Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836604

RESUMEN

The COVID-19 pandemic has reemphasized the need to identify safe and scalable therapeutics to slow or reverse symptoms of disease caused by newly emerging and reemerging viral pathogens. Recent clinical successes of monoclonal antibodies (mAbs) in therapy for viral infections demonstrate that mAbs offer a solution for these emerging biothreats. We have explored this with respect to Junin virus (JUNV), an arenavirus classified as a category A high-priority agent and the causative agent of Argentine hemorrhagic fever (AHF). There are currently no Food and Drug Administration-approved drugs available for preventing or treating AHF, although immune plasma from convalescent patients is used routinely to treat active infections. However, immune plasma is severely limited in quantity, highly variable in quality, and poses significant safety risks including the transmission of transfusion-borne diseases. mAbs offer a highly specific and consistently potent alternative to immune plasma that can be manufactured at large scale. We previously described a chimeric mAb, cJ199, that provided protection in a guinea pig model of AHF. To adapt this mAb to a format more suitable for clinical use, we humanized the mAb (hu199) and evaluated it in a cynomolgus monkey model of AHF with two JUNV isolates, Romero and Espindola. While untreated control animals experienced 100% lethality, all animals treated with hu199 at 6 d postinoculation (dpi) survived, and 50% of animals treated at 8 dpi survived. mAbs like hu199 may offer a safer, scalable, and more reproducible alternative to immune plasma for rare viral diseases that have epidemic potential.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Antivirales/farmacología , Fiebre Hemorrágica Americana/prevención & control , Virus Junin/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Cobayas , Fiebre Hemorrágica Americana/sangre , Humanos , Macaca fascicularis
2.
Cell Rep ; 15(7): 1514-1526, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27160900

RESUMEN

Previous efforts to identify cross-neutralizing antibodies to the receptor-binding site (RBS) of ebolavirus glycoproteins have been unsuccessful, largely because the RBS is occluded on the viral surface. We report a monoclonal antibody (FVM04) that targets a uniquely exposed epitope within the RBS; cross-neutralizes Ebola (EBOV), Sudan (SUDV), and, to a lesser extent, Bundibugyo viruses; and shows protection against EBOV and SUDV in mice and guinea pigs. The antibody cocktail ZMapp™ is remarkably effective against EBOV (Zaire) but does not cross-neutralize other ebolaviruses. By replacing one of the ZMapp™ components with FVM04, we retained the anti-EBOV efficacy while extending the breadth of protection to SUDV, thereby generating a cross-protective antibody cocktail. In addition, we report several mutations at the base of the ebolavirus glycoprotein that enhance the binding of FVM04 and other cross-reactive antibodies. These findings have important implications for pan-ebolavirus vaccine development and defining broadly protective antibody cocktails.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Ebolavirus/fisiología , Epítopos/inmunología , Glicoproteínas/metabolismo , Fiebre Hemorrágica Ebola/inmunología , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/ultraestructura , Anticuerpos Neutralizantes , Anticuerpos Antivirales/química , Sitios de Unión , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/ultraestructura , Cobayas , Células HEK293 , Humanos , Cinética , Ratones Endogámicos BALB C , Modelos Moleculares , Mutación/genética , Coloración Negativa , Pruebas de Neutralización , Resultado del Tratamiento
3.
Proc Natl Acad Sci U S A ; 113(16): 4458-63, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044104

RESUMEN

Countermeasures against potential biothreat agents remain important to US Homeland Security, and many of these pharmaceuticals could have dual use in the improvement of global public health. Junin virus, the causative agent of Argentine hemorrhagic fever (AHF), is an arenavirus identified as a category A high-priority agent. There are no Food and Drug Administration (FDA) approved drugs available for preventing or treating AHF, and the current treatment option is limited to administration of immune plasma. Whereas immune plasma demonstrates the feasibility of passive immunotherapy, it is limited in quantity, variable in quality, and poses safety risks such as transmission of transfusion-borne diseases. In an effort to develop a monoclonal antibody (mAb)-based alternative to plasma, three previously described neutralizing murine mAbs were expressed as mouse-human chimeric antibodies and evaluated in the guinea pig model of AHF. These mAbs provided 100% protection against lethal challenge when administered 2 d after infection (dpi), and one of them (J199) was capable of providing 100% protection when treatment was initiated 6 dpi and 92% protection when initiated 7 dpi. The efficacy of J199 is superior to that previously described for all other evaluated drugs, and its high potency suggests that mAbs like J199 offer an economical alternative to immune plasma and an effective dual use (bioterrorism/public health) therapeutic.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Fiebre Hemorrágica Americana/tratamiento farmacológico , Fiebre Hemorrágica Americana/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Cobayas , Humanos , Virus Junin , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología
4.
Viruses ; 8(3): 71, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-27072420

RESUMEN

Iminosugars that are competitive inhibitors of endoplasmic reticulum (ER) α-glucosidases have been demonstrated to have antiviral activity against a diverse set of viruses. A novel iminosugar, UV-4B, has recently been shown to provide protection against lethal infections with dengue and influenza A (H1N1) viruses in mice. In the current study, the breadth of activity of UV-4B against influenza was examined ex vivo and in vivo. Efficacy of UV-4B against influenza A and B viruses was shown in primary human bronchial epithelial cells, a principal target tissue for influenza. Efficacy of UV-4B against influenza A (H1N1 and H3N2 subtypes) and influenza B was demonstrated using multiple lethal mouse models with readouts including mortality and weight loss. Clinical trials are ongoing to demonstrate safety of UV-4B and future studies to evaluate antiviral activity against influenza in humans are planned.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Antivirales/administración & dosificación , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , Animales , Antivirales/farmacología , Peso Corporal , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/virología , Humanos , Ratones , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Análisis de Supervivencia , Resultado del Tratamiento
5.
Antiviral Res ; 129: 93-98, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26946111

RESUMEN

The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10-20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Inhibidores de Glicósido Hidrolasas/farmacología , Dengue Grave/tratamiento farmacológico , alfa-Glucosidasas/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Animales , Anticuerpos Antivirales/sangre , Acrecentamiento Dependiente de Anticuerpo/efectos de los fármacos , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Células Cultivadas , Chlorocebus aethiops , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Drogas en Investigación , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/enzimología , Inhibidores de Glicósido Hidrolasas/administración & dosificación , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Humanos , Concentración 50 Inhibidora , Ratones , Monocitos/virología , Receptores de Interferón/deficiencia , Serogrupo , Dengue Grave/virología , Células Vero
6.
J Immunol Methods ; 430: 33-42, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26826278

RESUMEN

An electrochemiluminescent (ECL)-based multiplex immunoassay using Meso-Scale Discovery (MSD) technology was developed for detecting antibody response toward 10 Staphylococcus aureus (S. aureus) exotoxins. These 10 antigens included three different groups of toxins: 1) single component pore-forming toxins such as alpha- and delta-hemolysins, 2) the bicomponent pore-forming toxin Panton-Valentine leukocidin (PVL), comprised of LukS-PV and LukF-PV subunits, and 3) enterotoxin/superantigens - Staphylococcal enterotoxins A (SEA), B (SEB), C1 (SEC1), D (SED), K (SEK) and Toxic shock syndrome toxin-1 (TSST-1). Assay development included optimization steps with a conventional SEB ELISA-based serological assay and then optimized parameters were transferred and re-optimized in a singleplex ECL format. Finally, two pentaplex solid-phase ECL formats were developed. As proof of concept, one set of pentaplex ECL data was compared with conventional ELISA results. During the assay development controls were screened and developed for both the singleplex and multiplex assays. ECL-based multiplex assays were more sensitive with a wide dynamic range and proved more time-efficient than conventional ELISAs. Using the newly developed ECL method we showed, for the first time, that delta-hemolysin toxin can induce an immune response as antibody titers could be detected.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Toxinas Bacterianas/inmunología , Inmunoensayo/métodos , Inmunoglobulina G/sangre , Mediciones Luminiscentes/métodos , Staphylococcus aureus/inmunología , Proteínas Bacterianas/inmunología , Enterotoxinas/inmunología , Exotoxinas/inmunología , Proteínas Hemolisinas/inmunología , Humanos , Leucocidinas/inmunología , Superantígenos/inmunología
7.
J Virol ; 90(1): 279-91, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468532

RESUMEN

UNLABELLED: Filoviruses cause highly lethal viral hemorrhagic fever in humans and nonhuman primates. Current immunotherapeutic options for filoviruses are mostly specific to Ebola virus (EBOV), although other members of Filoviridae such as Sudan virus (SUDV), Bundibugyo virus (BDBV), and Marburg virus (MARV) have also caused sizeable human outbreaks. Here we report a set of pan-ebolavirus and pan-filovirus monoclonal antibodies (MAbs) derived from cynomolgus macaques immunized repeatedly with a mixture of engineered glycoproteins (GPs) and virus-like particles (VLPs) for three different filovirus species. The antibodies recognize novel neutralizing and nonneutralizing epitopes on the filovirus glycoprotein, including conserved conformational epitopes within the core regions of the GP1 subunit and a novel linear epitope within the glycan cap. We further report the first filovirus antibody binding to a highly conserved epitope within the fusion loop of ebolavirus and marburgvirus species. One of the antibodies binding to the core GP1 region of all ebolavirus species and with lower affinity to MARV GP cross neutralized both SUDV and EBOV, the most divergent ebolavirus species. In a mouse model of EBOV infection, this antibody provided 100% protection when administered in two doses and partial, but significant, protection when given once at the peak of viremia 3 days postinfection. Furthermore, we describe novel cocktails of antibodies with enhanced protective efficacy compared to individual MAbs. In summary, the present work describes multiple novel, cross-reactive filovirus epitopes and innovative combination concepts that challenge the current therapeutic models. IMPORTANCE: Filoviruses are among the most deadly human pathogens. The 2014-2015 outbreak of Ebola virus disease (EVD) led to more than 27,000 cases and 11,000 fatalities. While there are five species of Ebolavirus and several strains of marburgvirus, the current immunotherapeutics primarily target Ebola virus. Since the nature of future outbreaks cannot be predicted, there is an urgent need for therapeutics with broad protective efficacy against multiple filoviruses. Here we describe a set of monoclonal antibodies cross-reactive with multiple filovirus species. These antibodies target novel conserved epitopes within the envelope glycoprotein and exhibit protective efficacy in mice. We further present novel concepts for combination of cross-reactive antibodies against multiple epitopes that show enhanced efficacy compared to monotherapy and provide complete protection in mice. These findings set the stage for further evaluation of these antibodies in nonhuman primates and development of effective pan-filovirus immunotherapeutics for use in future outbreaks.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Filoviridae/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Proteínas Virales/inmunología , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/aislamiento & purificación , Anticuerpos Antivirales/uso terapéutico , Reacciones Cruzadas , Modelos Animales de Enfermedad , Femenino , Inmunización Pasiva , Macaca , Ratones Endogámicos BALB C , Análisis de Supervivencia , Resultado del Tratamiento
8.
Viruses ; 7(5): 2404-27, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25984714

RESUMEN

Iminosugars are capable of targeting the life cycles of multiple viruses by blocking host endoplasmic reticulum α-glucosidase enzymes that are required for competent replication of a variety of enveloped, glycosylated viruses. Iminosugars as a class are approved for use in humans with diseases such as diabetes and Gaucher's disease, providing evidence for safety of this class of compounds. The in vitro antiviral activity of iminosugars has been described in several publications with a subset of these demonstrating in vivo activity against flaviviruses, herpesviruses, retroviruses and filoviruses. Although there is compelling non-clinical in vivo evidence of antiviral efficacy, the efficacy of iminosugars as antivirals has yet to be demonstrated in humans. In the current study, we report a novel iminosugar, UV-12, which has efficacy against dengue and influenza in mouse models. UV-12 exhibits drug-like properties including oral bioavailability and good safety profile in mice and guinea pigs. UV-12 is an example of an iminosugar with activity against multiple virus families that should be investigated in further safety and efficacy studies and demonstrates potential value of this drug class as antiviral therapeutics.


Asunto(s)
Antivirales/uso terapéutico , Dengue/tratamiento farmacológico , Iminoazúcares/uso terapéutico , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Cobayas , Iminoazúcares/farmacología , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Orthomyxoviridae/efectos de los fármacos , Resultado del Tratamiento
9.
Viruses ; 6(9): 3663-82, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25256396

RESUMEN

Sequence determination of complete or coding-complete genomes of viruses is becoming common practice for supporting the work of epidemiologists, ecologists, virologists, and taxonomists. Sequencing duration and costs are rapidly decreasing, sequencing hardware is under modification for use by non-experts, and software is constantly being improved to simplify sequence data management and analysis. Thus, analysis of virus disease outbreaks on the molecular level is now feasible, including characterization of the evolution of individual virus populations in single patients over time. The increasing accumulation of sequencing data creates a management problem for the curators of commonly used sequence databases and an entry retrieval problem for end users. Therefore, utilizing the data to their fullest potential will require setting nomenclature and annotation standards for virus isolates and associated genomic sequences. The National Center for Biotechnology Information's (NCBI's) RefSeq is a non-redundant, curated database for reference (or type) nucleotide sequence records that supplies source data to numerous other databases. Building on recently proposed templates for filovirus variant naming [ ()////-], we report consensus decisions from a majority of past and currently active filovirus experts on the eight filovirus type variants and isolates to be represented in RefSeq, their final designations, and their associated sequences.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Filoviridae/genética , Evolución Molecular , Filoviridae/clasificación , Humanos , Selección Genética
10.
Arch Virol ; 159(5): 1229-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24190508

RESUMEN

Specific alterations (mutations, deletions, insertions) of virus genomes are crucial for the functional characterization of their regulatory elements and their expression products, as well as a prerequisite for the creation of attenuated viruses that could serve as vaccine candidates. Virus genome tailoring can be performed either by using traditionally cloned genomes as starting materials, followed by site-directed mutagenesis, or by de novo synthesis of modified virus genomes or parts thereof. A systematic nomenclature for such recombinant viruses is necessary to set them apart from wild-type and laboratory-adapted viruses, and to improve communication and collaborations among researchers who may want to use recombinant viruses or create novel viruses based on them. A large group of filovirus experts has recently proposed nomenclatures for natural and laboratory animal-adapted filoviruses that aim to simplify the retrieval of sequence data from electronic databases. Here, this work is extended to include nomenclature for filoviruses obtained in the laboratory via reverse genetics systems. The previously developed template for natural filovirus genetic variant naming, (/)///-, is retained, but we propose to adapt the type of information added to each field for cDNA clone-derived filoviruses. For instance, the full-length designation of an Ebola virus Kikwit variant rescued from a plasmid developed at the US Centers for Disease Control and Prevention could be akin to "Ebola virus H.sapiens-rec/COD/1995/Kikwit-abc1" (with the suffix "rec" identifying the recombinant nature of the virus and "abc1" being a placeholder for any meaningful isolate designator). Such a full-length designation should be used in databases and the methods section of publications. Shortened designations (such as "EBOV H.sap/COD/95/Kik-abc1") and abbreviations (such as "EBOV/Kik-abc1") could be used in the remainder of the text, depending on how critical it is to convey information contained in the full-length name. "EBOV" would suffice if only one EBOV strain/variant/isolate is addressed.


Asunto(s)
Filoviridae/clasificación , Filoviridae/genética , Virus Reordenados/clasificación , Virus Reordenados/genética , Genoma Viral
11.
Virology ; 441(2): 135-45, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23582637

RESUMEN

The Ebola virus (EBOV) RNA-dependent RNA polymerase (RdRp) complex consists of the catalytic subunit of the polymerase, L, and its cofactor VP35. Using immunofluorescence analysis and coimmunoprecipitation assays, we mapped the VP35 binding site on L. A core binding domain spanning amino acids 280-370 of L was sufficient to mediate weak interaction with VP35, while the entire N-terminus up to amino acid 380 was required for strong VP35-L binding. Interestingly, the VP35 binding site overlaps with an N-terminal L homo-oligomerization domain in a non-competitive manner. N-terminal L deletion mutants containing the VP35 binding site were able to efficiently block EBOV replication and transcription in a minigenome system suggesting the VP35 binding site on L as a potential target for the development of antivirals.


Asunto(s)
Ebolavirus/fisiología , Nucleoproteínas/metabolismo , Mapeo de Interacción de Proteínas , Multimerización de Proteína , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Animales , Línea Celular , Humanos , Inmunoprecipitación , Microscopía Confocal , Microscopía Fluorescente , Proteínas de la Nucleocápside , Dominios y Motivos de Interacción de Proteínas
14.
J Pharm Sci ; 100(12): 5156-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21858822

RESUMEN

The filoviruses, Ebola virus and Marburg virus, cause severe hemorrhagic fever with up to 90% human mortality. Virus-like particles of EBOV (eVLPs) and MARV (mVLPs) are attractive vaccine candidates. For the development of stable vaccines, the conformational stability of these two enveloped VLPs produced in insect cells was characterized by various spectroscopic techniques over a wide pH and temperature range. Temperature-induced aggregation of the VLPs at various pH values was monitored by light scattering. Temperature/pH empirical phase diagrams (EPDs) of the two VLPs were constructed to summarize the large volume of data generated. The EPDs show that both VLPs lose their conformational integrity above about 50°C-60°C, depending on solution pH. The VLPs were maximally thermal stable in solution at pH 7-8, with a significant reduction in stability at pH 5 and 6. They were much less stable in solution at pH 3-4 due to increased susceptibility of the VLPs to aggregation. The characterization data and conformational stability profiles from these studies provide a basis for selection of optimized solution conditions for further vaccine formulation and long-term stability studies of eVLPs and mVLPs.


Asunto(s)
Ebolavirus/metabolismo , Enfermedad del Virus de Marburg/metabolismo , Marburgvirus/metabolismo , Virión/química , Animales , Anticuerpos Antivirales , Baculoviridae/genética , Baculoviridae/metabolismo , Línea Celular , Vacunas contra el Virus del Ébola/análisis , Vacunas contra el Virus del Ébola/química , Vacunas contra el Virus del Ébola/metabolismo , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/inmunología , Vectores Genéticos , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Insectos , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/química , Marburgvirus/genética , Marburgvirus/inmunología , Conformación Molecular , Temperatura , Vacunas de Partículas Similares a Virus/análisis , Vacunas de Partículas Similares a Virus/química , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/metabolismo
15.
Mol Cell Proteomics ; 9(12): 2690-703, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20702783

RESUMEN

An assessment of the total protein composition of filovirus (ebolavirus and marburgvirus) virions is currently lacking. In this study, liquid chromatography-linked tandem mass spectrometry of purified ebola and marburg virions was performed to identify associated cellular proteins. Host proteins involved in cell adhesion, cytoskeleton, cell signaling, intracellular trafficking, membrane organization, and chaperones were identified. Significant overlap exists between this data set and proteomic studies of disparate viruses, including HIV-1 and influenza A, generated in multiple cell types. However, the great majority of proteins identified here have not been previously described to be incorporated within filovirus particles. Host proteins identified by liquid chromatography-linked tandem mass spectrometry could lack biological relevance because they represent protein contaminants in the virus preparation, or because they are incorporated within virions by chance. These issues were addressed using siRNA library-mediated gene knockdown (targeting each identified virion-associated host protein), followed by filovirus infection. Knockdown of several host proteins (e.g. HSPA5 and RPL18) significantly interfered with ebolavirus and marburgvirus infection, suggesting specific and relevant virion incorporation. Notably, select siRNAs inhibited ebolavirus, but enhanced marburgvirus infection, suggesting important differences between the two viruses. The proteomic analysis presented here contributes to a greater understanding of filovirus biology and potentially identifies host factors that can be targeted for antiviral drug development.


Asunto(s)
Filoviridae/metabolismo , Proteómica , Interferencia de ARN , Proteínas Virales/metabolismo , Virión/metabolismo , Secuencia de Aminoácidos , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Técnicas de Silenciamiento del Gen , Microscopía Fluorescente , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Espectrometría de Masas en Tándem , Proteínas Virales/química , Proteínas Virales/genética
16.
Antimicrob Agents Chemother ; 54(5): 2152-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20211898

RESUMEN

There exists an urgent need to develop licensed drugs and vaccines for the treatment or prevention of filovirus infections. FGI-103 is a low-molecular-weight compound that was discovered through an in vitro screening assay utilizing a variant of Zaire ebolavirus (ZEBOV) that expresses green fluorescent protein. In vitro analyses demonstrated that FGI-103 also exhibits antiviral activity against wild-type ZEBOV and Sudan ebolavirus, as well as Marburgvirus (MARV) strains Ci67 and Ravn. In vivo administration of FGI-103 as a single intraperitoneal dose of 10 mg/kg delivered 24 h after infection is sufficient to completely protect mice against a lethal challenge with a mouse-adapted strain of either ZEBOV or MARV-Ravn. In a murine model of ZEBOV infection, delivery of FGI-103 reduces viremia and the viral burden in kidney, liver, and spleen tissues and is associated with subdued and delayed proinflammatory cytokine responses and tissue pathology. Taken together, these results identify a promising antiviral therapeutic candidate for the treatment of filovirus infections.


Asunto(s)
Amidinas/farmacología , Antivirales/farmacología , Benzofuranos/farmacología , Infecciones por Filoviridae/tratamiento farmacológico , Filoviridae/efectos de los fármacos , Amidinas/química , Animales , Antivirales/química , Benzofuranos/química , Chlorocebus aethiops , Citocinas/metabolismo , Ebolavirus/efectos de los fármacos , Ebolavirus/genética , Femenino , Filoviridae/genética , Infecciones por Filoviridae/inmunología , Infecciones por Filoviridae/patología , Proteínas Fluorescentes Verdes/genética , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Células Hep G2 , Humanos , Riñón/citología , Hígado/patología , Hígado/virología , Masculino , Enfermedad del Virus de Marburg/tratamiento farmacológico , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/patología , Marburgvirus/efectos de los fármacos , Marburgvirus/genética , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Bibliotecas de Moléculas Pequeñas , Células Vero
17.
Antiviral Res ; 83(3): 245-51, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19523489

RESUMEN

We report herein the identification of a small molecule therapeutic, FGI-106, which displays potent and broad-spectrum inhibition of lethal viral hemorrhagic fevers pathogens, including Ebola, Rift Valley and Dengue Fever viruses, in cell-based assays. Using mouse models of Ebola virus, we further demonstrate that FGI-106 can protect animals from an otherwise lethal infection when used either in a prophylactic or therapeutic setting. A single treatment, administered 1 day after infection, is sufficient to protect animals from lethal Ebola virus challenge. Cell-based assays also identified inhibitory activity against divergent virus families, which supports a hypothesis that FGI-106 interferes with a common pathway utilized by different viruses. These findings suggest FGI-106 may provide an opportunity for targeting viral diseases.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Ebolavirus/efectos de los fármacos , Animales , Línea Celular Tumoral , Supervivencia Celular , Quimioprevención , Virus del Dengue/efectos de los fármacos , Femenino , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Ensayo de Placa Viral
18.
J Virol ; 83(9): 4508-19, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225002

RESUMEN

We have previously shown that the first transcription start signal (TSS) of Zaire Ebola virus (ZEBOV) is involved in formation of an RNA secondary structure regulating VP30-dependent transcription activation. Interestingly, transcription of Marburg virus (MARV) minigenomes occurs independently of VP30. In this study, we analyzed the structure of the MARV 3' noncoding region and its influence on VP30 necessity. Secondary structure formation of the TSS of the first gene was experimentally determined and showed substantial differences from the structure formed by the ZEBOV TSS. Chimeric MARV minigenomes mimicking the ZEBOV-specific RNA secondary structure were neither transcribed nor replicated. Mapping of the MARV genomic replication promoter revealed that the region homologous to the sequence involved in formation of the regulatory ZEBOV RNA structure is part of the MARV promoter. The MARV promoter is contained within the first 70 nucleotides of the genome and consists of two elements separated by a spacer region, comprising the TSS of the first gene. Mutations within the spacer abolished transcription activity and led to increased replication, indicating competitive transcription and replication initiation. The second promoter element is located within the nontranslated region of the first gene and consists of a stretch of three UN(5) hexamers. Recombinant full-length MARV clones, in which the three conserved U residues were substituted, could not be rescued, underlining the importance of the UN(5) hexamers for replication activity. Our data suggest that differences in the structure of the genomic replication promoters might account for the different transcription strategies of Marburg and Ebola viruses.


Asunto(s)
Ebolavirus/química , Ebolavirus/metabolismo , Marburgvirus/química , Marburgvirus/metabolismo , Conformación de Ácido Nucleico , ARN no Traducido/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Ebolavirus/genética , Regulación Viral de la Expresión Génica , Genoma Viral/genética , Humanos , Marburgvirus/genética , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Alineación de Secuencia , Transcripción Genética/genética , Activación Transcripcional , Replicación Viral
19.
Virology ; 370(1): 33-44, 2008 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17904180

RESUMEN

In this study we report the development and optimization of two minigenome rescue systems for Nipah virus, a member of the Paramyxoviridae family. One is mediated by the T7 RNA polymerase supplied either by a constitutively expressing cell line or by transfection of expression plasmids and is thus independent from infection with a helper virus. The other approach is based on RNA polymerase I-driven transcription, a unique approach for paramyxovirus reverse genetics technology. Minigenome rescue was evaluated by reporter gene activities of (i) the two different minigenome transcription systems, (ii) genomic versus antigenomic-oriented minigenomes, (iii) different ratios of the viral protein expression plasmids, and (iv) time course experiments. The high efficiency and reliability of the established systems allowed for downscaling to 96-well plates. This served as a basis for the development of a high-throughput screening system for potential antivirals that target replication and transcription of Nipah virus without the need of high bio-containment. Using this system we were able to identify two compounds that reduced minigenome activity.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , Genoma Viral/genética , Virus Nipah/fisiología , Plásmidos/genética , ARN Polimerasa I/metabolismo , ARN Viral/metabolismo , Proteínas Virales/metabolismo , Antivirales/farmacología , Línea Celular , Cloranfenicol O-Acetiltransferasa/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Virus Nipah/efectos de los fármacos , Virus Nipah/genética , Paramyxoviridae/genética , Paramyxoviridae/metabolismo , ARN Polimerasa I/genética , ARN Viral/efectos de los fármacos , ARN Viral/genética , Transcripción Genética/efectos de los fármacos , Transfección , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
20.
Antimicrob Agents Chemother ; 50(3): 984-93, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16495261

RESUMEN

Phosphorodiamidate morpholino oligomers (PMO) are a class of uncharged single-stranded DNA analogs modified such that each subunit includes a phosphorodiamidate linkage and morpholine ring. PMO antisense agents have been reported to effectively interfere with the replication of several positive-strand RNA viruses in cell culture. The filoviruses, Marburg virus and Ebola virus (EBOV), are negative-strand RNA viruses that cause up to 90% lethality in human outbreaks. There is currently no commercially available vaccine or efficacious therapeutic for any filovirus. In this study, PMO conjugated to arginine-rich cell-penetrating peptide (P-PMO) and nonconjugated PMO were assayed for the ability to inhibit EBOV infection in cell culture and in a mouse model of lethal EBOV infection. A 22-mer P-PMO designed to base pair with the translation start site region of EBOV VP35 positive-sense RNA generated sequence-specific and time- and dose-dependent inhibition of EBOV amplification in cell culture. The same oligomer provided complete protection to mice when administered before or after an otherwise lethal infection of EBOV. A corresponding nonconjugated PMO, as well as nonconjugated truncated versions of 16 and 19 base residues, provided length-dependent protection to mice when administered prophylactically. Together, these data suggest that antisense PMO and P-PMO have the potential to control EBOV infection and are promising therapeutic candidates.


Asunto(s)
Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/prevención & control , Morfolinas/farmacología , Proteínas Virales/farmacología , Animales , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Ebolavirus/genética , Femenino , Fiebre Hemorrágica Ebola/inmunología , Cinética , Ratones , Ratones Endogámicos C57BL , Péptidos/química , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero , Proteínas Reguladoras y Accesorias Virales , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA