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










Base de datos
Intervalo de año de publicación
2.
Curr Med Chem ; 28(40): 8384-8391, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33563150

RESUMEN

Progressive globalization of our society brings not only worldwide integration, it also increases and promotes our exposure to new viral pathogens with evident impacts on our global health. Especially with the emergence of SARS-CoV-2, our biomedical research infrastructure has never been more compelled to rapidly develop antiviral regimens that demonstrate improved efficacy against these pathogens. Here, we showcase 3 poignant antivirals against the lucrative target, RNA-dependent RNA polymerase (RdRP) of RNA viruses - a timely and relevant topic given the present efforts against COVID-19. While effective drug designs against RdRP are important, their benefit and potential as a standard of care truly relies on them standing out in well-designed clinical trials.


Asunto(s)
COVID-19 , ARN Polimerasa Dependiente del ARN , Antivirales/farmacología , Antivirales/uso terapéutico , Diseño de Fármacos , Humanos , ARN Viral , SARS-CoV-2
3.
Antiviral Res ; 171: 104598, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31513822

RESUMEN

As antiviral drug resistance develops and new viruses emerge there is a pressing need to develop strategies to rapidly develop antiviral therapeutics. Here we use phospho-specific flow cytometry to assess perturbations of many different cellular signaling pathways during treatment with drug combinations that are highly effective in blocking Herpes simplex virus type 1 (HSV-1) infection. We discovered two antiviral drug combinations act on distinct signaling pathways, either STAT1 or S6 phosphorylation, to block HSV-1 infection. We focused on upregulation of S6 phosphorylation by HSV-1 infection, and our subsequent finding that ribavirin antagonizes this upregulation of S6 phosphorylation. We go on to show that the S6 kinase inhibitor SL0101 blocks HSV-1 replication in vitro and in an in vivo animal model of HSV-1 infection. Overall, we have used an unbiased analysis of cellular signaling pathways during treatment by antiviral drug combinations to discover a novel antiviral drug target against HSV-1 infection. The outcomes of the approach we present highlight the importance of analyzing how antiviral drugs modulate cellular and pathogen-induced signaling as a method to discover new drug therapy targets.


Asunto(s)
Antivirales/farmacología , Ribavirina/farmacología , Ribosomas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Biomarcadores , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Herpes Simple/tratamiento farmacológico , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Humanos , Ratones , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factor de Transcripción STAT1/metabolismo , Replicación Viral/efectos de los fármacos
4.
J Immunol Res ; 2019: 8685312, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31089479

RESUMEN

Type I Interferons were first described for their profound antiviral abilities in cell culture and animal models, and later, they were translated into potent antiviral therapeutics. However, as additional studies into the function of Type I Interferons progressed, it was also seen that pathogenic viruses have coevolved to encode potent mechanisms allowing them to evade or suppress the impact of Type I Interferons on their replication. For chronic viral infections, such as HIV and many of the AIDS-associated viruses, including HTLV, HCV, KSHV, and EBV, the clinical efficacy of Type I Interferons is limited by these mechanisms. Here, we review some of the ways that HIV and AIDS-associated viruses thrive in Type I Interferon-rich environments via mechanisms that block the function of this important antiviral cytokine. Overall, a better understanding of these mechanisms creates avenues to better understand the innate immune response to these viruses as well as plan the development of antivirals that would allow the natural antiviral effect of Type I Interferons to manifest during these infections.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Infecciones por VIH/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferón Tipo I/inmunología , Transducción de Señal , Síndrome de Inmunodeficiencia Adquirida/virología , Animales , Ensayos Clínicos como Asunto , VIH/inmunología , Humanos , Inmunidad Innata , Ratones
5.
Innate Immun ; 24(8): 490-500, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30282499

RESUMEN

This study investigates the modulation of Type I IFN induction of an antiviral state by HIV. IFNs, including IFN-α, are key innate immune cytokines that activate the JAK/STAT pathway leading to the expression of IFN-stimulated genes. IFN-stimulated gene expression establishes the antiviral state, limiting viral infection in IFN-α-stimulated microenvironments. Our previous studies have shown that HIV proteins disrupt the induction of IFN-α by degradation of IFN-ß promoter stimulator-1, an adaptor protein for the up-regulation and release of IFN-α into the local microenvironment via the retinoic acid-inducible gene 1-like receptor signaling pathway. However, IFN-α is still released from other sources such as plasmacytoid dendritic cells via TLR-dependent recognition of HIV. Here we report that the activation of the JAK/STAT pathway by IFN-α stimulation is disrupted by HIV proteins Vpu and Nef, which both reduce IFN-α induction of STAT1 phosphorylation. Thus, HIV would still be able to avoid antiviral protection induced by IFN-α in the local microenvironment. These findings show that HIV blocks multiple signaling points that would lead to the up-regulation of IFN-stimulated genes, allowing more effective replication in IFN-α-rich environments.


Asunto(s)
Infecciones por VIH/inmunología , VIH/fisiología , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Microambiente Celular , Células HEK293 , Humanos , Evasión Inmune , Inmunidad Innata , Interferón-alfa/metabolismo , Fosforilación , Transducción de Señal , Replicación Viral
6.
Cell Host Microbe ; 21(3): 334-343, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28279345

RESUMEN

New influenza vaccines that provide effective and broad protection are desperately needed. Live attenuated viruses are attractive vaccine candidates because they can elicit both humoral and cellular immune responses. However, recent formulations of live attenuated influenza vaccines (LAIVs) have not been protective. We combined high-coverage transposon mutagenesis of influenza virus with a rapid high-throughput screening for attenuation to generate W7-791, a live attenuated mutant virus strain. W7-791 produced only a transient asymptomatic infection in adult and neonatal mice even at doses 100-fold higher than the LD50 of the parent strain. A single administration of W7-791 conferred full protection to mice against lethal challenge with H1N1, H3N2, and H5N1 strains, and improved viral clearance in ferrets. Adoptive transfer of T cells from W7-791-immunized mice conferred heterologous protection, indicating a role for T cell-mediated immunity. These studies present an LAIV development strategy to rapidly generate and screen entire libraries of viral clones.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/aislamiento & purificación , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Protección Cruzada , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Hurones , Pruebas Genéticas , Inmunidad Heteróloga , Vacunas contra la Influenza/administración & dosificación , Ratones , Mutagénesis Insercional , Infecciones por Orthomyxoviridae/inmunología , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/aislamiento & purificación
7.
PLoS One ; 10(9): e0137951, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26375588

RESUMEN

Our main objective of this study was to determine how Human Immunodeficiency Virus (HIV) avoids induction of the antiviral Type I Interferon (IFN) system. To limit viral infection, the innate immune system produces important antiviral cytokines such as the IFN. IFN set up a critical roadblock to virus infection by limiting further replication of a virus. Usually, IFN production is induced by the recognition of viral nucleic acids by innate immune receptors and subsequent downstream signaling. However, the importance of IFN in the defense against viruses has lead most pathogenic viruses to evolve strategies to inhibit host IFN induction or responses allowing for increased pathogenicity and persistence of the virus. While the adaptive immune responses to HIV infection have been extensively studied, less is known about the balance between induction and inhibition of innate immune defenses, including the antiviral IFN response, by HIV infection. Here we show that HIV infection of T cells does not induce significant IFN production even IFN I Interferon production. To explain this paradox, we screened HIV proteins and found that two HIV encoded proteins, Vpu and Nef, strongly antagonize IFN induction, with expression of these proteins leading to loss of expression of the innate immune viral RNA sensing adaptor protein, IPS-1 (IFN-ß promoter stimulator-1). We hypothesize that with lower levels of IPS-1 present, infected cells are defective in mounting antiviral responses allowing HIV to replicate without the normal antiviral actions of the host IFN response. Using cell lines as well as primary human derived cells, we show that HIV targeting of IPS-1 is key to limiting IFN induction. These findings describe how HIV infection modulates IFN induction providing insight into the mechanisms by which HIV establishes infection and persistence in a host.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Replicación Viral/inmunología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Citocinas/metabolismo , Infecciones por VIH/patología , Infecciones por VIH/virología , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Humanos , Transducción de Señal
8.
J Microbiol Biol Educ ; 15(2): 232-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25574286

RESUMEN

Over the past years, professional students have had extensive exposure to clinical cases during basic science classes. With this in mind, we have taken this clinical case exposure moment to be an opportune time to introduce the ethics of working with patients during biomedical research. Our goal is to present a straightforward assignment that allows for active student research into the facts of the Tuskegee Experiment of the 1900s. The assignment provides the necessary background to allow for a student-centered discussion on the ethical issues of the events and ramifications of what happened. Thus, in educating a class on the event's happenings, one concomitantly creates a platform for meaningful discussion on the principles and ethics of patient care. We have found that an ethics-infused event such as the Tuskegee Experiment is an excellent way to introduce students to these topics.

9.
Int J Nanomedicine ; 7: 2281-92, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654513

RESUMEN

BACKGROUND: Infectious diseases cause many molecular assemblies and pathways within cellular signaling networks to function aberrantly. The most effective way to treat complex, diseased cellular networks is to apply multiple drugs that attack the problem from many fronts. However, determining the optimal combination of several drugs at specific dosages to reach an endpoint objective is a daunting task. METHODS: In this study, we applied an experimental feedback system control (FSC) method and rapidly identified optimal drug combinations that inhibit herpes simplex virus-1 infection, by only testing less than 0.1% of the total possible drug combinations. RESULTS: Using antiviral efficacy as the criterion, FSC quickly identified a highly efficacious drug cocktail. This cocktail contained high dose ribavirin. Ribavirin, while being an effective antiviral drug, often induces toxic side effects that are not desirable in a therapeutic drug combination. To screen for less toxic drug combinations, we applied a second FSC search in cascade and used both high antiviral efficacy and low toxicity as criteria. Surprisingly, the new drug combination eliminated the need for ribavirin, but still blocked viral infection in nearly 100% of cases. CONCLUSION: This cascade search provides a versatile platform for rapid discovery of new drug combinations that satisfy multiple criteria.


Asunto(s)
Antivirales/administración & dosificación , Descubrimiento de Drogas/métodos , Herpes Simple/tratamiento farmacológico , Herpesvirus Humano 1/efectos de los fármacos , Algoritmos , Animales , Antivirales/farmacología , Antivirales/toxicidad , Combinación de Medicamentos , Retroalimentación , Herpes Simple/metabolismo , Ratones , Modelos Biológicos , Células 3T3 NIH
10.
Proc Natl Acad Sci U S A ; 109(11): 4239-44, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371602

RESUMEN

Type I and type II interferons (IFNs) are cytokines that establish the cellular antiviral state through the induction of IFN-stimulated genes (ISGs). We sought to understand the basis of the antiviral activity induced by type I and II IFNs in relation to the functions of their ISGs. Based on gene expression studies, we systematically identified antiviral ISGs by performing blinded, functional screens on 288 type I and type II ISGs. We assessed and validated the antiviral activity of these ISGs against an RNA virus, vesicular stomatitis virus (VSV), and a DNA virus, murine gammaherpes virus (MHV-68). Overall, we identified 34 ISGs that elicited an antiviral effect on the replication of either one or both viruses. Fourteen ISGs have uncharacterized antiviral functions. We further defined ISGs that affect critical life-cycle processes in expression of VSV protein and MHV-68 immediate-early genes. Two previously undescribed antiviral ISGs, TAP1 and BMP2, were further validated. TAP1-deficient fibroblasts were more susceptible to VSV infection but less so to MHV-68 infection. On the other hand, exogenous BMP2 inhibits MHV-68 lytic growth but did not affect VSV growth. These results delineate common and distinct sets of type I and type II IFN-induced genes as well as identify unique ISGs that have either broad or specific antiviral effects on these viruses.


Asunto(s)
Virus ADN/inmunología , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Vesiculovirus/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Células de la Médula Ósea/citología , Virus ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Interferón Tipo I/farmacología , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Vesiculovirus/efectos de los fármacos
12.
Curr Opin Immunol ; 23(1): 57-64, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21123041

RESUMEN

Type I interferons (IFNs) are cytokines of the innate immune system that induce antiviral protein expression in response to viral infection. Various proteins and pathways have been shown to recognize nucleic acid ligands especially from RNA viruses. Here, we will review recent developments including transcription of DNA virus genomes into RNA ligands, and the recognition of viruses by TLR2 for interferon induction. The induced IFNs activate many interferon stimulated genes (ISGs) that have direct antiviral effects. Recent studies have identified IFITM proteins as the first ISG to inhibit viral entry processes and revealed mechanistic understanding of known antiviral ISGs such as ISG15 and Viperin.


Asunto(s)
Interferón Tipo I/inmunología , Virosis/inmunología , Animales , ADN Viral/genética , Humanos , Receptor Toll-Like 2/inmunología , Transcripción Genética
13.
Cell Host Microbe ; 5(2): 166-78, 2009 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-19218087

RESUMEN

A conserved herpesviral kinase, designated ORF36 in murine gamma-herpesvirus 68 (MHV-68), plays multiple vital roles in the viral life cycle. Here, we show by screening mutant viruses that ORF36 counteracts the antiviral type I interferon (IFN) response. ORF36 specifically binds to the activated form of interferon regulatory factor 3 (IRF-3) in the nucleus, inhibiting IRF-3 interaction with the cotranscriptional activator CBP and thereby suppressing the recruitment of RNA polymerase II to the interferon beta promoter. The anti-IFN function of ORF36 is conserved among herpesvirus subfamilies, although the conserved kinase activity is not absolutely required for this function. MHV-68 lacking ORF36 induces a greater interferon response and is attenuated in vitro and in vivo, where acute viral infection in the lung and latency in the spleen are compromised. Our data suggest that herpesviruses have evolved within their conserved kinase an anti-IFN activity critical for evasion of host immunity and for persistence.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Interferón Tipo I/biosíntesis , Proteínas Quinasas/metabolismo , Rhadinovirus/patogenicidad , Proteínas Virales/metabolismo , Animales , Pulmón/inmunología , Pulmón/virología , Proteínas de la Membrana/metabolismo , Ratones , Fosfoproteínas/metabolismo , Unión Proteica , Proteínas Quinasas/genética , Rhadinovirus/genética , Bazo/inmunología , Bazo/virología , Proteínas Virales/genética , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
14.
J Virol ; 82(5): 2208-17, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18077715

RESUMEN

Innate immune responses against viral infection, especially the induction of type I interferon, are critical for limiting the replication of the virus. Although it has been shown that DNA can induce type I interferon, to date no natural DNA ligand of a virus that induces type I interferon has been described. Here we screened the genome of murine gammaherpesvirus 68 with mutations at various genomic locations to map the region of DNA that induces type I interferon. A repetitive region termed the 100-base-pair repeat region is a ligand that is both necessary and sufficient for the viral genomic DNA to induce type I interferon. A region colinear with this ligand in the genome of Kaposi's sarcoma-associated herpesvirus also induces type I interferon. We have thus defined a repetitive region of the genomes of gammaherpesviruses as the first natural DNA virus ligand that induces type I interferon.


Asunto(s)
ADN Viral/genética , Gammaherpesvirinae/genética , Interferón Tipo I/biosíntesis , Animales , Secuencia de Bases , Células Cultivadas , Cromosomas Artificiales Bacterianos , Cartilla de ADN , ADN Viral/metabolismo , Humanos , Ligandos , Ratones , Mutación
15.
J Immunol ; 177(1): 298-305, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16785525

RESUMEN

Langerhans cells (LC) are a unique subset of dendritic cells (DC), present in the epidermis and serving as the first line of defense against pathogens invading the skin. To investigate the role of human LCs in innate immune responses, we examined TLR expression and function of LC-like DCs derived from CD34+ progenitor cells and compared them to DCs derived from peripheral blood monocytes (monocyte-derived DC; Mo-DC). LC-like DCs and Mo-DCs expressed TLR1-10 mRNAs at comparable levels. Although many of the TLR-induced cytokine patterns were similar between the two cell types, stimulation with the TLR3 agonist poly(I:C) triggered significantly higher amounts of the IFN-inducible chemokines CXCL9 (monokine induced by IFN-gamma) and CXCL11 (IFN-gamma-inducible T cell alpha chemoattractant) in LC-like DCs as compared with Mo-DCs. Supernatants from TLR3-activated LC-like DCs reduced intracellular replication of vesicular stomatitis virus in a type I IFN-dependent manner. Finally, CXCL9 colocalized with LCs in skin biopsy specimens from viral infections. Together, our data suggest that LCs exhibit a direct antiviral activity that is dependent on type I IFN as part of the innate immune system.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Células de Langerhans/inmunología , Células de Langerhans/virología , Receptor Toll-Like 3/fisiología , Línea Celular , Células Cultivadas , Quimiocina CXCL11 , Quimiocina CXCL9 , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/metabolismo , Células Dendríticas/metabolismo , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/virología , Humanos , Interferón Tipo I/fisiología , Queratinocitos/inmunología , Queratinocitos/metabolismo , Queratinocitos/virología , Células de Langerhans/metabolismo , Ligandos , Molusco Contagioso/inmunología , Molusco Contagioso/virología , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 3/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Verrugas/inmunología , Verrugas/virología
16.
J Clin Invest ; 115(5): 1369-78, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15864354

RESUMEN

Little is known about the role of CD1d-restricted T cells in antiviral immune responses. Here we show that the lytic replication cycle of the Kaposi sarcoma-associated herpesvirus (KSHV) promotes downregulation of cell-surface CD1d. This is caused by expression of the 2 modulator of immune recognition (MIR) proteins of the virus, each of which promotes the loss of surface CD1d expression following transfection into uninfected cells. Inhibition of CD1d surface expression is due to ubiquitination of the CD1d alpha-chain on a unique lysine residue in its cytoplasmic tail, which triggers endocytosis. Unlike MIR-mediated MHC class I downregulation, however, CD1d downregulation does not appear to include accelerated lysosomal degradation. MIR2-induced downregulation of CD1d results in reduced activation of CD1d-restricted T cells in vitro. KSHV modulation of CD1d expression represents a strategy for viral evasion of innate host immune responses and implicates CD1d-restricted T cells as regulators of this viral infection.


Asunto(s)
Antígenos CD1/genética , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , Replicación Viral/fisiología , Antígenos CD1/biosíntesis , Antígenos CD1/inmunología , Antígenos CD1d , Regulación hacia Abajo/fisiología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/inmunología , Humanos , Ubiquitina/metabolismo , Proteínas Virales/metabolismo
17.
J Biol Chem ; 277(8): 6124-30, 2002 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-11751860

RESUMEN

Kaposi's sarcoma-associated herpesvirus encodes two related proteins, MIR1 and MIR2, that lead to reduction of the cell surface levels of major histocompatibility complex class I and other polypeptides involved in immune recognition. MIR1 and MIR2 do not affect the assembly or transport of their target proteins through the secretory pathway; rather, they act to enhance the selective endocytosis of target chains from the cell surface. Sequence inspection reveals that the modulator of immune recognition (MIR) proteins contain an NH(2)-terminal zinc finger of the plant homeodomain (PHD) subfamily, two transmembrane (TM) domains, and a C-terminal conserved region (CR). Here we examine the transmembrane topology and functional organization of MIR2. Both the PHD domain and the CR are disposed cytosolically and are essential for MIR-mediated endocytosis. MIR proteins form homo-oligomers; this activity is independent of the PHD and CR elements and maps instead to the TM regions. Analysis of chimeras between MIR1 and MIR2 reveals that the TM regions also mediate target selectivity. Mutations that ablate the PHD or CR regions generate dominant negative phenotypes for major histocompatibility complex class I endocytosis. These findings suggest a domain organization for the MIR proteins, with the TM regions involved in target selection and the cytosolic PHD and CR domains involved in the possible recruitment of cellular machinery that directly or indirectly regulates internalization of target molecules.


Asunto(s)
Endocitosis/fisiología , Herpesvirus Humano 8/fisiología , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Vectores Genéticos , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Virales/química , Proteínas Virales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...