Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 19458, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34593911

RESUMO

Efficacious therapeutics for Ebola virus disease are in great demand. Ebola virus infections mediated by mucosal exposure, and aerosolization in particular, present a novel challenge due to nontypical massive early infection of respiratory lymphoid tissues. We performed a randomized and blinded study to compare outcomes from vehicle-treated and remdesivir-treated rhesus monkeys in a lethal model of infection resulting from aerosolized Ebola virus exposure. Remdesivir treatment initiated 4 days after exposure was associated with a significant survival benefit, significant reduction in serum viral titer, and improvements in clinical pathology biomarker levels and lung histology compared to vehicle treatment. These observations indicate that remdesivir may have value in countering aerosol-induced Ebola virus disease.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/farmacologia , Administração Intravenosa , Aerossóis , Alanina/administração & dosagem , Alanina/farmacologia , Animais , Antivirais/administração & dosagem , Modelos Animais de Doenças , Feminino , Doença pelo Vírus Ebola/sangue , Estimativa de Kaplan-Meier , Fígado/efeitos dos fármacos , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Linfonodos/virologia , Macaca mulatta , Masculino , Distribuição Aleatória , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Síndrome de Resposta Inflamatória Sistêmica/virologia , Carga Viral/efeitos dos fármacos , Viremia/tratamento farmacológico
2.
Appl Biosaf ; 26(1): 23-32, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36033961

RESUMO

Introduction: Failure of an existing effluent decontamination system (EDS) prompted the consideration of commercial off-the-shelf solutions for decontamination of containment laboratory waste. A bleach-based chemical EDS was purchased to serve as an interim solution. Methods: Studies were conducted in the laboratory to validate inactivation of Bacillus spores with bleach in complex matrices containing organic simulants including fetal bovine serum, humic acid, and animal room sanitation effluent. Results: These studies demonstrated effective decontamination of >106 spores at a free chlorine concentration of ≥5700 parts per million with a 2-hour contact time. Translation of these results to biological validation of the bleach-based chemical EDS required some modifications to the system and its operation. Discussion: The chemical EDS was validated for the treatment of biosafety levels 3 and 4 waste effluent using laboratory-prepared spore packets along with commercial biological indicators; however, several issues and lessons learned identified during the process of onboarding are also discussed, including bleach product source, method of validation, dechlorination, and treated waste disposal.

3.
J Infect Dis ; 222(11): 1894-1901, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-32479636

RESUMO

Marburg virus (MARV) is a filovirus with documented human case-fatality rates of up to 90%. Here, we evaluated the therapeutic efficacy of remdesivir (GS-5734) in nonhuman primates experimentally infected with MARV. Beginning 4 or 5 days post inoculation, cynomolgus macaques were treated once daily for 12 days with vehicle, 5 mg/kg remdesivir, or a 10-mg/kg loading dose followed by 5 mg/kg remdesivir. All vehicle-control animals died, whereas 83% of animals receiving a 10-mg/kg loading dose of remdesivir survived, as did 50% of animals receiving a 5-mg/kg remdesivir regimen. Remdesivir-treated animals exhibited improved clinical scores, lower plasma viral RNA, and improved markers of kidney function, liver function, and coagulopathy versus vehicle-control animals. The small molecule remdesivir showed therapeutic efficacy in this Marburg virus disease model with treatment initiation 5 days post inoculation, supporting further assessment of remdesivir for the treatment of Marburg virus disease in humans.


Assuntos
Antimetabólitos/uso terapêutico , Antivirais/uso terapêutico , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus/efeitos dos fármacos , Doenças dos Macacos/tratamento farmacológico , Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Animais , Modelos Animais de Doenças , Feminino , Estimativa de Kaplan-Meier , Macaca fascicularis , Masculino , Doença do Vírus de Marburg/mortalidade , Doença do Vírus de Marburg/patologia , Doença do Vírus de Marburg/virologia , Doenças dos Macacos/mortalidade , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , RNA Viral
4.
Viruses ; 12(1)2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31941095

RESUMO

Recent Ebola virus (EBOV) outbreaks in West Africa and the Democratic Republic of the Congo have highlighted the urgent need for approval of medical countermeasures for treatment and prevention of EBOV disease (EVD). Until recently, when successes were achieved in characterizing the efficacy of multiple experimental EVD therapeutics in humans, the only feasible way to obtain data regarding potential clinical benefits of candidate therapeutics was by conducting well-controlled animal studies. Nonclinical studies are likely to continue to be important tools for screening and development of new candidates with improved pharmacological properties. Here, we describe a natural history study to characterize the time course and order of progression of the disease manifestations of EVD in rhesus monkeys. In 12 rhesus monkeys exposed by the intramuscular route to 1000 plaque-forming units of EBOV, multiple endpoints were monitored for 28 days following exposure. The disease progressed rapidly with mortality events occurring 7-10 days after exposure. Key disease manifestations observed consistently across the infected animals included, but were not limited to, viremia, fever, systemic inflammation, coagulopathy, lymphocytolysis, renal tubular necrosis with mineralization, and hepatocellular degeneration and necrosis.


Assuntos
Modelos Animais de Doenças , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/fisiopatologia , Macaca mulatta/virologia , Animais , Progressão da Doença , Feminino , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/mortalidade , Injeções Intramusculares , Masculino
5.
Cell Host Microbe ; 24(3): 405-416.e3, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30173956

RESUMO

Sexual transmission of filoviruses was first reported in 1968 after an outbreak of Marburg virus (MARV) disease and recently caused flare-ups of Ebola virus disease in the 2013-2016 outbreak. How filoviruses establish testicular persistence and are shed in semen remain unknown. We discovered that persistent MARV infection of seminiferous tubules, an immune-privileged site that harbors sperm production, is a relatively common event in crab-eating macaques that survived infection after antiviral treatment. Persistence triggers severe testicular damage, including spermatogenic cell depletion and inflammatory cell invasion. MARV mainly persists in Sertoli cells, leading to breakdown of the blood-testis barrier formed by inter-Sertoli cell tight junctions. This disruption is accompanied by local infiltration of immunosuppressive CD4+Foxp3+ regulatory T cells. Our study elucidates cellular events associated with testicular persistence that may promote sexual transmission of filoviruses and suggests that targeting immunosuppression may be warranted to clear filovirus persistence in damaged immune-privileged sites.


Assuntos
Doença do Vírus de Marburg/virologia , Marburgvirus/fisiologia , Doenças dos Primatas/virologia , Testículo/virologia , Animais , Macaca , Masculino , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/metabolismo , Doenças dos Primatas/imunologia , Doenças dos Primatas/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/virologia , Sobreviventes , Linfócitos T Reguladores/imunologia , Junções Íntimas/metabolismo , Junções Íntimas/virologia
6.
AAPS PharmSciTech ; 19(1): 395-412, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28752471

RESUMO

Systemically administered interferons are rapidly cleared from the circulation thus requiring frequent dosing to maintain the therapeutic levels of circulating interferon. This is particularly problematic for their use in the treatment of chronic diseases. The purpose of this study was to evaluate the potential of proprietary calcium phosphate (CaP) particles to deliver biologically active interferon alpha (IFNα) via the lungs into systemic circulation. Recombinant human IFNα-2a was formulated with proprietary CaP particles. In vitro biological activity of IFNα was assessed for its potential to activate IFN-induced cellular pathways in HEK-Blu-IFN α/ß cell cultures. Antiviral activity was evaluated against vesicular stomatitis virus (VSV) infection of HeLa cells. Male BALB/c mice were used to evaluate the absorption of IFNα from CaP-IFNα across the lungs following intratracheal (IT) instillation. Serum IFNα concentrations up to 9 h post-treatment were determined. Data were analyzed to obtain pharmacokinetic (PK) parameters. Data from these studies indicated that IFNα formulated with CaP retains its biological activity, and it is transported into circulation in a dose-dependent manner. PK analysis showed larger than two-fold area under the serum concentration-time curve (AUC) for CaP-IFNα compared to non-formulated IFNα administered IT. The IFNα formulated with CaP had two-fold longer half-life (t1/2) and mean residence time (MRT) relative to IFNα alone administered by injection. Clearance of CaP-IFNα was slower than IFNα administered IM or IT. Relative bioavailability of CaP-IFNα was 1.3-fold of IFNα injection and twofold of IFNα administered IT. Furthermore, inhalation of aerosolized CaP did not indicate any lung toxicity in animals.


Assuntos
Antivirais/administração & dosagem , Fosfatos de Cálcio/química , Interferon-alfa/administração & dosagem , Pulmão/metabolismo , Administração por Inalação , Aerossóis , Animais , Antivirais/farmacocinética , Antivirais/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Células HeLa , Humanos , Interferon alfa-2 , Interferon-alfa/farmacocinética , Interferon-alfa/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia
7.
Infect Immun ; 85(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28760930

RESUMO

The obligate intracellular parasite Toxoplasma gondii can actively infect any nucleated cell type, including cells from the immune system. The rapid transfer of T. gondii from infected dendritic cells to effector natural killer (NK) cells may contribute to the parasite's sequestration and shielding from immune recognition shortly after infection. However, subversion of NK cell functions, such as cytotoxicity or production of proinflammatory cytokines, such as gamma interferon (IFN-γ), upon parasite infection might also be beneficial to the parasite. In the present study, we investigated the effects of T. gondii infection on NK cells. In vitro, infected NK cells were found to be poor at killing target cells and had reduced levels of IFN-γ production. This could be attributed in part to the inability of infected cells to form conjugates with their target cells. However, even upon NK1.1 cross-linking of NK cells, the infected NK cells also exhibited poor degranulation and IFN-γ production. Similarly, NK cells infected in vivo were also poor at killing target cells and producing IFN-γ. Increased levels of transforming growth factor ß production, as well as increased levels of expression of SHP-1 in the cytosol of infected NK cells upon infection, were observed in infected NK cells. However, the phosphorylation of STAT4 was not altered in infected NK cells, suggesting that transcriptional regulation mediates the reduced IFN-γ production, which was confirmed by quantitative PCR. These data suggest that infection of NK cells by T. gondii impairs NK cell recognition of target cells and cytokine release, two mechanisms that independently could enhance T. gondii survival.


Assuntos
Imunomodulação , Células Matadoras Naturais/microbiologia , Células Matadoras Naturais/fisiologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Interações Hospedeiro-Parasita , Interferon gama/biossíntese , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6/biossíntese , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Fator de Transcrição STAT4/metabolismo , Toxoplasma/fisiologia , Fator de Crescimento Transformador beta/biossíntese
8.
Cell Microbiol ; 18(11): 1537-1550, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27018989

RESUMO

The obligate intracellular parasite Toxoplasma gondii exploits cells of the immune system to disseminate. Upon infection, parasitized dendritic cells (DCs) and microglia exhibit a hypermigratory phenotype in vitro that has been associated with enhancing parasite dissemination in vivo in mice. One unresolved question is how parasites commandeer parasitized cells to achieve systemic dissemination by a 'Trojan-horse' mechanism. By chromatography and mass spectrometry analyses, we identified an orthologue of the 14-3-3 protein family, T. gondii 14-3-3 (Tg14-3-3), as mediator of DC hypermotility. We demonstrate that parasite-derived polypeptide fractions enriched for Tg14-3-3 or recombinant Tg14-3-3 are sufficient to induce the hypermotile phenotype when introduced by protein transfection into murine DCs, human DCs or microglia. Further, gene transfer of Tg14-3-3 by lentiviral transduction induced hypermotility in primary human DCs. In parasites expressing Tg14-3-3 in a ligand-regulatable fashion, overexpression of Tg14-3-3 was correlated with induction of hypermotility in parasitized DCs. Localization studies in infected DCs identified Tg14-3-3 within the parasitophorous vacuolar space and a rapid recruitment of host cell 14-3-3 to the parasitophorous vacuole membrane. The present work identifies a determinant role for Tg14-3-3 in the induction of the migratory activation of immune cells by T. gondii. Collectively, the findings reveal Tg14-3-3 as a novel target for an intracellular pathogen that acts by hijacking the host cell's migratory properties to disseminate.


Assuntos
Proteínas 14-3-3/fisiologia , Células Dendríticas/fisiologia , Proteínas de Protozoários/fisiologia , Toxoplasma/fisiologia , Animais , Movimento Celular , Células Cultivadas , Células Dendríticas/parasitologia , Interações Hospedeiro-Parasita , Humanos , Camundongos Endogâmicos C57BL , Vacúolos/metabolismo , Vacúolos/parasitologia
9.
Cell Microbiol ; 18(3): 413-23, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26348250

RESUMO

Plasmodium falciparum (P. falciparum)-induced effects on the phenotype of human dendritic cells (DC) could contribute to poor induction of long-lasting protective immunity against malaria. DC ability to present antigens to naïve T cells, thus initiating adaptive immune responses depends on complex switches in chemokine receptors, production of soluble mediators and expression of molecules enabling antigen-presentation and maturation. To examine the cellular basis of these processes in the context of malaria, we performed detailed analysis of early events following exposure of human monocyte-derived DC to natural hemozoin (nHZ) and the synthetic analog of its heme core, ß-hematin. DC exposed to either molecule produced high levels of the inflammatory chemokine MCP-1, showed continuous high expression of the inflammatory chemokine receptor CCR5, no upregulation of the lymphoid homing receptor CCR7 and no cytoskeletal actin redistribution with loss of podosomes. DC partially matured as indicated by increased expression of major histocompatibility complex (MHC) class II and CD86 following nHZ and ß-hematin exposure, however there was a lack in expression of the maturation marker CD83 following nHZ but not ß-hematin exposure. Overall our data demonstrate that exposure to nHZ partially impairs the capacity of DC to mature, an effect in part differential to ß-hematin.


Assuntos
Células Dendríticas/fisiologia , Hemeproteínas/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Malária Falciparum/metabolismo , Antígenos CD/metabolismo , Antígeno B7-2/metabolismo , Quimiocina CCL2/metabolismo , Células Dendríticas/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hemeproteínas/farmacologia , Humanos , Imunoglobulinas/metabolismo , Lipopolissacarídeos/farmacologia , Malária Falciparum/parasitologia , Glicoproteínas de Membrana/metabolismo , Podossomos/efeitos dos fármacos , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptores CCR7/genética , Receptores CCR7/metabolismo , Antígeno CD83
10.
Int J Parasitol ; 44(2): 85-90, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24184911

RESUMO

While the spread of Toxoplasma gondii within the infected human or animal host is associated with pathology, the pathways of dissemination have remained enigmatic. From the time point of entry into the gut, to the quiescent chronic infection in the central nervous system, Toxoplasma is detected and surveyed by immune cells that populate the tissues, for example dendritic cells. Paradoxically, this protective migratory function of leukocytes appears to be targeted by Toxoplasma to mediate its dissemination in the organism. Recent findings show that tightly regulated events take place shortly after host cell invasion that promote the migratory activation of infected dendritic cells. Here, we review the emerging knowledge on how this obligate intracellular protozoan orchestrates the subversion of leukocytes to achieve systemic dissemination and reach peripheral organs where pathology manifests.


Assuntos
Leucócitos/fisiologia , Toxoplasma/fisiologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Interações Hospedeiro-Parasita , Humanos
11.
Cell Microbiol ; 15(10): 1735-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23534541

RESUMO

Host cell manipulation is an important feature of the obligate intracellular parasite Toxoplasma gondii. Recent reports have shown that the tachyzoite stages subvert dendritic cells (DC) as a conduit for dissemination (Trojan horse) during acute infection. To examine the cellular basis of these processes, we performed a detailed analysis of the early events following tachyzoite invasion of human monocyte-derived DC. We demonstrate that within minutes after tachyzoite penetration, profound morphological changes take place in DC that coincide with a migratory activation. Active parasite invasion of DC led to cytoskeletal actin redistribution with loss of adhesive podosome structures and redistribution of integrins (CD18 and CD11c), that concurred with the onset of DC hypermotility in vitro. Inhibition of parasite rhoptry secretion and invasion, but not inhibition of parasite or host cell protein synthesis, abrogated the onset of morphological changes and hypermotility in DC dose-dependently. Also, infected DC, but not by-stander DC, exhibited upregulation of C-C chemokine receptor 7 (CCR7). Yet, the onset of parasite-induced DC hypermotility preceded chemotactic migratory responsesin vitro. Collectively, present data reveal that invasion of DC by T. gondii initiates a series of regulated events, including rapid cytoskeleton rearrangements, hypermotility and chemotaxis, that promote the migratory activation of DC.


Assuntos
Movimento Celular , Citoesqueleto/metabolismo , Células Dendríticas/fisiologia , Células Dendríticas/parasitologia , Endocitose , Interações Hospedeiro-Patógeno , Toxoplasma/fisiologia , Células Cultivadas , Quimiotaxia , Humanos
12.
PLoS Pathog ; 8(12): e1003051, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236276

RESUMO

During acute infection in human and animal hosts, the obligate intracellular protozoan Toxoplasma gondii infects a variety of cell types, including leukocytes. Poised to respond to invading pathogens, dendritic cells (DC) may also be exploited by T. gondii for spread in the infected host. Here, we report that human and mouse myeloid DC possess functional γ-aminobutyric acid (GABA) receptors and the machinery for GABA biosynthesis and secretion. Shortly after T. gondii infection (genotypes I, II and III), DC responded with enhanced GABA secretion in vitro. We demonstrate that GABA activates GABA(A) receptor-mediated currents in T. gondii-infected DC, which exhibit a hypermigratory phenotype. Inhibition of GABA synthesis, transportation or GABA(A) receptor blockade in T. gondii-infected DC resulted in impaired transmigration capacity, motility and chemotactic response to CCL19 in vitro. Moreover, exogenous GABA or supernatant from infected DC restored the migration of infected DC in vitro. In a mouse model of toxoplasmosis, adoptive transfer of infected DC pre-treated with GABAergic inhibitors reduced parasite dissemination and parasite loads in target organs, e.g. the central nervous system. Altogether, we provide evidence that GABAergic signaling modulates the migratory properties of DC and that T. gondii likely makes use of this pathway for dissemination. The findings unveil that GABA, the principal inhibitory neurotransmitter in the brain, has activation functions in the immune system that may be hijacked by intracellular pathogens.


Assuntos
Movimento Celular/imunologia , Células Dendríticas/imunologia , Transdução de Sinais/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Ácido gama-Aminobutírico/imunologia , Animais , Células Cultivadas , Quimiocina CCL19/imunologia , Células Dendríticas/parasitologia , Humanos , Camundongos , Receptores de GABA-A/imunologia , Toxoplasmose/patologia
13.
PLoS One ; 7(3): e32123, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22403627

RESUMO

The obligate intracellular parasite Neospora caninum disseminates across the placenta and the blood-brain barrier, to reach sites where it causes severe pathology or establishes chronic persistent infections. The mechanisms used by N. caninum to breach restrictive biological barriers remain elusive. To examine the cellular basis of these processes, migration of different N. caninum isolates (Nc-1, Nc-Liverpool, Nc-SweB1 and the Spanish isolates: Nc-Spain 3H, Nc-Spain 4H, Nc-Spain 6, Nc-Spain 7 and Nc-Spain 9) was studied in an in vitro model based on a placental trophoblast-derived BeWo cell line. Here, we describe that infection of dendritic cells (DC) by N. caninum tachyzoites potentiated translocation of parasites across polarized cellular monolayers. In addition, powered by the parasite's own gliding motility, extracellular N. caninum tachyzoites were able to transmigrate across cellular monolayers. Altogether, the presented data provides evidence of two putative complementary pathways utilized by N. caninum, in an isolate-specific fashion, for passage of restrictive cellular barriers. Interestingly, adoptive transfer of tachyzoite-infected DC in mice resulted in increased parasitic loads in various organs, e.g. the central nervous system, compared to infections with free parasites. Inoculation of pregnant mice with infected DC resulted in an accentuated vertical transmission to the offspring with increased parasitic loads and neonatal mortality. These findings reveal that N. caninum exploits the natural cell trafficking pathways in the host to cross cellular barriers and disseminate to deep tissues. The findings are indicative of conserved dissemination strategies among coccidian apicomplexan parasites.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/parasitologia , Espaço Intracelular/parasitologia , Neospora/fisiologia , Placenta/citologia , Placenta/parasitologia , Animais , Linhagem Celular , Polaridade Celular , Coccidiose/congênito , Coccidiose/transmissão , Espaço Extracelular/parasitologia , Feminino , Humanos , Transmissão Vertical de Doenças Infecciosas , Masculino , Camundongos , Movimento , Neospora/patogenicidade , Fenótipo , Gravidez , Especificidade da Espécie , Trofoblastos/citologia , Trofoblastos/parasitologia
14.
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
15.
J Virol ; 84(24): 12646-57, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943977

RESUMO

Tetherin and IFITM3 are recently identified interferon-induced cellular proteins that restrict infections by retroviruses and filoviruses and of influenza virus and flaviviruses, respectively. In our efforts to further explore their antiviral activities against other viruses and determine their antiviral mechanisms, we found that the two antiviral proteins potently inhibit the infection of vesicular stomatitis virus (VSV), a prototype member of the Rhabdoviridae family. Taking advantage of this well-studied virus infection system, we show that although both tetherin and IFITM3 are plasma membrane proteins, tetherin inhibits virion particle release from infected cells, while IFITM3 disrupts an early event after endocytosis of virion particles but before primary transcription of incoming viral genomes. Furthermore, we demonstrate that both the N-terminal 21 amino acid residues and C-terminal transmembrane region of IFITM3 are required for its antiviral activity. Collectively, our work sheds light on the mechanisms by which tetherin and IFITM3 restrict infection with rhabdoviruses and possibly other pathogenic viruses.


Assuntos
Antígenos CD/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Estomatite Vesicular/prevenção & controle , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vírion/fisiologia , Replicação Viral/fisiologia , Antígenos CD/genética , Northern Blotting , Western Blotting , Células Cultivadas , Endocitose , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Genoma Viral , Humanos , Proteínas de Membrana/genética , Mutação/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Estomatite Vesicular/genética , Internalização do Vírus
16.
J Virol ; 84(16): 8332-41, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534863

RESUMO

Interferons (IFNs) are key mediators of the host innate antiviral immune response. To identify IFN-stimulated genes (ISGs) that instigate an antiviral state against two medically important flaviviruses, West Nile virus (WNV) and dengue virus (DENV), we tested 36 ISGs that are commonly induced by IFN-alpha for antiviral activity against the two viruses. We discovered that five ISGs efficiently suppressed WNV and/or DENV infection when they were individually expressed in HEK293 cells. Mechanistic analyses revealed that two structurally related cell plasma membrane proteins, IFITM2 and IFITM3, disrupted early steps (entry and/or uncoating) of the viral infection. In contrast, three IFN-induced cellular enzymes, viperin, ISG20, and double-stranded-RNA-activated protein kinase, inhibited steps in viral proteins and/or RNA biosynthesis. Our results thus imply that the antiviral activity of IFN-alpha is collectively mediated by a panel of ISGs that disrupt multiple steps of the DENV and WNV life cycles.


Assuntos
Vírus da Dengue/imunologia , Interações Hospedeiro-Patógeno , Interferon-alfa/imunologia , Proteínas de Membrana/imunologia , Proteínas/imunologia , Proteínas de Ligação a RNA/imunologia , Vírus do Nilo Ocidental/imunologia , Linhagem Celular , Exonucleases/biossíntese , Exonucleases/imunologia , Exorribonucleases , Regulação da Expressão Gênica , Humanos , Proteínas de Membrana/biossíntese , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Proteínas/metabolismo , Proteínas de Ligação a RNA/biossíntese , Internalização do Vírus , Replicação Viral/imunologia , eIF-2 Quinase/biossíntese , eIF-2 Quinase/imunologia
17.
Antiviral Res ; 83(1): 53-60, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19501257

RESUMO

Type III interferons (IFN), IFN-lambda or IL-28/29, are new members of the IFN super-family. Except for using distinct receptors, type I and type III IFNs share the same major post receptor signaling components to activate the transcription of a similar set of IFN-stimulated genes (ISGs). To examine the antiviral effects of the new type IFNs against West Nile virus (WNV), we compared the antiviral effects of IFN-alpha and IFN-lambda on WNV virus-like particle (VLP) infection and replicon replication in Huh7.5 and Hela cells. The results revealed that (i) both types of IFNs could efficiently prevent the WNV infection, but IFN-alpha demonstrated a stronger antiviral efficacy; (ii) WNV genome replication in VLP-infected cells and replicon-containing cell lines could only be inhibited by IFN-alpha, but not IFN-lambda; (iii) in agreement with the observed antiviral effects, only IFN-lambda-induced activation of JAK-STAT signaling pathway and induction of ISG expression were completely inhibited in WNV replicon-containing cell lines, but IFN-alpha signal transduction was either unaffected or only partially inhibited in Huh7.5 or Hela cells by the virus. Hence, the differential inhibition of WNV on IFN-alpha and IFN-lambda signal transduction implies that the receptors of the two types of IFNs, but not the common post receptor signaling components, could be selectively targeted either directly by WNV nonstructural proteins or indirectly by the cellular responses induced by the virus infection to inhibit the signal transduction of the cytokines.


Assuntos
Antivirais/farmacologia , Interleucinas/farmacologia , Vírus do Nilo Ocidental/efeitos dos fármacos , Linhagem Celular , Humanos , Interferon-alfa/farmacologia , Interferons , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...