Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(3): 113788, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38461415

RESUMEN

Histone deacetylases (HDACs) regulate gene expression and innate immunity. Previously, we showed that HDAC5 is degraded during Vaccinia virus (VACV) infection and is a restriction factor for VACV and herpes simplex virus type 1. Here, we report that HDAC5 promotes interferon regulatory factor 3 (IRF3) activation downstream of Toll-IL-1 receptor (TIR) domain-containing adaptor molecule-1 or Sendai virus-mediated stimulation without requiring HDAC activity. Loss of HDAC5-mediated IRF3 activation is restored by re-introduction of HDAC5 but not HDAC1 or HDAC4. The antiviral activity of HDAC5 is antagonized by VACV protein C6 and orthologs from the orthopoxviruses cowpox, rabbitpox, camelpox, monkeypox, and variola. Infection by many of these viruses induces proteasomal degradation of HDAC5, and expression of C6 alone can induce HDAC5 degradation. Mechanistically, C6 binds to the dimerization domain of HDAC5 and prevents homodimerization and heterodimerization with HDAC4. Overall, this study describes HDAC5 as a positive regulator of IRF3 activation and provides mechanistic insight into how the poxviral protein C6 binds to HDAC5 to antagonize its function.


Asunto(s)
Orthopoxvirus , Virus de la Viruela , Monkeypox virus/metabolismo , Virus de la Viruela/metabolismo , Orthopoxvirus/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Virus Vaccinia/fisiología , Histona Desacetilasas/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166218, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34311080

RESUMEN

Throughout history, pandemics of infectious diseases caused by emerging viruses have spread worldwide. Evidence from previous outbreaks demonstrated that pregnant women are at high risk of contracting the diseases and suffering from adverse outcomes. However, while some viruses can cause major health complications for the mother and her fetus, others do not appear to affect pregnancy. Viral surface proteins bind to specific receptors on the cellular membrane of host cells and begin therewith the infection process. During pregnancy, the molecular features of these proteins may determine specific target cells in the placenta, which may explain the different outcomes. In this review, we display information on Variola, Influenza, Zika and Corona viruses focused on their surface proteins, effects on pregnancy, and possible target placental cells. This will contribute to understanding viral entry during pregnancy, as well as to develop strategies to decrease the incidence of obstetrical problems in current and future infections.


Asunto(s)
Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , Proteínas del Envoltorio Viral/metabolismo , Virosis/virología , Femenino , Humanos , Placenta/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Virus de la Viruela/metabolismo , Virus de la Viruela/patogenicidad , Virosis/metabolismo , Virus Zika/metabolismo , Virus Zika/patogenicidad
3.
Virology ; 501: 107-114, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27898336

RESUMEN

Most poxviruses encode a homolog of a ~200,000-kDa membrane protein originally identified in variola virus. We investigated the importance of the ectromelia virus (ECTV) homolog C15 in a natural infection model. In cultured mouse cells, the replication of a mutant virus with stop codons near the N-terminus (ECTV-C15Stop) was indistinguishable from a control virus (ECTV-C15Rev). However, for a range of doses injected into the footpads of BALB/c mice there was less mortality with the mutant. Similar virus loads were present at the site of infection with mutant or control virus whereas there was less ECTV-C15Stop in popliteal and inguinal lymph nodes, spleen and liver indicating decreased virus spread and replication. The latter results were supported by immunohistochemical analyses. Decreased spread was evidently due to immune modulatory activity of C15, rather than to an intrinsic viral function, as the survival of infected mice depended on CD4+ and CD8+ T cells.


Asunto(s)
Virus de la Ectromelia/metabolismo , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/metabolismo , Ectromelia Infecciosa/virología , Proteínas de la Membrana/metabolismo , Proteínas Virales/metabolismo , Animales , Modelos Animales de Enfermedad , Virus de la Ectromelia/genética , Ectromelia Infecciosa/genética , Ectromelia Infecciosa/patología , Femenino , Humanos , Hígado/patología , Hígado/virología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Bazo/patología , Bazo/virología , Virus de la Viruela/genética , Virus de la Viruela/metabolismo , Proteínas Virales/genética , Virulencia
4.
BioDrugs ; 30(1): 9-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26820996

RESUMEN

An unusually high production of cytokines or chemokines as well as increased complement activation can drive development of chronic inflammatory autoimmune diseases. State-of-the-art biological therapies, recombinant receptors, or specific antibodies that target immune and inflammatory mediators are now effectively used. However, these newer drugs are not equally effective for all patients and can cause adverse effects, making the search for new immunomodulatory proteins of great importance. The poxviruses--first and foremost, the variola (smallpox) virus, which is highly pathogenic in man--code for numerous highly evolved and extraordinarily effective immunomodulatory proteins that bind cytokines, chemokines, and proteins of the complement system. The discovery of and investigation into immune modulators from the variola virus has great potential for guiding new and effective drugs for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Proteínas Virales/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Quimiocinas/inmunología , Citocinas/inmunología , Diseño de Fármacos , Humanos , Factores Inmunológicos/inmunología , Poxviridae/metabolismo , Virus de la Viruela/metabolismo
5.
Cell Death Dis ; 6: e1680, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25766319

RESUMEN

Subversion of host cell apoptosis is an important survival strategy for viruses to ensure their own proliferation and survival. Certain viruses express proteins homologous in sequence, structure and function to mammalian pro-survival B-cell lymphoma 2 (Bcl-2) proteins, which prevent rapid clearance of infected host cells. In vaccinia virus (VV), the virulence factor F1L was shown to be a potent inhibitor of apoptosis that functions primarily be engaging pro-apoptotic Bim. Variola virus (VAR), the causative agent of smallpox, harbors a homolog of F1L of unknown function. We show that VAR F1L is a potent inhibitor of apoptosis, and unlike all other characterized anti-apoptotic Bcl-2 family members lacks affinity for the Bim Bcl-2 homology 3 (BH3) domain. Instead, VAR F1L engages Bid BH3 as well as Bak and Bax BH3 domains. Unlike its VV homolog, variola F1L only protects against Bax-mediated apoptosis in cellular assays. Crystal structures of variola F1L bound to Bid and Bak BH3 domains reveal that variola F1L forms a domain-swapped Bcl-2 fold, which accommodates Bid and Bak BH3 in the canonical Bcl-2-binding groove, in a manner similar to VV F1L. Despite the observed conservation of structure and sequence, variola F1L inhibits apoptosis using a startlingly different mechanism compared with its VV counterpart. Our results suggest that unlike during VV infection, Bim neutralization may not be required during VAR infection. As molecular determinants for the human-specific tropism of VAR remain essentially unknown, identification of a different mechanism of action and utilization of host factors used by a VAR virulence factor compared with its VV homolog suggest that studying VAR directly may be essential to understand its unique tropism.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Virales/metabolismo , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Línea Celular , Humanos , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Virus de la Viruela/metabolismo , Proteínas Virales/genética
6.
Bull Exp Biol Med ; 157(2): 249-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24952494

RESUMEN

The biological characteristics of a 17-kDa protein synthesized in bacterial cells, a TNF-binding domain (VARV-TNF-BP) of a 47-kDa variola virus CrmB protein (VARV-CrmB) consisting of TNF-binding and chemokine-binding domains, were studied. Removal of the C-terminal chemokine-binding domain from VARV-CrmB protein was inessential for the efficiency of its inhibition of TNF cytotoxicity towards L929 mouse fibroblast culture and for TNF-induced oxidative metabolic activity of mouse blood leukocytes. The results of this study could form the basis for further studies of VARV-TNF-BP mechanisms of activity for prospective use in practical medicine.


Asunto(s)
Factor de Necrosis Tumoral alfa/metabolismo , Virus de la Viruela/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Línea Celular , Ratones , Oxidación-Reducción/efectos de los fármacos , Unión Proteica , Factor de Necrosis Tumoral alfa/toxicidad
7.
PLoS Pathog ; 8(1): e1002475, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22241999

RESUMEN

Type 1 interferons (T1-IFNs) play a major role in antiviral defense, but when or how they protect during infections that spread through the lympho-hematogenous route is not known. Orthopoxviruses, including those that produce smallpox and mousepox, spread lympho-hematogenously. They also encode a decoy receptor for T1-IFN, the T1-IFN binding protein (T1-IFNbp), which is essential for virulence. We demonstrate that during mousepox, T1-IFNs protect the liver locally rather than systemically, and that the T1-IFNbp attaches to uninfected cells surrounding infected foci in the liver and the spleen to impair their ability to receive T1-IFN signaling, thus facilitating virus spread. Remarkably, this process can be reversed and mousepox cured late in infection by treating with antibodies that block the biological function of the T1-IFNbp. Thus, our findings provide insights on how T1-IFNs function and are evaded during a viral infection in vivo, and unveil a novel mechanism for antibody-mediated antiviral therapy.


Asunto(s)
Anticuerpos Antivirales/farmacología , Virus de la Ectromelia/metabolismo , Ectromelia Infecciosa/inmunología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Factores de Virulencia/antagonistas & inhibidores , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Cricetinae , Virus de la Ectromelia/inmunología , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/tratamiento farmacológico , Ectromelia Infecciosa/metabolismo , Femenino , Hígado/inmunología , Hígado/metabolismo , Hígado/virología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Virus de la Viruela/inmunología , Virus de la Viruela/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo , Acoplamiento Viral/efectos de los fármacos
8.
PLoS Pathog ; 7(7): e1002162, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21829356

RESUMEN

Pathogens have evolved sophisticated mechanisms to evade detection and destruction by the host immune system. Large DNA viruses encode homologues of chemokines and their receptors, as well as chemokine-binding proteins (CKBPs) to modulate the chemokine network in host response. The SECRET domain (smallpox virus-encoded chemokine receptor) represents a new family of viral CKBPs that binds a subset of chemokines from different classes to inhibit their activities, either independently or fused with viral tumor necrosis factor receptors (vTNFRs). Here we present the crystal structures of the SECRET domain of vTNFR CrmD encoded by ectromelia virus and its complex with chemokine CX3CL1. The SECRET domain adopts a ß-sandwich fold and utilizes its ß-sheet I surface to interact with CX3CL1, representing a new chemokine-binding manner of viral CKBPs. Structure-based mutagenesis and biochemical analysis identified important basic residues in the 40s loop of CX3CL1 for the interaction. Mutation of corresponding acidic residues in the SECRET domain also affected the binding for other chemokines, indicating that the SECRET domain binds different chemokines in a similar manner. We further showed that heparin inhibited the binding of CX3CL1 by the SECRET domain and the SECRET domain inhibited RAW264.7 cell migration induced by CX3CL1. These results together shed light on the structural basis for the SECRET domain to inhibit chemokine activities by interfering with both chemokine-GAG and chemokine-receptor interactions.


Asunto(s)
Quimiocina CX3CL1/química , Receptores del Factor de Necrosis Tumoral/química , Virus de la Viruela/química , Proteínas Virales/química , Sustitución de Aminoácidos , Animales , Línea Celular , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Relación Estructura-Actividad , Virus de la Viruela/genética , Virus de la Viruela/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
9.
Protein Cell ; 1(12): 1084-92, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21213103

RESUMEN

Vaccinia virus complement control protein (VCP) is one of the proteins encoded by vaccinia virus to modulate the host inflammatory response. VCP modulates the inflammatory response and protects viral habitat by inhibiting the classical and the alternative pathways of complement activation. The extended structure of VCP, mobility between its sequential domains, charge distribution and type of residues at the binding regions are factors that have been identified to influence its ability to bind to complement proteins. We report that a Lister strain of vaccinia virus encodes a VCP homolog (Lis VCP) that is functional, glycosylated, has two amino acids less than the well-characterized VCP from vaccinia virus WR strain (WR VCP), and the human smallpox inhibitor of complement enzymes (SPICE) from variola virus. The glycosylated VCP of Lister is immunogenic in contrast to the weak immunogenicity of the nonglycosylated VCP. Lis VCP is the only orthopoxviral VCP homolog found to be glycosylated, and we speculate that glycosylation influences its pattern of complement inhibition. We also correlate dimerization of VCP observed only in mammalian and baculovirus expression systems to higher levels of activity than monomers, observed in the yeast expression system.


Asunto(s)
Proteínas Recombinantes/metabolismo , Virus Vaccinia/inmunología , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Dimerización , Expresión Génica , Glicosilación , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Viruela/inmunología , Viruela/metabolismo , Relación Estructura-Actividad , Virus Vaccinia/química , Virus de la Viruela/química , Virus de la Viruela/inmunología , Virus de la Viruela/metabolismo , Proteínas Virales/genética , Proteínas Virales/farmacología
10.
Mol Biol (Mosk) ; 44(6): 1054-63, 2010.
Artículo en Ruso | MEDLINE | ID: mdl-21290827

RESUMEN

Orthopoxviruses bear in their genomes several genes coding for homologous secreted proteins able to bind tumor necrosis factor. Different species of the genus possess different sets of these tumor necrosis factor-binding proteins. Viriola virus encodes the only one of them named CrmB. Despite sharing high sequence identity, CrmB proteins belonging to distinct orthopoxviral species were shown to significantly differ by their physico-chemical and biological properties. We modeled spatial structures of tumor necrosis factor receptor domains of variola and cowpox virus CrmB proteins bound to either murine, or human or mutated human tumor necrosis factor. In the sequence of last the arginine residue at position 31 is substituted with glutamine that is characteristic for murine tumor necrosis factor. Theoretical analysis of modeled ligand-receptor complexes revealed that the least stable should be the complex of cowpox virus CrmB with human tumor necrosis factor, and that arginine to glutamine substitution at position 31 should significantly stabilize binding of corresponding human tumor necrosis factor mutant to cowpox virus CrmB. Experimental evaluation of recombinant variola and cowpox virus CrmB efficiencies in inhibiting cytotoxic effect of all these tumor necrosis factors have approved our predictions.


Asunto(s)
Virus de la Viruela Vacuna/metabolismo , Modelos Moleculares , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Arginina/genética , Virus de la Viruela Vacuna/genética , Glutamina/genética , Humanos , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/química , Virus de la Viruela/genética , Proteínas Virales/química , Proteínas Virales/genética
11.
FASEB J ; 24(5): 1479-88, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20019241

RESUMEN

Variola virus (VARV) caused smallpox, one of the most devastating human diseases and the first to be eradicated, but its deliberate release represents a dangerous threat. Virulent orthopoxviruses infecting humans, such as monkeypox virus (MPXV), could fill the niche left by smallpox eradication and the cessation of vaccination. However, immunomodulatory activities and virulence determinants of VARV and MPXV remain largely unexplored. We report the molecular characterization of the VARV- and MPXV-secreted type I interferon-binding proteins, which interact with the cell surface after secretion and prevent type I interferon responses. The proteins expressed in the baculovirus system have been purified, and their interferon-binding properties characterized by surface plasmon resonance. The ability of these proteins to inhibit a broad range of interferons was investigated to identify potential adaptation to the human immune system. Furthermore, we demonstrate by Western blot and activity assays the expression of the type I interferon inhibitor during VARV and MPXV infections. These findings are relevant for the design of new vaccines and therapeutics to smallpox and emergent virulent orthopoxviruses because the type I interferon-binding protein is a major virulence factor in animal models, vaccination with this protein induces protective immunity, and its neutralization prevents disease progression.


Asunto(s)
Interferón Tipo I/antagonistas & inhibidores , Monkeypox virus/inmunología , Mpox/inmunología , Viruela/inmunología , Virus de la Viruela/inmunología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunomodulación , Monkeypox virus/metabolismo , Monkeypox virus/patogenicidad , Virus de la Viruela/metabolismo , Virus de la Viruela/patogenicidad , Vacunas Virales/inmunología
12.
J Interferon Cytokine Res ; 30(3): 123-34, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20038204

RESUMEN

The type III interferon (IFN) family elicits an antiviral response that is nearly identical to that evoked by IFN-alpha/beta. However, these cytokines (known as IFN-lambda1, 2, and 3) signal through a distinct receptor, and thus may be resistant to the evasion strategies used by some viruses to avoid the IFN-alpha/beta response. Orthopoxviruses are highly resistant to IFN-alpha/beta because they encode well-characterized immunomodulatory proteins that inhibit IFN activity. These include a secreted receptor (B18R) that neutralizes IFN-alpha/beta, and a cytoplasmic protein (E3L) that blocks IFN-alpha/beta effector functions in infected cells. We therefore determined the ability of these immunomodulators to abrogate the IFN-lambda-induced antiviral response. We found that (i) vaccinia virus (VACV) replication is resistant to IFN-lambda antiviral activity; (ii) neither VACV B18R nor the variola virus homolog B20R neutralizes IFN-lambda; (iii) VACV E3L inhibits the IFN-lambda-mediated antiviral response through a PKR-dependent pathway; (iv) VACV infection inhibits IFN-lambdaR-mediated signal transduction and gene expression. These results demonstrate differential sensitivity of IFN-lambda to multiple distinct evasion mechanisms employed by a single virus.


Asunto(s)
Regulación de la Expresión Génica , Factores Inmunológicos/metabolismo , Interferones/antagonistas & inhibidores , Receptor de Interferón alfa y beta/metabolismo , Virus Vaccinia/fisiología , Proteínas Virales/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Células HeLa , Humanos , Factores Inmunológicos/genética , Interferones/genética , Interferones/farmacología , Ratones , Proteínas de Unión al ARN/metabolismo , Receptor de Interferón alfa y beta/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Vaccinia/fisiopatología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Virus de la Viruela/metabolismo , Vesiculovirus/metabolismo , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
13.
Biochemistry (Mosc) ; 74(12): 1356-62, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19961417

RESUMEN

Gel-filtration chromatographic separation of the lysate of Sf21 insect cells infected with recombinant baculovirus BVi67 containing the gene for TNF-binding protein (CrmB) of variola virus (VARV) revealed that hTNF-cytotoxicity neutralization activity is associated with a fraction corresponding mainly to high molecular weight proteins (above 500 kDa) and less with fractions corresponding to proteins of 270 or 90 kDa. The recombinant VARV-CrmB protein has been purified by affinity chromatography. Difference in the experimentally determined and estimated (according to amino acid composition) VARV-CrmB molecular weight is due to glycosylation of the recombinant protein expressed in the insect cells. VARV-CrmB neutralizes in vitro the cytotoxic effect of hTNF and hLTalpha, and its TNF-neutralizing activity is two to three orders of magnitude higher compared to the analogous effects of type I and II soluble TNF receptors, comparable with the activity of mAb MAK195, and somewhat lower than the effect of the commercial drug Remicade.


Asunto(s)
Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Antiinflamatorios/farmacología , Anticuerpos Monoclonales/farmacología , Humanos , Infliximab , Linfotoxina-alfa/antagonistas & inhibidores , Linfotoxina-alfa/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación
14.
Proc Natl Acad Sci U S A ; 106(22): 9045-50, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19451633

RESUMEN

Identification of the binary interactions between viral and host proteins has become a valuable tool for investigating viral tropism and pathogenesis. Here, we present the first systematic protein interaction screening of the unique variola virus proteome by using yeast 2-hybrid screening against a variety of human cDNA libraries. Several protein-protein interactions were identified, including an interaction between variola G1R, an ankryin/F-box containing protein, and human nuclear factor kappa-B1 (NF-kappaB1)/p105. This represents the first direct interaction between a pathogen-encoded protein and NF-kappaB1/p105. Orthologs of G1R are present in a variety of pathogenic orthopoxviruses, but not in vaccinia virus, and expression of any one of these viral proteins blocks NF-kappaB signaling in human cells. Thus, proteomic screening of variola virus has the potential to uncover modulators of the human innate antiviral responses.


Asunto(s)
Interacciones Huésped-Patógeno , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Proteómica , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Biblioteca de Genes , Humanos , Subunidad p50 de NF-kappa B/metabolismo , Orthopoxvirus/metabolismo , Orthopoxvirus/patogenicidad , Técnicas del Sistema de Dos Híbridos
15.
Curr Opin Investig Drugs ; 9(8): 865-70, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18666034

RESUMEN

The smallpox virus can potentially be used as a biological weapon and pose a serious threat to human populations, thus rendering the development of measures against it particularly important. There is a lack of effective antiviral drugs for the treatment of smallpox and, as a result, a number of viral as well as host targets are being explored in preclinical and clinical studies to assess their efficacy as potential therapeutic agents. This review describes the latest approaches used to develop drugs against smallpox.


Asunto(s)
Antivirales/uso terapéutico , Sistemas de Liberación de Medicamentos , Viruela/tratamiento farmacológico , Animales , Antivirales/farmacología , Bioterrorismo , Diseño de Fármacos , Quimioterapia Combinada , Humanos , Transducción de Señal/efectos de los fármacos , Viruela/fisiopatología , Virus de la Viruela/efectos de los fármacos , Virus de la Viruela/metabolismo
16.
J Virol ; 81(18): 9891-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17581984

RESUMEN

Variola virus, the causative agent of smallpox, enters and exits the host via the respiratory route. To better understand the pathogenesis of poxvirus infection and its interaction with respiratory epithelia, we used vaccinia virus and examined its interaction with primary cultures of well-differentiated human airway epithelia. We found that vaccinia virus preferentially infected the epithelia through the basolateral membrane and released viral progeny across the apical membrane. Despite infection and virus production, epithelia retained tight junctions, transepithelial electrical conductance, and a steep transepithelial concentration gradient of virus, indicating integrity of the epithelial barrier. In fact, during the first four days of infection, epithelial height and cell number increased. These morphological changes and maintenance of epithelial integrity required vaccinia virus growth factor, which was released basolaterally, where it activated epidermal growth factor 1 receptors. These data suggest a complex interaction between the virus and differentiated airway epithelia; the virus preferentially enters the cells basolaterally, exits apically, and maintains epithelial integrity by stimulating growth factor receptors.


Asunto(s)
Mucosa Respiratoria/metabolismo , Viruela/metabolismo , Virus Vaccinia/crecimiento & desarrollo , Vaccinia/metabolismo , Internalización del Virus , Diferenciación Celular , Conductividad Eléctrica , Receptores ErbB/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Viruela/patología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Uniones Estrechas/virología , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Vaccinia/patología , Virus Vaccinia/metabolismo , Virus de la Viruela/crecimiento & desarrollo , Virus de la Viruela/metabolismo , Esparcimiento de Virus/fisiología
17.
Virology ; 358(1): 211-20, 2007 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-16979683

RESUMEN

Interleukin-18 (IL-18) plays an important role in host defense against microbial pathogens. Many poxviruses encode homologous IL-18 binding proteins (IL-18BP) that neutralize IL-18 activity. Here, we examined whether IL-18BP neutralizes IL-18 activity by binding to the same region of IL-18 where IL-18 receptor (IL-18R) binds. We introduced alanine substitutions to known receptor binding sites of human IL-18 and found that only the substitution of Leu5 reduced the binding affinity of IL-18 with IL-18BP of variola virus (varvIL-18BP) by more than 4-fold. The substitutions of Lys53 and Ser55, which were not previously known to be part of the receptor binding site but that are spatially adjacent to Leu5, reduced the binding affinity to varvIL-18BP by approximately 100- and 7-fold, respectively. These two substitutions also reduced the binding affinity with human IL-18R alpha subunit (hIL-18Ralpha) by 4- and 2-fold, respectively. Altogether, our data show that varvIL-18BP prevents IL-18 from binding to IL-18R by interacting with three residues that are part of the binding site for hIL-18Ralpha.


Asunto(s)
Interleucina-18/metabolismo , Receptores de Interleucina-18/metabolismo , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Interleucina-18/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , Resonancia por Plasmón de Superficie
18.
Biochim Biophys Acta ; 1764(11): 1710-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17070121

RESUMEN

Tumor necrosis factor (TNF), a potent proinflammatory and antiviral cytokine, is a critical extracellular immune regulator targeted by poxviruses through the activity of virus-encoded family of TNF-binding proteins (CrmB, CrmC, CrmD, and CrmE). The only TNF-binding protein from variola virus (VARV), the causative agent of smallpox, infecting exclusively humans, is CrmB. Here we have aligned the amino acid sequences of CrmB proteins from 10 VARV, 14 cowpox virus (CPXV), and 22 monkeypox virus (MPXV) strains. Sequence analyses demonstrated a high homology of these proteins. The regions homologous to cd00185 domain of the TNF receptor family, determining the specificity of ligand-receptor binding, were found in the sequences of CrmB proteins. In addition, a comparative analysis of the C-terminal SECRET domain sequences of CrmB proteins was performed. The differences in the amino acid sequences of these domains characteristic of each particular orthopoxvirus species were detected. It was assumed that the species-specific distinctions between the CrmB proteins might underlie the differences in these physicochemical and biological properties. The individual recombinant proteins VARV-CrmB, MPXV-CrmB, and CPXV-CrmB were synthesized in a baculovirus expression system in insect cells and isolated. Purified VARV-CrmB was detectable as a dimer with a molecular weight of 90 kDa, while MPXV- and CPXV-CrmBs, as monomers when fractioned by non-reducing SDS-PAGE. The CrmB proteins of VARV, MPXV, and CPXV differed in the efficiencies of inhibition of the cytotoxic effects of human, mouse, or rabbit TNFs in L929 mouse fibroblast cell line. Testing of CrmBs in the experimental model of LPS-induced shock using SPF BALB/c mice detected a pronounced protective effect of VARV-CrmB. Thus, our data demonstrated the difference in anti-TNF activities of VARV-, MPXV-, and CPXV-CrmBs and efficiency of VARV-CrmB rather than CPXV- or MPXV-CrmBs against LPS-induced mortality in mice.


Asunto(s)
Virus de la Viruela Vacuna/metabolismo , Monkeypox virus/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Ratones , Datos de Secuencia Molecular , Receptores del Factor de Necrosis Tumoral/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Spodoptera , Proteínas Virales/química
19.
Mol Biol (Mosk) ; 39(6): 1055-62, 2005.
Artículo en Ruso | MEDLINE | ID: mdl-16358743

RESUMEN

DNA fragments containing genes for coding IFN-gamma-binding proteins (IFNgammaBPs) of variola virus (VARV) and monkeypox virus (MPXV) were obtained from viral genomes using PCR. Isolated genes coding desired proteins were expressed in the insect Sf21 cells using baculovirus expression system. Secreted recombinant IFNgammaBPs were isolated from culture medium of infected Sf21 cells through affinity chromatography procedure. SDS-PAAG and Western blot analysis of culture medium of infected insect cells and preparations of purified recombinant IFNgammaBPs indicated that recombinant viral proteins were dimerized even in the absence of ligand (hIFNgamma) unlike their cell (eucaryotic) analogs. Biological activity of the recombinant IFNgammaBPs were studied in the test of protective effect inhibition of hIFNgamma on L68 cells infected with murine encephalomyocarditis virus. It was shown that recombinant IFNgammaBPs had dose-dependent IFNgamma-inhibiting activity. A possibility of the elaboration of new therapeutics for anti-hIFNgamma therapy on the base of IFNgammaBPs is discussed.


Asunto(s)
Antivirales/antagonistas & inhibidores , Interferón gamma/antagonistas & inhibidores , Monkeypox virus/metabolismo , Virus de la Viruela/metabolismo , Proteínas Virales/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Interferón gamma/farmacología , Datos de Secuencia Molecular , Monkeypox virus/genética , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Virus de la Viruela/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
J Clin Invest ; 115(2): 379-87, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15690085

RESUMEN

The EGF-like domain of smallpox growth factor (SPGF) targets human ErbB-1, inducing tyrosine phosphorylation of certain host cellular substrates via activation of the receptor's kinase domain and thereby facilitating viral replication. Given these findings, low molecular weight organic inhibitors of ErbB-1 kinases might function as antiviral agents against smallpox. Here we show that CI-1033 and related 4-anilinoquinazolines inhibit SPGF-induced human cellular DNA synthesis, protein tyrosine kinase activation, and c-Cbl association with ErbB-1 and resultant internalization. Infection of monkey kidney BSC-40 and VERO-E6 cells in vitro by variola strain Solaimen is blocked by CI-1033, primarily at the level of secondary viral spreading. In an in vivo lethal vaccinia virus pneumonia model, CI-1033 alone promotes survival of animals, augments systemic T cell immunity and, in conjunction with a single dose of anti-L1R intracellular mature virus particle-specific mAb, fosters virtually complete viral clearance of the lungs of infected mice by the eighth day after infection. Collectively, these findings show that chemical inhibitors of host-signaling pathways exploited by viral pathogens may represent potent antiviral therapies.


Asunto(s)
Receptores ErbB/metabolismo , Sustancias de Crecimiento/metabolismo , Morfolinas/farmacología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Viruela/tratamiento farmacológico , Virus de la Viruela/metabolismo , Proteínas Virales/metabolismo , Animales , Chlorocebus aethiops , ADN/biosíntesis , Receptores ErbB/antagonistas & inhibidores , Células HeLa , Humanos , Masculino , Ratones , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología , Neumonía/virología , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-cbl , Viruela/metabolismo , Viruela/patología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Vaccinia/tratamiento farmacológico , Vaccinia/inmunología , Vaccinia/patología , Virus Vaccinia/metabolismo , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...