Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 16(1): 110, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29699566

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has been increasing by 0.5% per year in the United States. PDAC portends a dismal prognosis and novel therapies are needed. This study describes the generation and characterization of a novel oncolytic chimeric orthopoxvirus for the treatment of pancreatic cancer. METHODS: After chimerization and high-throughput screening, CF33 was chosen from 100 new chimeric orthopoxvirus isolates for its ability to kill pancreatic cancer cells. In vitro cytotoxicity was assayed in six pancreatic cancer cell lines. In vivo efficacy and toxicity were evaluated in PANC-1 and MIA PaCa-2 xenograft models. RESULTS: CF33 caused rapid killing of six pancreatic cancer cells lines in vitro, releasing damage-associated molecular patterns, and regression of PANC-1 injected and non-injected distant xenografts in vivo after a single low intratumoral dose of 103 plaque-forming units. Using luciferase imaging, CF33 was noted to preferentially replicate in tumors which corresponds to the low viral titers found in solid organs. CONCLUSION: The low dose of CF33 required to treat pancreatic cancer in this preclinical study may ease the manufacturing and dosing challenges currently facing oncolytic viral therapy.


Assuntos
Terapia Viral Oncolítica , Orthopoxvirus/fisiologia , Neoplasias Pancreáticas/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral , Quimera , Citotoxicidade Imunológica , Relação Dose-Resposta Imunológica , Humanos , Luciferases/metabolismo , Orthopoxvirus/isolamento & purificação , Neoplasias Pancreáticas/patologia , Replicação Viral
2.
Viruses ; 9(8)2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28786952

RESUMO

Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.


Assuntos
Apoptose , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Infecções por Poxviridae/virologia , Poxviridae/fisiologia , Animais , Caspases/metabolismo , Humanos , Leporipoxvirus/patogenicidade , Leporipoxvirus/fisiologia , Molluscipoxvirus/patogenicidade , Molluscipoxvirus/fisiologia , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/patogenicidade , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/fisiopatologia , Transdução de Sinais , Proteínas Virais/metabolismo , Virulência , Replicação Viral
3.
Methods Mol Biol ; 1581: 121-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28374246

RESUMO

Poxviruses cause many diseases in humans and animals worldwide, and there is a need for vaccines with improved safety and good efficacy. In addition, poxvirus vectors are widely used as recombinant vaccines for various infectious diseases and as recombinant and oncolytic vaccines for cancer. One concern with poxvirus vaccine vectors is that some poxviruses can infect a developing fetus and cause fetal loss or congenital disease. This can be an issue both for patients receiving a vaccine and for pregnant health care providers, including doctors, nurses, and veterinarians, who might receive accidental exposure to the poxvirus by injection or during patient care. We describe here a method for analyzing the safety of virus exposure in pregnant mammals using a mouse model testing vaccinia, canarypox, and raccoonpox virus vectors.


Assuntos
Infecções por Poxviridae/diagnóstico , Poxviridae/patogenicidade , Animais , Chlorocebus aethiops , Feminino , Camundongos , Orthopoxvirus/genética , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/fisiologia , Gravidez , Vacínia/diagnóstico , Células Vero , Carga Viral , Vacinas Virais/genética , Replicação Viral
4.
PLoS Pathog ; 11(9): e1005148, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26325270

RESUMO

Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.


Assuntos
Anticorpos Neutralizantes/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Modelos Moleculares , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Proteínas do Envelope Viral/antagonistas & inibidores , Tropismo Viral , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/uso terapêutico , Afinidade de Anticorpos , Especificidade de Anticorpos , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Complexo Antígeno-Anticorpo/metabolismo , Chlorocebus aethiops , Feminino , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Mutação , Orthopoxvirus/imunologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/prevenção & controle , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Vacinas Sintéticas/química , Vacinas Sintéticas/genética , Vacinas Sintéticas/metabolismo , Vacinas Sintéticas/uso terapêutico , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/metabolismo , Vacinas Virais/uso terapêutico
5.
Vestn Ross Akad Med Nauk ; (12): 4-8, 2013.
Artigo em Russo | MEDLINE | ID: mdl-24741936

RESUMO

UNLABELLED: Currently one of the most promising approaches in development of cancer virotherapy is based on the ability of oncolytic viruses to selective infection and lysis of tumor cells. AIM: The goal of the study was to identify and evaluate perspective oncolytic viruses capable of selectively destroying human glioma cells. PATIENTS AND METHODS: Original GB2m, GA14m and GB22m glioma cell cultures derived from patients were used for evaluating in vitro oncolytic activity of some typical orthopoxviruses, adenoviruses and parvoviruses. RESULTS: The oncolytic activity in the human glioma cell models was confirmed for LIVP and WR strains of vaccinia virus, Adel2 and Ad2del strains with deletions within E1B/55K gene and derived from human adenoviruses type 2 and 5, respectively. CONCLUSIONS: We consider these oncolytic viruses as promising agents for the treatment of human malignant glioma.


Assuntos
Glioma , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Adenoviridae/fisiologia , Técnicas de Cultura de Células , Glioma/terapia , Glioma/virologia , Humanos , Orthopoxvirus/fisiologia , Parvovirus/fisiologia , Células Tumorais Cultivadas/virologia , Fenômenos Fisiológicos Virais
6.
Adv Virus Res ; 71: 135-71, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18585528

RESUMO

As a family of viruses, poxviruses collectively exhibit a broad host range and most of the individual members are capable of replicating in a wide array of cell types from various host species, at least in vitro. At the cellular level, poxvirus tropism is dependent not upon specific cell surface receptors, but rather upon: (1) the ability of the cell to provide intracellular complementing factors needed for productive virus replication, and (2) the ability of the specific virus to successfully manipulate intracellular signaling networks that regulate cellular antiviral processes downstream of virus entry. The large genomic coding capacity of poxviruses enables the virus to express a unique collection of viral proteins that function as host range factors, which specifically target and manipulate host signaling pathways to establish optimal cellular conditions for viral replication. Functionally, the known host range factors from poxviruses have been associated with manipulation of a diverse array of cellular targets, which includes cellular kinases and phosphatases, apoptosis, and various antiviral pathways. To date, only a small number of poxvirus host range genes have been identified and studied, and only a handful of these have been functionally characterized. For this reason, poxvirus host range factors represent a potential gold mine for the discovery of novel pathogen-host protein interactions. This review summarizes our current understanding of the mechanisms by which the known poxvirus host range genes, and their encoded factors, expand tropism through the manipulation of host cell intracellular signaling pathways.


Assuntos
Interações Hospedeiro-Patógeno , Myxoma virus/fisiologia , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Regulação Viral da Expressão Gênica , Humanos , Myxoma virus/química , Myxoma virus/genética , Orthopoxvirus/química , Orthopoxvirus/genética , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/fisiopatologia , Transdução de Sinais , Tropismo , Proteínas Virais/química , Proteínas Virais/genética , Replicação Viral
7.
Virol J ; 4: 8, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17224068

RESUMO

The potential use of variola virus, the causative agent of smallpox, as a bioweapon and the endemic presence of monkeypox virus in Africa demonstrate the need for better therapies for orthopoxvirus infections. Chemotherapeutic approaches to control viral infections have been less successful than those targeting bacterial infections. While bacteria commonly reproduce themselves outside of cells and have metabolic functions against which antibiotics can be directed, viruses replicate in the host cells using the cells' metabolic pathways. This makes it very difficult to selectively target the virus without damaging the host. Therefore, the development of antiviral drugs against poxviruses has initially focused on unique properties of the viral replication cycle or of viral proteins that can be selectively targeted. However, recent advances in molecular biology have provided insights into host factors that represent novel drug targets. The latest anti-poxvirus drugs are kinase inhibitors, which were originally developed to treat cancer progression but in addition block egress of poxviruses from infected cells. This review will summarize the current understanding of anti-poxvirus drugs and will give an overview of the development of the latest second generation poxvirus drugs.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Orthopoxvirus/efeitos dos fármacos , Fosfotransferases/antagonistas & inibidores , Animais , Antivirais/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Humanos , Camundongos , Orthopoxvirus/classificação , Orthopoxvirus/enzimologia , Orthopoxvirus/fisiologia , Infecções por Poxviridae/tratamento farmacológico , Infecções por Poxviridae/virologia , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
8.
J Virol ; 79(20): 13139-49, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16189015

RESUMO

ST-246 is a low-molecular-weight compound (molecular weight = 376), that is potent (concentration that inhibited virus replication by 50% = 0.010 microM), selective (concentration of compound that inhibited cell viability by 50% = >40 microM), and active against multiple orthopoxviruses, including vaccinia, monkeypox, camelpox, cowpox, ectromelia (mousepox), and variola viruses. Cowpox virus variants selected in cell culture for resistance to ST-246 were found to have a single amino acid change in the V061 gene. Reengineering this change back into the wild-type cowpox virus genome conferred resistance to ST-246, suggesting that V061 is the target of ST-246 antiviral activity. The cowpox virus V061 gene is homologous to vaccinia virus F13L, which encodes a major envelope protein (p37) required for production of extracellular virus. In cell culture, ST-246 inhibited plaque formation and virus-induced cytopathic effects. In single-cycle growth assays, ST-246 reduced extracellular virus formation by 10 fold relative to untreated controls, while having little effect on the production of intracellular virus. In vivo oral administration of ST-246 protected BALB/c mice from lethal infection, following intranasal inoculation with 10x 50% lethal dose (LD(50)) of vaccinia virus strain IHD-J. ST-246-treated mice that survived infection acquired protective immunity and were resistant to subsequent challenge with a lethal dose (10x LD(50)) of vaccinia virus. Orally administered ST-246 also protected A/NCr mice from lethal infection, following intranasal inoculation with 40,000x LD(50) of ectromelia virus. Infectious virus titers at day 8 postinfection in liver, spleen, and lung from ST-246-treated animals were below the limits of detection (<10 PFU/ml). In contrast, mean virus titers in liver, spleen, and lung tissues from placebo-treated mice were 6.2 x 10(7), 5.2 x 10(7), and 1.8 x 10(5) PFU/ml, respectively. Finally, oral administration of ST-246 inhibited vaccinia virus-induced tail lesions in Naval Medical Research Institute mice inoculated via the tail vein. Taken together, these results validate F13L as an antiviral target and demonstrate that an inhibitor of extracellular virus formation can protect mice from orthopoxvirus-induced disease.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Indóis/farmacologia , Orthopoxvirus/efeitos dos fármacos , Infecções por Poxviridae/prevenção & controle , Administração Oral , Sequência de Aminoácidos , Animais , Antivirais/efeitos adversos , Antivirais/química , Benzamidas/efeitos adversos , Benzamidas/química , Efeito Citopatogênico Viral/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Vírus da Ectromelia/isolamento & purificação , Ectromelia Infecciosa/prevenção & controle , Feminino , Indóis/efeitos adversos , Indóis/química , Isoindóis , Fígado/virologia , Pulmão/virologia , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Peso Molecular , Orthopoxvirus/isolamento & purificação , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Alinhamento de Sequência , Baço/virologia , Vacínia/prevenção & controle , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Montagem de Vírus/efeitos dos fármacos
9.
J Virol ; 78(22): 12147-56, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15507601

RESUMO

By using a homology-based bioinformatics approach, a structural model of the vaccinia virus (VV) I7L proteinase was developed. A unique chemical library of approximately 51,000 compounds was computationally queried to identify potential active site inhibitors. The resulting biased subset of compounds was assayed for both toxicity and the ability to inhibit the growth of VV in tissue culture cells. A family of chemotypically related compounds was found which exhibits selective activity against orthopoxviruses, inhibiting VV with 50% inhibitory concentrations of 3 to 12 microM. These compounds exhibited no significant cytotoxicity in the four cell lines tested and did not inhibit the growth of other organisms such as Saccharomyces cerevisiae, Pseudomonas aeruginosa, adenovirus, or encephalomyocarditis virus. Phenotypic analyses of virus-infected cells were conducted in the presence of active compounds to verify that the correct biochemical step (I7L-mediated core protein processing) was being inhibited. Electron microscopy of compound-treated VV-infected cells indicated a block in morphogenesis. Compound-resistant viruses were generated and resistance was mapped to the I7L open reading frame. Transient expression with the mutant I7L gene rescued the ability of wild-type virus to replicate in the presence of compound, indicating that this is the only gene necessary for resistance. This novel class of inhibitors has potential for development as an efficient antiviral drug against pathogenic orthopoxviruses, including smallpox.


Assuntos
Antivirais/farmacologia , Orthopoxvirus/efeitos dos fármacos , Sequência de Aminoácidos , Farmacorresistência Viral , Células HeLa , Humanos , Dados de Sequência Molecular , Orthopoxvirus/fisiologia , Replicação Viral/efeitos dos fármacos
10.
Methods Mol Biol ; 269: 51-64, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15114007

RESUMO

Poxvirus DNA is not infectious because the initiation of the infective process requires proteins encapsidated along with the virus genome. However, infectious virus can be produced if purified poxvirus DNA is transfected into cells previously infected with another poxvirus. This process is termed heterologous reactivation if the infecting virus is different from the transfected virus. We describe a method in which the high-frequency recombination and replication reactions catalyzed by the Leporipoxvirus, Shope fibroma virus (SFV), can be coupled with SFV-promoted reactivation reactions to rapidly construct recombinant vaccinia viruses in high yields (25-100% recombinant progeny). The reactivated vaccinia viruses are easily purified free of the SFV helper virus by plating mixed populations of virus on cells that support only the growth of vaccinia virus. These heterologous reactivation reactions can be used to manipulate the structure of virus genomes and produce viruses that express recombinant proteins at high levels. We illustrate the method by polymerase chain reaction (PCR) cloning the gene encoding green fluorescent protein (GFP), then using double-strand break repair reactions to produce a recombinant virus that expresses high levels of GFP.


Assuntos
DNA Viral/genética , Leporipoxvirus/genética , Orthopoxvirus/genética , Vaccinia virus/genética , Animais , Linhagem Celular , Cricetinae , Leporipoxvirus/fisiologia , Orthopoxvirus/fisiologia , Recombinação Genética , Vaccinia virus/crescimento & desenvolvimento
11.
J Cell Biochem ; 91(6): 1099-108, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15048867

RESUMO

Viruses utilize a variety of strategies to evade the host immune response and replicate in the cells they infect. The comparatively large genomes of the Orthopoxviruses and gammaherpesviruses encode several immunomodulatory proteins that are homologous to component of the innate immune system of host cells, which are reviewed here. However, the viral mechanisms used to survive host responses are quite distinct between these two virus families. Poxviruses undergo continuous lytic replication in the host cytoplasm while expressing many genes that inhibit innate immune responses. In contrast, herpesviruses persist in a latent state during much of their lifecycle while expressing only a limited number of relatively non-immunogenic viral proteins, thereby avoiding the adaptive immune response. Poxviruses suppress, whereas latent gammaherpesviruses activate, signaling by NF-kappaB, yet both viruses target similar host signaling pathways to suppress the apoptotic response. Here, modulation of apoptotic and NF-kappaB signal transduction pathways are examined as examples of common pathways appropriated in contrasting ways by herpesviruses and poxviruses.


Assuntos
Apoptose/fisiologia , Gammaherpesvirinae/fisiologia , NF-kappa B/metabolismo , Orthopoxvirus/fisiologia , Transdução de Sinais/fisiologia , Proteínas Virais/metabolismo , Animais , Apoptose/genética , Apoptose/imunologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Gammaherpesvirinae/genética , Gammaherpesvirinae/imunologia , Regulação Viral da Expressão Gênica/genética , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Camundongos , NF-kappa B/genética , Orthopoxvirus/genética , Orthopoxvirus/imunologia , Receptores de Citocinas/genética , Receptores de Citocinas/imunologia , Receptores de Citocinas/metabolismo , Transdução de Sinais/genética , Proteínas Virais/imunologia , Latência Viral/fisiologia , Replicação Viral/fisiologia
12.
J Virol ; 77(13): 7281-90, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12805426

