum of antiviral activity against other picornaviruses, which highlights its potential in the development of antiviral agents.


Assuntos
Antivirais/farmacologia , Enterovirus/efeitos dos fármacos , Enterovirus/enzimologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Cães , Enterovirus/genética , Infecções por Enterovirus/tratamento farmacológico , Células HeLa , Humanos , RNA Polimerase Dependente de RNA/genética , Proteínas Recombinantes/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero
15.
Bioorg Med Chem Lett ; 19(7): 1950-5, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19251415

RESUMO

A series of thiourea derivatives were synthesized and their antiviral activity was evaluated in a cell-based HCV subgenomic replicon assay. SAR studies revealed that the chain length and the position of the alkyl linker largely influenced the in vitro anti-HCV activity of this class of potent antiviral agents. Among this series of compounds synthesized, the thiourea derivative with a six-carbon alkyl linker at the meta-position of the central phenyl ring (10) was identified as the most potent anti-HCV inhibitor (EC(50) = 0.047 microM) with a selectivity index of 596.


Assuntos
Antivirais/síntese química , Hepacivirus/efeitos dos fármacos , Tioureia/síntese química , Antivirais/química , Antivirais/farmacologia , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Replicon/genética , Relação Estrutura-Atividade , Tioureia/análogos & derivados , Tioureia/química , Tioureia/farmacologia
16.
Bioorg Med Chem Lett ; 19(15): 4134-8, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19539472

RESUMO

An efficient synthetic methodology to provide indole, 2,3-dihydro-indole, and 3,4-dihydro-2H-quinoline-1-carbothioic acid amide derivatives is described. These conformationally restricted heterobicyclic scaffolds were evaluated as a novel class of HCV inhibitors. Introduction of an acyl group at the NH(2) of the thiourea moiety has been found to enhance inhibitory activity. The chain length and the position of the alkyl group on the indoline aromatic ring markedly influenced anti-HCV activity. The indoline scaffold was more potent than the corresponding indole and tetrahydroquinoline scaffolds and analogue 31 displayed excellent activity (EC(50)=510nM) against HCV without significant cytotoxicity (CC(50) >50microM).


Assuntos
Antivirais/síntese química , Hepacivirus/metabolismo , Hepatite C/tratamento farmacológico , Indóis/síntese química , Quinolinas/síntese química , Antivirais/farmacologia , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Desenho de Fármacos , Humanos , Indóis/farmacologia , Modelos Químicos , Conformação Molecular , Estrutura Molecular , Estrutura Terciária de Proteína , Quinolinas/química , Quinolinas/farmacologia , Relação Estrutura-Atividade , Proteínas não Estruturais Virais/química
17.
Bioorg Med Chem Lett ; 19(21): 6063-8, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19796940

RESUMO

A novel class of arylthiourea HCV inhibitors bearing various functionalities, such as cyclic urea, cyclic thiourea, urea, and thiourea, on the alkyl linker were designed and synthesized. Herein we report the synthesis and structure-activity relationships (SARs) of this novel class of arylthiourea derivatives that showed potent inhibitory activities against HCV in the cell-based subgenomic HCV replicon assay. Among compounds tested, the new carbazole derivative 64, which has an eight-carbon linkage between the phenyl and carbazole rings and a tolyl group at the N-9 position of carbazole, was found to possess strong anti-HCV activity (EC50=0.031 microM), lower cytotoxicity (CC50 >50 microM), and higher selectivity index (SI >1612) compared to its derivatives.


Assuntos
Antivirais/síntese química , Carbazóis/síntese química , Hepacivirus/efeitos dos fármacos , Tioureia/análogos & derivados , Antivirais/química , Antivirais/toxicidade , Carbazóis/química , Carbazóis/farmacologia , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Relação Estrutura-Atividade , Tioureia/síntese química , Tioureia/química , Tioureia/farmacologia , Tioureia/toxicidade , Replicação Viral/efeitos dos fármacos
18.
Antiviral Res ; 172: 104636, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654671

RESUMO

Dengue virus (DENV) is a global health problem that affects approximately 3.9 billion people worldwide. Since safety concerns were raised for the only licensed vaccine, Dengvaxia, and since the present treatment is only supportive care, the development of more effective therapeutic anti-DENV agents is urgently needed. In this report, we identified a potential small-molecule inhibitor, BP34610, via cell-based high-throughput screening (HTS) of 12,000 compounds using DENV-2 reporter viruses. BP34610 reduced the virus yields of type 2 DENV-infected cells with a 50% effective concentration (EC50) and selectivity index value of 0.48 ±â€¯0.06 µM and 197, respectively. Without detectable cytotoxicity, the compound inhibited not only all four serotypes of DENV but also Japanese encephalitis virus (JEV). Time-of-addition experiments suggested that BP34610 may act at an early stage of DENV virus infection. Sequencing analyses of several individual clones derived from BP34610-resistant viruses revealed a consensus amino acid substitution (S397P) in the N-terminal stem region of the E protein. Introduction of S397P into the DENV reporter viruses conferred an over 14.8-fold EC90 shift for BP34610. Importantly, the combination of BP34610 with a viral replication inhibitor, ribavirin, displayed synergistic enhancement of anti-DENV-2 activity. Our results identify an effective small-molecule inhibitor, BP34610, which likely targets the DENV E protein. BP34610 could be developed as an anti-flavivirus agent in the future.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Proteínas do Envelope Viral/efeitos dos fármacos , Animais , Antivirais/toxicidade , Linhagem Celular , Dengue/tratamento farmacológico , Sinergismo Farmacológico , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Ribavirina/farmacologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
19.
Eur J Med Chem ; 167: 245-268, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30772607

RESUMO

A medicinal chemistry program based on the small-molecule HCV NS5A inhibitor daclatasvir has led to the discovery of dimeric phenylthiazole compound 8, a novel and potent HCV NS5A inhibitor. The subsequent SAR studies and optimization revealed that the cycloalkyl amide derivatives 27a-29a exhibited superior potency against GT1b with GT1b EC50 values at picomolar concentration. Interestingly, high diastereospecificity for HCV inhibition was observed in this class with the (1R,2S,1'R,2'S) diastereomer displaying the highest GT1b inhibitory activity. The best inhibitor 27a was found to be 3-fold more potent (GT1b EC50 = 0.003 nM) than daclatasvir (GT1b EC50 = 0.009 nM) against GT1b, and no detectable in vitro cytotoxicity was observed (CC50 > 50 µM). Pharmacokinetic studies demonstrated that compound 27a had an excellent pharmacokinetic profiles with a superior oral exposure and desired bioavailability after oral administration in both rats and dogs, and therefore it was selected as a developmental candidate for the treatment of HCV infection.


Assuntos
Descoberta de Drogas , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Tiazóis/farmacocinética , Proteínas não Estruturais Virais/antagonistas & inibidores , Amidas/química , Animais , Disponibilidade Biológica , Cães , Humanos , Ratos , Sialiltransferases/antagonistas & inibidores , Relação Estrutura-Atividade , Tiazóis/química , Tiazóis/uso terapêutico
20.
J Biomed Sci ; 15(3): 291-300, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18196474

RESUMO

Pyridyl imidazolidinone is a novel class of capsid binder which can inhibit enterovirus 71 (EV71). In this study, we tested the susceptibility of six recombinant viruses with different single-site mutations in VP1. Eleven modified pyridyl imidazolidinones were synthesized and used to probe the interaction between these compounds and the EV71 VP1 protein. We found that the D31N or E98K mutant viruses were susceptible to bulkier compounds, which suggested that mutations at these two sites in VP1 may widen the hydrophobic pocket of VP1, allowing bulkier compounds to enter and interfere VP1-receptor binding. Additionally, the Y116H mutant was more resistant to pyridyl imidazolidinone compounds containing a methyl group in the central position of the hydrophobic linker. When a trifluoromethyl group was substituted for the methyl group in the middle of the linker chain, the inhibitory effect was totally abolished in the Y116H mutant, suggesting that the interaction between Tyr (Y) 116 of VP1 and the central position of the linker chain of pyridyl imidazolodinone is very important for drug efficacy. A V192M mutant was resistant to most of the derivatives, indicating that residue 192 is a key mutation for resistance to pyridyl imidazolidinone.


Assuntos
Antivirais/farmacologia , Enterovirus/efeitos dos fármacos , Imidazolidinas/farmacologia , Animais , Chlorocebus aethiops , DNA Viral , Resistência Microbiana a Medicamentos/genética , Enterovirus/genética , Enterovirus/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Testes de Neutralização , Células Vero , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA