Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 88(3): 1740-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24257621

RESUMO

In a recent clinical trial, balapiravir, a prodrug of a cytidine analog (R1479), failed to achieve efficacy (reducing viremia after treatment) in dengue patients, although the plasma trough concentration of R1479 remained above the 50% effective concentration (EC(50)). Here, we report experimental evidence to explain the discrepancy between the in vitro and in vivo results and its implication for drug development. R1479 lost its potency by 125-fold when balapiravir was used to treat primary human peripheral blood mononuclear cells (PBMCs; one of the major cells targeted for viral replication) that were preinfected with dengue virus. The elevated EC(50) was greater than the plasma trough concentration of R1479 observed in dengue patients treated with balapiravir and could possibly explain the efficacy failure. Mechanistically, dengue virus infection triggered PBMCs to generate cytokines, which decreased their efficiency of conversion of R1479 to its triphosphate form (the active antiviral ingredient), resulting in decreased antiviral potency. In contrast to the cytidine-based compound R1479, the potency of an adenosine-based inhibitor of dengue virus (NITD008) was much less affected. Taken together, our results demonstrate that viral infection in patients before treatment could significantly affect the conversion of the prodrug to its active form; such an effect should be calculated when estimating the dose efficacious for humans.


Assuntos
Antivirais/administração & dosagem , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Dengue/virologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Nucleosídeos/administração & dosagem , Animais , Citidina/administração & dosagem , Citidina/análogos & derivados , Citocinas/imunologia , Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Feminino , Humanos , Camundongos , Nucleosídeos/farmacologia , Pró-Fármacos/administração & dosagem
2.
Methods Mol Biol ; 1030: 269-81, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23821275

RESUMO

Dengue fever is an emerging mosquito-borne flaviviral disease that threatens 2.5 billion people worldwide. No clinically approved vaccine and antiviral therapy are currently available to prevent or treat dengue virus (DENV) infection. Vertebrate animals other than primates are not normally infectable with DENV; however, a small animal dengue infection model would greatly facilitate the development of a vaccine or an antiviral therapy. To this end, a rodent model for DENV infection has been established in IFN-α/ß and IFN-γ receptor-deficient (AG129) mice. This chapter describes the protocol for the DENV infection model in AG129 mice and testing of antiviral compounds by oral gavage or parenteral injection.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Animais , Antivirais/administração & dosagem , Linhagem Celular , Dengue/imunologia , Dengue/patologia , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Esquema de Medicação , Feminino , Masculino , Camundongos
3.
Sci Transl Med ; 4(139): 139ra83, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22723463

RESUMO

Dengue virus (DENV) is a mosquito-borne flavivirus that affects 2.5 billion people worldwide. There are four dengue serotypes (DENV1 to DENV4), and infection with one elicits lifelong immunity to that serotype but offers only transient protection against the other serotypes. Identification of the protective determinants of the human antibody response to DENV is a vital requirement for the design and evaluation of future preventative therapies and treatments. Here, we describe the isolation of a neutralizing antibody from a DENV1-infected patient. The human antibody 14c10 (HM14c10) binds specifically to DENV1. HM14c10 neutralizes the virus principally by blocking virus attachment; at higher concentrations, a post-attachment step can also be inhibited. In vivo studies show that the HM14c10 antibody has antiviral activity at picomolar concentrations. A 7 Å resolution cryoelectron microscopy map of Fab fragments of HM14c10 in a complex with DENV1 shows targeting of a discontinuous epitope that spans the adjacent surface of envelope protein dimers. As found previously, a human antibody specific for the related West Nile virus binds to a similar quaternary structure, suggesting that this could be an immunodominant epitope. These findings provide a structural and molecular context for durable, serotype-specific immunity to DENV infection.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Antivirais/uso terapêutico , Vírus da Dengue/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/farmacologia , Antivirais/farmacologia , Microscopia Crioeletrônica , Dengue/tratamento farmacológico , Vírus da Dengue/imunologia , Vírus da Dengue/ultraestrutura , Epitopos/imunologia , Humanos , Camundongos
4.
Antiviral Res ; 92(2): 369-71, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21854808

RESUMO

Dengue virus infection causes diseases in people, ranging from the acute febrile illness dengue fever, to life-threatening dengue hemorrhagic fever/dengue shock syndrome. We previously reported that a host cellular α-glucosidases I and II inhibitor, imino sugar CM-10-18, potently inhibited dengue virus replication in cultured cells, and significantly reduced viremia in dengue virus infected AG129 mice. In this report we show that CM-10-18 also significantly protects mice from death and/or disease progress in two mouse models of lethal dengue virus infection. Our results thus provide a strong support for the development of CM-10-18 or its derivatives as antiviral agents to treat servere dengue virus infections.


Assuntos
Antivirais/administração & dosagem , Dengue/tratamento farmacológico , Dengue/prevenção & controle , Inibidores Enzimáticos/administração & dosagem , Inibidores de Glicosídeo Hidrolases , Animais , Dengue/mortalidade , Dengue/patologia , Camundongos , Análise de Sobrevida , Resultado do Tratamento
5.
Antimicrob Agents Chemother ; 55(9): 4072-80, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21730119

RESUMO

We describe a novel translation inhibitor that has anti-dengue virus (DENV) activity in vitro and in vivo. The inhibitor was identified through a high-throughput screening using a DENV infection assay. The compound contains a benzomorphan core structure. Mode-of-action analysis indicated that the compound inhibits protein translation in a viral RNA sequence-independent manner. Analysis of the stereochemistry demonstrated that only one enantiomer of the racemic compound inhibits viral RNA translation. Medicinal chemistry was performed to eliminate a metabolically labile glucuronidation site of the compound to improve its in vivo stability. Pharmacokinetic analysis showed that upon a single subcutaneous dosing of 25 mg/kg of body weight in mice, plasma levels of the compound reached a C(max) (maximum plasma drug concentration) above the protein-binding-adjusted 90% effective concentration (EC(90)) value of 0.96 µM. In agreement with the in vivo pharmacokinetic results, treatment of DENV-infected mice with 25 mg/kg of compound once per day reduced peak viremia by about 40-fold. However, mice treated with 75 mg/kg of compound per day exhibited adverse effects. Collectively, our results demonstrate that the benzomorphan compounds inhibit DENV through suppression of RNA translation. The therapeutic window of the current compounds needs to be improved for further development.


Assuntos
Antivirais/farmacologia , Antivirais/farmacocinética , Vírus da Dengue/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Antivirais/química , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Cricetinae , Vírus da Dengue/genética , Feminino , Humanos , Camundongos , Estrutura Molecular , RNA Viral/genética , Ratos , Células Vero
6.
J Virol ; 85(15): 7775-87, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21632767

RESUMO

Dengue (DEN) is a mosquito-borne viral disease that has become an increasing economic and health burden for the tropical and subtropical world. The lack of an appropriate animal model of DEN has greatly impeded the study of its pathogenesis and the development of vaccines/antivirals. We recently reported a DEN virus 2 (DENV-2) strain (D2Y98P) that lethally infects immunocompromised AG129 mice, resulting in organ damage or dysfunction and increased vascular permeability, hallmarks of severe DEN in patients (G. K. Tan et al., PLoS Negl. Trop. Dis. 4:e672, 2010). Here we report the identification of one critical virulence determinant of strain D2Y98P. By mutagenesis, we showed that a Phe-to-Leu alteration at amino acid position 52 in nonstructural protein NS4B completely abolished the pathogenicity of the D2Y98P virus, as evidenced by a lack of lethality and the absence of histological signs of disease, which correlated with reduced viral titers and intact vascular permeability. Conversely, a Leu-to-Phe alteration at position 52 of NS4B in nonvirulent DENV-2 strain TSV01 led to 80% lethality and increased viremia. The NS4B(Phe52) viruses displayed enhanced RNA synthesis in mammalian cells but not in mosquito cells. The increased viral RNA synthesis was independent of the ability of NS4B to interfere with the host type I interferon response. Overall, our results demonstrate that Phe at position 52 in NS4B confers virulence in mice on two independent DENV-2 strains through enhancement of viral RNA synthesis. In addition to providing further insights into the functional role of NS4B protein, our findings further support a direct relationship between viral loads and DEN pathogenesis in vivo, consistent with observations in DEN patients.


Assuntos
Aminoácidos/química , DNA Viral/biossíntese , Vírus da Dengue/patogenicidade , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Vírus da Dengue/genética , Imunofluorescência , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas não Estruturais Virais/química , Virulência
7.
J Virol ; 85(13): 6548-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21507975

RESUMO

Viral replication relies on the host to supply nucleosides. Host enzymes involved in nucleoside biosynthesis are potential targets for antiviral development. Ribavirin (a known antiviral drug) is such an inhibitor that suppresses guanine biosynthesis; depletion of the intracellular GTP pool was shown to be the major mechanism to inhibit flavivirus. Along similar lines, inhibitors of the pyrimidine biosynthesis pathway could be targeted for potential antiviral development. Here we report on a novel antiviral compound (NITD-982) that inhibits host dihydroorotate dehydrogenase (DHODH), an enzyme required for pyrimidine biosynthesis. The inhibitor was identified through screening 1.8 million compounds using a dengue virus (DENV) infection assay. The compound contains an isoxazole-pyrazole core structure, and it inhibited DENV with a 50% effective concentration (EC(50)) of 2.4 nM and a 50% cytotoxic concentration (CC(50)) of >5 µM. NITD-982 has a broad antiviral spectrum, inhibiting both flaviviruses and nonflaviviruses with nanomolar EC(90)s. We also show that (i) the compound inhibited the enzymatic activity of recombinant DHODH, (ii) an NITD-982 analogue directly bound to the DHODH protein, (iii) supplementing the culture medium with uridine reversed the compound-mediated antiviral activity, and (iv) DENV type 2 (DENV-2) variants resistant to brequinar (a known DHODH inhibitor) were cross resistant to NITD-982. Collectively, the results demonstrate that the compound inhibits DENV through depleting the intracellular pyrimidine pool. In contrast to the in vitro potency, the compound did not show any efficacy in the DENV-AG129 mouse model. The lack of in vivo efficacy is likely due to the exogenous uptake of pyrimidine from the diet or to a high plasma protein-binding activity of the current compound.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Pirimidinas/antagonistas & inibidores , Animais , Antivirais/química , Antivirais/farmacocinética , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , Dengue/virologia , Vírus da Dengue/enzimologia , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , Di-Hidro-Orotato Desidrogenase , Modelos Animais de Doenças , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Pirimidinas/biossíntese , Sigmodontinae , Resultado do Tratamento , Células Vero , Replicação Viral/efeitos dos fármacos
8.
Antiviral Res ; 89(1): 26-34, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21073903

RESUMO

Cellular α-glucosidases I and II are enzymes that sequentially trim the three terminal glucoses in the N-linked oligosaccharides of viral envelope glycoproteins. This process is essential for the proper folding of viral glycoproteins and subsequent assembly of many enveloped viruses, including dengue virus (DENV). Imino sugars are substrate mimics of α-glucosidases I and II. In this report, we show that two oxygenated alkyl imino sugar derivatives, CM-9-78 and CM-10-18, are potent inhibitors of both α-glucosidases I and II in vitro and in treated animals, and efficiently inhibit DENV infection of cultured human cells. Pharmacokinetic studies reveal that both compounds are well tolerated at doses up to 100mg/kg in rats and have favorable pharmacokinetic properties and bioavailability in mice. Moreover, we showed that oral administration of either CM-9-78 or CM-10-18 reduces the peak viremia of DENV in mice. Interestingly, while treatment of DENV infected mice with ribavirin alone did not reduce the viremia, combination therapy of ribavirin with sub-effective dose of CM-10-18 demonstrated a significantly enhanced antiviral activity, as indicated by a profound reduction of the viremia. Our findings thus suggest that combination therapy of two broad-spectrum antiviral agents may provide a practically useful approach for the treatment of DENV infection.


Assuntos
Antivirais/administração & dosagem , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Inibidores de Glicosídeo Hidrolases , Ribavirina/administração & dosagem , Administração Oral , Animais , Antivirais/farmacocinética , Antivirais/farmacologia , Linhagem Celular , Dengue/prevenção & controle , Dengue/virologia , Modelos Animais de Doenças , Quimioterapia Combinada , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Concentração Inibidora 50 , Camundongos , Ribavirina/farmacologia , Viremia/prevenção & controle
9.
Antimicrob Agents Chemother ; 54(8): 3255-61, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20516277

RESUMO

Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen that infects humans. Neither a vaccine nor an antiviral therapy is currently available for DENV. Here, we report an adenosine nucleoside prodrug that potently inhibits DENV replication both in cell culture and in a DENV mouse model. NITD449 (2'-C-acetylene-7-deaza-7-carbamoyladenosine) was initially identified as a parental compound that inhibits all four serotypes of DENV with low cytotoxicity. However, in vivo pharmacokinetic studies indicated that NITD449 had a low level of exposure in plasma when dosed orally. To increase the oral bioavailability, we covalently linked isobutyric acids to the 3'- and 5'-hydroxyl groups of ribose via ester linkage to NITD449, leading to the prodrug NITD203 (3',5'-O-diisobutyryl-2'-C-acetylene-7-deaza-7-carbamoyl-adenosin). Pharmacokinetic analysis showed that upon oral dosing of the prodrug, NITD203 was readily converted to NITD449, resulting in improved exposure of the parental compound in plasma in both mouse and rat. In DENV-infected AG129 mice, oral dosing of the prodrug at 25 mg/kg of body weight reduced peak viremia by 30-fold. Antiviral spectrum analysis showed that NITD203 inhibited various flaviviruses (DENV, yellow fever virus, and West Nile virus) and hepatitis C virus but not Chikungunya virus (an alphavirus). Mode-of-action analysis, using a luciferase-reporting replicon, indicated that NITD203 inhibited DENV RNA synthesis. Although NITD203 exhibited potent in vitro and in vivo efficacies, the compound could not reach a satisfactory no-observable-adverse-effect level (NOAEL) in a 2-week in vivo toxicity study. Nevertheless, our results demonstrate that a prodrug approach using a nucleoside analog could potentially be developed for flavivirus antiviral therapy.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Pró-Fármacos/farmacologia , Replicação Viral/efeitos dos fármacos , Adenosina/farmacocinética , Adenosina/farmacologia , Adenosina/uso terapêutico , Animais , Antivirais/química , Antivirais/farmacocinética , Antivirais/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Dengue/virologia , Células Epiteliais/virologia , Ésteres/química , Humanos , Camundongos , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Ratos , Células Vero
10.
Antimicrob Agents Chemother ; 54(7): 2932-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20457821

RESUMO

We recently reported that (2R,3R,4R,5R)-2-(4-amino-pyrrolo[2,3-d]pyrimidin-7-yl)-3-ethynyl-5-hydroxy-methyl-tetrahydro-furan-3,4-diol is a potent inhibitor of dengue virus (DENV), with 50% effective concentration (EC(50)) and cytotoxic concentration (CC(50)) values of 0.7 microM and >100 microM, respectively. Here we describe the synthesis, structure-activity relationship, and antiviral characterization of the inhibitor. In an AG129 mouse model, a single-dose treatment of DENV-infected mice with the compound suppressed peak viremia and completely prevented death. Mode-of-action analysis using a DENV replicon indicated that the compound blocks viral RNA synthesis. Recombinant adenosine kinase could convert the compound to a monophosphate form. Suppression of host adenosine kinase, using a specific inhibitor (iodotubercidin) or small interfering RNA (siRNA), abolished or reduced the compound's antiviral activity in cell culture. Studies of rats showed that (14)C-labeled compound was converted to mono-, di-, and triphosphate metabolites in vivo. Collectively, the results suggest that this adenosine inhibitor is phosphorylated to an active (triphosphate) form which functions as a chain terminator for viral RNA synthesis.


Assuntos
Adenosina/farmacologia , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , RNA Viral/genética , Adenosina/química , Adenosina/metabolismo , Adenosina Quinase/genética , Adenosina Quinase/metabolismo , Animais , Antivirais/química , Antivirais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Masculino , Fosforilação , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Ratos , Ratos Wistar
11.
PLoS Negl Trop Dis ; 4(4): e672, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20436920

RESUMO

The spread of dengue (DEN) worldwide combined with an increased severity of the DEN-associated clinical outcomes have made this mosquito-borne virus of great global public health importance. Progress in understanding DEN pathogenesis and in developing effective treatments has been hampered by the lack of a suitable small animal model. Most of the DEN clinical isolates and cell culture-passaged DEN virus strains reported so far require either host adaptation, inoculation with a high dose and/or intravenous administration to elicit a virulent phenotype in mice which results, at best, in a productive infection with no, few, or irrelevant disease manifestations, and with mice dying within few days at the peak of viremia. Here we describe a non-mouse-adapted DEN2 virus strain (D2Y98P) that is highly infectious in AG129 mice (lacking interferon-alpha/beta and -gamma receptors) upon intraperitoneal administration. Infection with a high dose of D2Y98P induced cytokine storm, massive organ damage, and severe vascular leakage, leading to haemorrhage and rapid death of the animals at the peak of viremia. In contrast, very interestingly and uniquely, infection with a low dose of D2Y98P led to asymptomatic viral dissemination and replication in relevant organs, followed by non-paralytic death of the animals few days after virus clearance, similar to the disease kinetic in humans. Spleen damage, liver dysfunction and increased vascular permeability, but no haemorrhage, were observed in moribund animals, suggesting intact vascular integrity, a cardinal feature in DEN shock syndrome. Infection with D2Y98P thus offers the opportunity to further decipher some of the aspects of dengue pathogenesis and provides a new platform for drug and vaccine testing.


Assuntos
Vírus da Dengue/patogenicidade , Modelos Animais de Doenças , Dengue Grave/patologia , Dengue Grave/virologia , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Citocinas/metabolismo , Morte , Hemorragia/patologia , Humanos , Camundongos , Camundongos Knockout , Receptores de Interferon/deficiência
12.
Antiviral Res ; 85(3): 450-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20060421

RESUMO

Antiviral drug discovery is becoming increasingly important due to the global threat of viral disease pandemics. Many members of the genus Flavivirus are significant human pathogens, among which dengue virus (DENV) alone poses a public health threat to 2.5 billion worldwide, leading to 50-100 million human infections each year. Neither vaccine nor effective therapeutics is currently available for DENV. Development of a DENV vaccine has been challenging, because of the need to simultaneously immunize and induce a long-lasting protection against all four serotypes of DENV; an incompletely immunized individual may be sensitized to life-threatening dengue hemorrhagic fever or dengue shock syndrome. The challenges associated with vaccine development have underscored the importance of development of antiviral therapies for DENV and other flaviviruses. Here we review the strategies to identify inhibitors for DENV therapy. Both viral and host proteins essential for viral replication cycle are potential targets for antiviral development. Inhibitors could be identified by multiple approaches, including enzyme-based screening, viral replication-based screening, structure-based rational design, virtual screening, and fragment-based screening. The strategies discussed in this report should be applicable to antiviral development of other viruses.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Descoberta de Drogas/métodos , Humanos
13.
Proc Natl Acad Sci U S A ; 106(48): 20435-9, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19918064

RESUMO

Dengue virus (DENV), a mosquito-borne flavivirus, is a major public health threat. The virus poses risk to 2.5 billion people worldwide and causes 50 to 100 million human infections each year. Neither a vaccine nor an antiviral therapy is currently available for prevention and treatment of DENV infection. Here, we report a previously undescribed adenosine analog, NITD008, that potently inhibits DENV both in vitro and in vivo. In addition to the 4 serotypes of DENV, NITD008 inhibits other flaviviruses, including West Nile virus, yellow fever virus, and Powassan virus. The compound also suppresses hepatitis C virus, but it does not inhibit nonflaviviruses, such as Western equine encephalitis virus and vesicular stomatitis virus. A triphosphate form of NITD008 directly inhibits the RNA-dependent RNA polymerase activity of DENV, indicating that the compound functions as a chain terminator during viral RNA synthesis. NITD008 has good in vivo pharmacokinetic properties and is biologically available through oral administration. Treatment of DENV-infected mice with NITD008 suppressed peak viremia, reduced cytokine elevation, and completely prevented the infected mice from death. No observed adverse effect level (NOAEL) was achieved when rats were orally dosed with NITD008 at 50 mg/kg daily for 1 week. However, NOAEL could not be accomplished when rats and dogs were dosed daily for 2 weeks. Nevertheless, our results have proved the concept that a nucleoside inhibitor could be developed for potential treatment of flavivirus infections.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/metabolismo , Dengue/tratamento farmacológico , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Viremia/tratamento farmacológico , Adenosina/química , Animais , Antivirais/farmacocinética , Antivirais/uso terapêutico , Chlorocebus aethiops , Cães , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Camundongos , Estrutura Molecular , Nível de Efeito Adverso não Observado , Ratos , Células Vero
14.
Antiviral Res ; 84(3): 260-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19800368

RESUMO

The dengue virus envelope protein plays an essential role in viral entry by mediating fusion between the viral and host membranes. The crystal structure of the envelope protein shows a pocket (located at a "hinge" between Domains I and II) that can be occupied by ligand n-octyl-beta-D-glucoside (betaOG). Compounds blocking the betaOG pocket are thought to interfere with conformational changes in the envelope protein that are essential for fusion. Two fusion assays were developed to examine the anti-fusion activities of compounds. The first assay measures the cellular internalization of propidium iodide upon membrane fusion. The second assay measures the protease activity of trypsin upon fusion between dengue virions and trypsin-containing liposomes. We performed an in silico virtual screening for small molecules that can potentially bind to the betaOG pocket and tested these candidate molecules in the two fusion assays. We identified one compound that inhibits dengue fusion in both assays with an IC(50) of 6.8 microM and reduces viral titers with an EC(50) of 9.8 microM. Time-of-addition experiments showed that the compound was only active when present during viral infection but not when added 1h later, in agreement with a mechanism of action through fusion inhibition.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Aedes , Animais , Linhagem Celular , Cricetinae , Vírus da Dengue/química , Vírus da Dengue/fisiologia , Testes de Sensibilidade Microbiana , Ligação Proteica/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/química
15.
Eur J Immunol ; 39(10): 2809-21, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19637226

RESUMO

Monocytes and macrophages are target cells for dengue infection. Besides their potential role for virus replication, activated monocytes/macrophages produce cytokines that may be critical for dengue pathology. To study the in vivo role of monocytes and macrophages for virus replication, we depleted monocytes and macrophages in IFN-alphabetagammaR knockout mice with clodronate liposomes before dengue infection. Although less virus was first recovered in the draining LN in the absence of macrophages, monocyte/macrophage depletion eventually resulted in a ten-fold higher systemic viral titer. A massive infiltration of CD11b(+)CD11c(low)Ly6C(low) monocytes into infected organs was observed in parallel with increasing virus titers before viremia was controlled. Depletion of monocytes in the blood before or after local infection had no impact on virus titers, suggesting that monocytes are not required as "virus-shuttles". Our data provide evidence that systemic viremia is established independently of tissue macrophages present at the site of infection and blood monocytes. Instead, we demonstrate the importance of monocytes/macrophages for the control of dengue virus.


Assuntos
Vírus da Dengue/crescimento & desenvolvimento , Dengue/imunologia , Dengue/virologia , Macrófagos/citologia , Macrófagos/imunologia , Animais , Contagem de Células , Movimento Celular/imunologia , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Células Epidérmicas , Epiderme/virologia , Granulócitos/citologia , Células de Langerhans/virologia , Lipossomos , Linfa/virologia , Linfonodos/citologia , Linfonodos/virologia , Tecido Linfoide/citologia , Tecido Linfoide/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/virologia , Peritônio/citologia , Receptor de Interferon alfa e beta/genética , Receptores de Interferon/genética , Baço/citologia , Baço/virologia , Carga Viral , Proteínas não Estruturais Virais/metabolismo , Viremia , Receptor de Interferon gama
16.
J Gen Virol ; 90(Pt 4): 799-809, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19264660

RESUMO

The flavivirus envelope glycoprotein (E) is responsible for viral attachment and entry by membrane fusion. Its ectodomain is the primary target of the humoral immune response. In particular, the C-terminal Ig-like domain III of E, which is exposed at the surface of the viral particle, forms an attractive antigen for raising protective monoclonal antibodies (mAb). 9F12, a mouse mAb raised against a dengue virus (DENV) serotype 2 recombinant domain III, cross-reacts with corresponding domains from the other three DENV serotypes and also with West Nile virus. mAb 9F12 binds with nanomolar affinity to a conserved epitope that maps to the viral surface comprising residues 305, 307, 310 and 330 of the E protein. mAb 9F12 neutralizes all four DENV serotypes in plaque reduction assays. We expressed a single-chain Fv from 9F12 that retains the binding activity of the parent mAb. Adsorption and fusion inhibition assays indicate that mAb 9F12 prevents early steps of viral entry. Its virus inhibition activity and broad cross-reactivity makes mAb 9F12 a suitable candidate for optimization and humanization into a therapeutic antibody to treat severe infections by dengue.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Vírus da Dengue/classificação , Vírus da Dengue/imunologia , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Reações Cruzadas , Dengue/imunologia , Vírus da Dengue/genética , Mapeamento de Epitopos , Fusão de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Testes de Neutralização , Sorotipagem , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
17.
Antimicrob Agents Chemother ; 53(5): 1823-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19223625

RESUMO

The incidence of dengue fever epidemics has increased dramatically over the last few decades. However, no vaccine or antiviral therapies are available. Therefore, the need for safe and effective antiviral drugs has become imperative. The entry of dengue virus into a host cell is mediated by its major envelope (E) protein. The crystal structure of the E protein reveals a hydrophobic pocket that is presumably important for low-pH-mediated membrane fusion. High-throughput docking with this hydrophobic pocket was performed, and hits were evaluated in cell-based assays. Compound 6 was identified as one of the inhibitors and had an average 50% effective concentration of 119 nM against dengue virus serotype 2 in a human cell line. Mechanism-of-action studies demonstrated that compound 6 acts at an early stage during dengue virus infection. It arrests dengue virus in vesicles that colocalize with endocytosed dextran and inhibits NS3 expression. The inhibitors described in this report can serve as molecular probes for the study of the entry of flavivirus into host cells.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/patogenicidade , Bibliotecas de Moléculas Pequenas , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/química , Sítios de Ligação , Linhagem Celular , Cricetinae , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/crescimento & desenvolvimento , Humanos , Modelos Moleculares , Relação Estrutura-Atividade , Proteínas do Envelope Viral/antagonistas & inibidores
18.
J Infect Dis ; 195(5): 665-74, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17262707

RESUMO

Dengue fever is an emerging arboviral disease for which no vaccine or antiviral treatment exists and that causes thousands of fatalities each year. To develop an in vivo test system for antidengue drugs, AG129 mice, which are deficient for the interferon- alpha / beta and - gamma receptors, were injected with unadapted dengue virus, resulting in a dose-dependent transient viremia lasting several days and peaking on day 3 after infection. Additionally, nonstructural protein 1, increased levels of proinflammatory cytokines, and neutralizing IgM and IgG antibodies were found, and mice had splenomegaly. Oral administration of the antiviral compounds 7-deaza-2'-C-methyl-adenosine, N-nonyl-deoxynojirimycin, or 6-O-butanoyl castanospermine significantly reduced viremia in a dose-dependent manner, even after delayed treatment, leading to a reduction of splenomegaly and proinflammatory cytokine levels. The results validate this dengue viremia mouse model as a suitable system for testing antidengue drugs and indicate that antiviral treatment during the acute phase of dengue fever can reduce the severity of the disease.


Assuntos
Antivirais/uso terapêutico , Dengue/sangue , Dengue/tratamento farmacológico , Modelos Animais de Doenças , Viremia , Replicação Viral/efeitos dos fármacos , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , Animais , Antivirais/farmacologia , Dengue/imunologia , Relação Dose-Resposta a Droga , Indolizinas/farmacologia , Camundongos , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Fatores de Tempo , Tubercidina/análogos & derivados , Tubercidina/uso terapêutico , Viremia/tratamento farmacológico , Viremia/imunologia
19.
Mol Cell Biol ; 26(22): 8515-26, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16966369

RESUMO

Cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts (6-4PPs) comprise major UV-induced photolesions. If left unrepaired, these lesions can induce mutations and skin cancer, which is facilitated by UV-induced immunosuppression. Yet the contribution of lesion and cell type specificity to the harmful biological effects of UV exposure remains currently unclear. Using a series of photolyase-transgenic mice to ubiquitously remove either CPDs or 6-4PPs from all cells in the mouse skin or selectively from basal keratinocytes, we show that the majority of UV-induced acute effects to require the presence of CPDs in basal keratinocytes in the mouse skin. At the fundamental level of gene expression, CPDs induce the expression of genes associated with repair and recombinational processing of DNA damage, as well as apoptosis and a response to stress. At the organismal level, photolyase-mediated removal of CPDs, but not 6-4PPs, from the genome of only basal keratinocytes substantially diminishes the incidence of skin tumors; however, it does not affect the UVB-mediated immunosuppression. Taken together, these findings reveal a differential role of basal keratinocytes in these processes, providing novel insights into the skin's acute and chronic responses to UV in a lesion- and cell-type-specific manner.


Assuntos
Carcinoma/etiologia , Desoxirribodipirimidina Fotoliase/metabolismo , Terapia de Imunossupressão/métodos , Queratinócitos/efeitos da radiação , Neoplasias Cutâneas/etiologia , Pele/patologia , Animais , Apoptose , Carcinoma/genética , Carcinoma/prevenção & controle , Carnitina/análogos & derivados , Carnitina/genética , Carnitina/fisiologia , Análise por Conglomerados , Desoxirribodipirimidina Fotoliase/genética , Hiperplasia/etiologia , Queratina-14/genética , Queratinócitos/patologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Dímeros de Pirimidina , Tolerância a Radiação , Pele/efeitos da radiação , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/prevenção & controle , Transcrição Gênica , Raios Ultravioleta
20.
Curr Biol ; 15(2): 105-15, 2005 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-15668165

RESUMO

BACKGROUND: The high and steadily increasing incidence of ultraviolet-B (UV-B)-induced skin cancer is a problem recognized worldwide. UV introduces different types of damage into the DNA, notably cyclobutane pyrimidine dimers (CPDs) and (6-4) photoproducts (6-4PPs). If unrepaired, these photolesions can give rise to cell death, mutation induction, and onset of carcinogenic events, but the relative contribution of CPDs and 6-4PPs to these biological consequences of UV exposure is hardly known. Because placental mammals have undergone an evolutionary loss of photolyases, repair enzymes that directly split CPDs and 6-4PPs into the respective monomers in a light-dependent and lesion-specific manner, they can only repair UV-induced DNA damage by the elaborate nucleotide excision repair pathway. RESULTS: To assess the relative contribution of CPDs and 6-4PPs to the detrimental effects of UV light, we generated transgenic mice that ubiquitously express CPD-photolyase, 6-4PP-photolyase, or both, thereby allowing rapid light-dependent repair of CPDs and/or 6-4PPs in the skin. We show that the vast majority of (semi)acute responses in the UV-exposed skin (i.e., sunburn, apoptosis, hyperplasia, and mutation induction) can be ascribed to CPDs. Moreover, CPD-photolyase mice, in contrast to 6-4PP-photolyase mice, exhibit superior resistance to sunlight-induced tumorigenesis. CONCLUSIONS: Our data unequivocally identify CPDs as the principal cause of nonmelanoma skin cancer and provide genetic evidence that CPD-photolyase enzymes can be employed as effective tools to combat skin cancer.


Assuntos
Carcinoma/etiologia , Dano ao DNA/efeitos da radiação , Reparo do DNA/genética , Desoxirribodipirimidina Fotoliase/metabolismo , Dímeros de Pirimidina/metabolismo , Neoplasias Cutâneas/etiologia , Raios Ultravioleta , Animais , Apoptose/efeitos da radiação , Carcinoma/prevenção & controle , Linhagem Celular , Desoxirribodipirimidina Fotoliase/genética , Expressão Gênica , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...