RESUMO

Poxvirus DNA is not infectious because establishing an infection requires the activities of enzymes packaged in the virion. This barrier can be overcome by transfecting virus DNA into cells previously infected with another poxvirus, since the resident virus can provide the trans-acting systems needed to reactivate transfected DNA. In this study we show that cells infected with a leporipoxvirus, Shope fibroma virus (SFV), can reactivate vaccinia virus DNA. Similar heterologous packaging systems which used fowlpox-infected cells to reactivate vaccinia virus have been described, but SFV-infected cells promoted a far more efficient reaction that can produce virus titers exceeding 10(6) PFU/ micro g of transfected DNA. SFV-promoted reactions also exploit the hyperrecombinogenic systems previously characterized in SFV-infected cells, and these coupled recombination and reactivation reactions could be used to delete nonessential regions of the vaccinia virus genome and to reconstruct vaccinia virus from overlapping DNA fragments. SFV-catalyzed recombination reactions need only two 18- to 20-bp homologies to target PCR amplicons to restriction enzyme-cut vaccinia virus vectors, and this reaction feature was used to rapidly clone and express a gene encoding fluorescent green protein without the need for plaque purification or selectable markers. The ability of SFV-infected cells to reactivate fragments of vaccinia virus was ultimately limited by the number of recombinational exchanges required and one cannot reconstruct vaccinia virus from multiple PCR fragments spanning essential portions of the genome. These observations suggest that recombination is an integral part of poxvirus reactivation reactions and provide a useful new technique for altering the structure of poxvirus genomes.


Assuntos
DNA Viral/genética , Leporipoxvirus/genética , Orthopoxvirus/genética , Recombinação Genética , Ativação Viral , Animais , Sequência de Bases , Linhagem Celular , Cricetinae , Dano ao DNA , Reparo do DNA , Leporipoxvirus/fisiologia , Microscopia Confocal , Orthopoxvirus/fisiologia , Reação em Cadeia da Polimerase , Transfecção
13.
Virology ; 288(1): 175-87, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11543670

RESUMO

NF-kappaB comprises a family of transcription factors that regulate key immune processes. In this study, the effects of orthopoxvirus infection upon the activation of NF-kappaB were examined. During the early phase of infection, cowpox virus can inhibit the induction of NF-kappaB-regulated gene expression by interfering with the process of IkappaBalpha degradation. Although either okadaic acid or tumor necrosis factor (TNF) treatment of infected cells can induce IkappaBalpha phosphorylation, further processing of IkappaBalpha is inhibited. These results suggest that cowpox virus is capable of inhibiting the activation of NF-kappaB at a point where multiple signal transduction pathways converge. Other orthopoxviruses affect NF-kappaB activity, but in a type-specific manner. Raccoonpox virus and vaccinia virus (Copenhagen strain) negatively affect NF-kappaB induction by TNF. In contrast, the modified vaccinia virus Ankara strain induces NF-kappaB activation, even in the absence of other stimuli. These findings suggest that orthopoxviruses may affect a broad range of virus-host interactions through their effects upon NF-kappaB activation. Moreover, because of the central role for NF-kappaB in immune processes and disease, these type-specific effects may contribute significantly to the immunogenic and pathogenic properties of poxviruses.


Assuntos
Vírus da Varíola Bovina/fisiologia , Regulação da Expressão Gênica , Proteínas I-kappa B , NF-kappa B/metabolismo , Orthopoxvirus/fisiologia , Animais , Linhagem Celular , Cricetinae , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Rim , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Ácido Okadáico/farmacologia , Osteossarcoma , Fosforilação , Guaxinins , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia , Vaccinia virus/fisiologia
14.
Arch Immunol Ther Exp (Warsz) ; 44(5-6): 373-8, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-9017154

RESUMO

An 11-year global WHO campaign for eradication of smallpox finished in October 1977 as the result of Edward Jenner's primary success in 1796, who for the first time applied human vaccination against variola virus (VARV). The 200th anniversary of this happening is a good occasion to summarize the current status of the knowledge about the role of B and T lymphocytes in the control of orthopoxvirus infections. This short review concentrates on general characteristics of orthopoxviruses and the immune response to infection, mainly by vaccinia virus (VV) and ectromelia virus (EV).


Assuntos
Orthopoxvirus/imunologia , Infecções por Poxviridae/prevenção & controle , Vacinas Virais , Animais , Antígenos Virais/imunologia , Reservatórios de Doenças , Glicoproteínas/imunologia , História do Século XVIII , Humanos , Imunidade Celular , Orthopoxvirus/genética , Orthopoxvirus/fisiologia , Infecções por Poxviridae/imunologia , Varíola/história , Varíola/prevenção & controle , Vacina Antivariólica/história , Vacinação/história , Proteínas do Envelope Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA