Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2225: 63-75, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108657

RESUMO

Myxoma virus (MYXV) has proven to be an effective candidate for oncolytic virotherapy in many preclinical cancer models. As a nonhuman pathogen, MYXV does not need to overcome any preexisting antiviral immunity, and its DNA cannot integrate into the host genome, making it an extremely safe vector. Moreover, the large dsDNA genome of MYXV allows the insertion of multiple transgenes and the design of engineered recombinant oncolytic viruses (OVs) with enhanced immunostimulatory or other desired properties. In this chapter, we describe detailed protocols for the generation and characterization of transgene-armed recombinant MYXV vectors.


Assuntos
Engenharia Genética/métodos , Genoma Viral , Myxoma virus/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Animais , Chlorocebus aethiops , Clonagem Molecular/métodos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Myxoma virus/metabolismo , Vírus Oncolíticos/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Coelhos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transgenes , Células Vero
2.
Methods Mol Biol ; 2225: 163-177, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108662

RESUMO

Viruses engineered to express fluorescent proteins can be used with live-cell imaging techniques to monitor the progression of infection in real time. Here we describe a set of methods to track infection spreading from one cell population to another as well as to visualize transfer of virions between cells. This approach is extended to multiplexing with physiological readouts of cell death, which can be correlated with single-cell resolution to viral infection.


Assuntos
Rastreamento de Células/métodos , Engenharia Genética/métodos , Myxoma virus/genética , Imagem Óptica/métodos , Proteínas Virais/genética , Vírion/genética , Células A549 , Apoptose/genética , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Myxoma virus/crescimento & desenvolvimento , Myxoma virus/metabolismo , Imagem Óptica/estatística & dados numéricos , Proteínas Virais/metabolismo , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Replicação Viral/genética , Proteína Vermelha Fluorescente
3.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32669330

RESUMO

Unlike RNA viruses, most DNA viruses replicate their genomes with high-fidelity polymerases that rarely make base substitution errors. Nevertheless, experimental evolution studies have revealed rapid acquisition of adaptive mutations during serial passage of attenuated vaccinia virus (VACV). One way in which adaptation can occur is by an accordion mechanism in which the gene copy number increases followed by base substitutions and, finally, contraction of the gene copy number. Here, we show rapid acquisition of multiple adaptive mutations mediated by a gene-inactivating frameshift mechanism during passage of an attenuated VACV. Attenuation had been achieved by exchanging the VACV A8R intermediate transcription factor gene with the myxoma virus ortholog. A total of seven mutations in six different genes occurred in three parallel passages of the attenuated virus. The most frequent mutations were single-nucleotide insertions or deletions within runs of five to seven As or Ts, although a deletion of 11 nucleotides also occurred, leading to frameshifts and premature stop codons. During 10 passage rounds, the attenuated VACV was replaced by the mutant viruses. At the end of the experiment, virtually all remaining viruses had one fixed mutation and one or more additional mutations. Although nucleotide substitutions in the transcription apparatus accounted for two low-frequency mutations, frameshifts in genes encoding protein components of the mature virion, namely, A26L, G6R, and A14.5L, achieved 74% to 98% fixation. The adaptive role of the mutations was confirmed by making recombinant VACV with A26L or G6R or both deleted, which increased virus replication levels and decreased particle/PFU ratios.IMPORTANCE Gene inactivation is considered to be an important driver of orthopoxvirus evolution. Whereas cowpox virus contains intact orthologs of genes present in each orthopoxvirus species, numerous genes are inactivated in all other members of the genus. Inactivation of additional genes can occur upon extensive passaging of orthopoxviruses in cell culture leading to attenuation in vivo, a strategy for making vaccines. Whether inactivation of multiple viral genes enhances replication in the host cells or has a neutral effect is unknown in most cases. Using an experimental evolution protocol involving serial passages of an attenuated vaccinia virus, rapid acquisition of inactivating frameshift mutations occurred. After only 10 passage rounds, the starting attenuated vaccinia virus was displaced by viruses with one fixed mutation and one or more additional mutations. The high frequency of multiple inactivating mutations during experimental evolution simulates their acquisition during normal evolution and extensive virus passaging to make vaccine strains.


Assuntos
Adaptação Biológica/genética , Mutação da Fase de Leitura , Myxoma virus/genética , Fatores de Transcrição/genética , Vaccinia virus/genética , Proteínas Virais/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Códon sem Sentido , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Dosagem de Genes , Aptidão Genética , Myxoma virus/metabolismo , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinação Genética , Fatores de Transcrição/metabolismo , Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Sequenciamento Completo do Genoma
4.
Methods Mol Biol ; 1826: 73-86, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30194594

RESUMO

The serpin family of serine proteinase inhibitors plays key roles in the maintenance of mammalian homeostasis. Virus-encoded serpins disrupt the balance of mammalian proteases to facilitate virus replication in the infected host. DNA viruses, in particular members of the poxvirus family, have acquired multiple copies of the functional serpins which are essential for viral pathogenesis. Virus-encoded serpins have proven to be very effective inhibitors of host proteases and thus are very attractive candidate molecules as immunomodulatory drugs. With this chapter we explain approaches to identifying immune-modulating viral serpins.


Assuntos
Myxoma virus , Serpinas , Proteínas Virais , Animais , Linhagem Celular , Myxoma virus/genética , Myxoma virus/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Coelhos , Serpinas/genética , Serpinas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29343569

RESUMO

Poxviruses are large, DNA viruses whose protein capsid is surrounded by one or more lipid envelopes. Embedded into these lipid envelopes are three conserved viral proteins which are thought to mediate binding of virions to target cells. While the function of these proteins has been studied in vitro, their specific roles during the pathogenesis of poxviral disease remain largely unclear. Here we present data demonstrating that the putative chondroitin binding protein M083 from the leporipoxvirus myxoma virus is a significant virulence factor during infection of susceptible Oryctolagus rabbits. Removal of M083 results in a reduced capacity of virus to spread beyond the regional lymph nodes and completely eliminates infection-mediated mortality. In vitro, removal of M083 results in only minor intracellular replication defects but causes a significant reduction in the ability of myxoma virus to spread from infected epithelial cells onto primary lymphocytes. We hypothesize that the physiological role of M083 is therefore to mediate the spread of myxoma virus onto rabbit lymphocytes, allowing these cells to disseminate virus throughout infected rabbits.IMPORTANCE Poxviruses represent both a class of human pathogens and potential therapeutic agents for the treatment of human malignancy. Understanding the basic biology of these agents is therefore significant to human health in a variety of ways. While the mechanisms mediating poxviral binding have been well studied in vitro, how these mechanisms impact poxviral pathogenesis in vivo remains unclear. The current study advances our understanding of how poxviral binding impacts viral pathogenesis by demonstrating that the putative chondroitin binding protein M083 plays a critical role during the pathogenesis of myxoma virus in susceptible Oryctolagus rabbits by impacting viral dissemination through changes in the transfer of virions onto primary splenocytes.


Assuntos
Linfócitos/virologia , Myxoma virus , Proteínas Virais , Células A549 , Animais , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Myxoma virus/genética , Myxoma virus/metabolismo , Myxoma virus/patogenicidade , Coelhos , Proteínas Virais/genética , Proteínas Virais/metabolismo
6.
Virology ; 503: 94-102, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28157624

RESUMO

In humans, deleterious mutations in the sterile α motif domain protein 9 (SAMD9) gene are associated with cancer, inflammation, weakening of the immune response, and developmental arrest. However, the biological function of SAMD9 and its sequence-structure relationships remain to be characterized. Previously, we found that an essential host range factor, M062 protein from myxoma virus (MYXV), antagonized the function of human SAMD9. In this study, we examine the interaction between M062 and human SAMD9 to identify regions that are critical to SAMD9 function. We also characterize the in vitro kinetics of the interaction. In an infection assay, exogenous expression of SAMD9 N-terminus leads to a potent inhibition of wild-type MYXV infection. We reason that this effect is due to the sequestration of viral M062 by the exogenously expressed N-terminal SAMD9 region. Our studies reveal the first molecular insight into viral M062-dependent mechanisms that suppress human SAMD9-associated antiviral function.


Assuntos
Myxoma virus/metabolismo , Proteínas/antagonistas & inibidores , Proteínas Virais/metabolismo , Células A549 , Animais , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Especificidade de Hospedeiro , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Myxoma virus/genética , Estrutura Terciária de Proteína , Proteínas/genética , Proteínas Virais/genética
7.
Proc Natl Acad Sci U S A ; 112(48): 14858-63, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26578811

RESUMO

Human sterile alpha motif domain-containing 9 (SAMD9) protein is a host restriction factor for poxviruses, but it can be overcome by some poxvirus host-range proteins that share homology with vaccinia virus C7 protein. To understand the mechanism of action for this important family of host-range factors, we determined the crystal structures of C7 and myxoma virus M64, a C7 family member that is unable to antagonize SAMD9. Despite their different functions and only 23% sequence identity, the two proteins have very similar overall structures, displaying a previously unidentified fold comprised of a compact 12-stranded antiparallel ß-sandwich wrapped in two short α helices. Extensive structure-guided mutagenesis of C7 identified three loops clustered on one edge of the ß sandwich as critical for viral replication and binding with SAMD9. The loops are characterized with functionally important negatively charged, positively charged, and hydrophobic residues, respectively, together forming a unique "three-fingered molecular claw." The key residues of the claw are not conserved in two C7 family members that do not antagonize SAMD9 but are conserved in distantly related C7 family members from four poxvirus genera that infect diverse mammalian species. Indeed, we found that all in the latter group of proteins bind SAMD9. Taken together, our data indicate that diverse mammalian poxviruses use a conserved molecular claw in a C7-like protein to target SAMD9 and overcome host restriction.


Assuntos
Myxoma virus/química , Proteínas/química , Vaccinia virus/química , Proteínas Virais/química , Cristalografia por Raios X , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Myxoma virus/genética , Myxoma virus/metabolismo , Estrutura Secundária de Proteína , Proteínas/genética , Proteínas/metabolismo , Vaccinia virus/genética , Vaccinia virus/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
8.
Virology ; 464-465: 134-145, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25068401

RESUMO

Although the production of single gene knockout viruses is a useful strategy to study viral gene functions, the redundancy of many host interactive genes within a complex viral genome can obscure their collective functions. In this study, a rabbit-specific poxvirus, myxoma virus (MYXV), was genetically altered to disrupt multiple members of the poxviral ankyrin-repeat (ANK-R) protein superfamily, M-T5, M148, M149 and M150. A particularly robust activation of the NF-κB pathway was observed in A549 cells following infection with the complete ANK-R knockout (vMyx-ANKsKO). Also, an increased release of IL-6 was only observed upon infection with vMyx-ANKsKO. In virus-infected rabbit studies, vMyx-ANKsKO was the most extensively attenuated and produced the smallest primary lesion of all ANK-R mutant constructs. This study provides the first insights into the shared functions of the poxviral ANK-R protein superfamily in vitro and in vivo.


Assuntos
Myxoma virus/patogenicidade , Infecções por Poxviridae/veterinária , Coelhos/imunologia , Coelhos/virologia , Proteínas Virais/metabolismo , Animais , Repetição de Anquirina , Deleção de Genes , Interleucina-6/genética , Interleucina-6/imunologia , Myxoma virus/genética , Myxoma virus/metabolismo , NF-kappa B/genética , NF-kappa B/imunologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/virologia , Coelhos/genética , Proteínas Virais/química , Proteínas Virais/genética , Virulência
9.
Clin Immunol ; 153(2): 254-63, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24845791

RESUMO

Many viruses encode virulence factors to facilitate their own survival by modulating a host's inflammatory response. One of these factors, secreted from cells infected with myxoma virus, is the serine proteinase inhibitor (serpin) Serp-1. Because Serp-1 had demonstrated anti-inflammatory properties in arterial injury models and viral infections, it was cloned and evaluated for therapeutic efficacy in collagen-induced arthritis (CIA). Clinical severity was significantly lower in the Serp-1 protocols (p<0.0001) and blinded radiographs indicated that the Serp-1 group had significantly less erosions than the controls (p<0.01). Delayed-type hypersensitivity was lower in the Serp-1 group but antibody titers to type II collagen were not significantly altered. Recipients had minimal histopathologic synovial changes and did not develop neutralizing antibodies to Serp-1. These results indicate that Serp-1 impedes the pathogenesis of CIA and suggests that the therapeutic potential of serine proteinase inhibitors in inflammatory joint diseases, such as rheumatoid arthritis, should be investigated further.


Assuntos
Artrite Experimental/prevenção & controle , Inibidores de Serina Proteinase/farmacologia , Serpinas/farmacologia , Proteínas Virais/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos/sangue , Anticorpos/imunologia , Artrite Experimental/diagnóstico por imagem , Artrite Experimental/imunologia , Contagem de Células Sanguíneas , Células CHO , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Feminino , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/prevenção & controle , Immunoblotting , Articulações/efeitos dos fármacos , Articulações/imunologia , Articulações/patologia , Dados de Sequência Molecular , Myxoma virus/genética , Myxoma virus/metabolismo , Radiografia , Ratos , Ratos Endogâmicos , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/imunologia , Serpinas/genética , Serpinas/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
10.
Am J Vet Res ; 73(8): 1252-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22849686

RESUMO

OBJECTIVE: To determine the oncolytic efficacy of an attenuated form of myxoma virus lacking the serp2 gene in canine tumor cells. SAMPLE: Primary cells were isolated from tumors that were surgically removed from dogs and from connective tissue obtained from the cadaver of a dog. Cells of various established cell lines from tumors and nontumorous tissues were obtained. PROCEDURES: Experiments were performed with cells in monolayer culture. Cell cultures were inoculated with wild-type myxoma viruses or myxoma viruses lacking the serp2 gene, and measures of cytopathic effects, viral growth kinetics, and cell death and apoptosis were determined. RESULTS: Myxoma viruses replicated in cells of many of the primary and established canine tumor cell lines. Canine tumor cells in which expression of activated protein kinase B was upregulated were more permissive to myxoma virus infection than were cells in which expression of activated protein kinase B was not upregulated. Myxoma viruses lacking the serp2 gene caused more cytopathic effects in canine tumor cells because of apoptosis than did wild-type myxoma viruses. CONCLUSIONS AND CLINICAL RELEVANCE: Results of the present study indicated myxoma viruses lacking the serp2 gene may be useful for treatment of cancer in dogs. Impact for Human Medicine-Results of the present study may be useful for development of novel oncolytic treatments for tumors in humans.


Assuntos
Doenças do Cão/terapia , Myxoma virus/fisiologia , Neoplasias/veterinária , Terapia Viral Oncolítica , Proteínas Virais/metabolismo , Animais , Apoptose , Western Blotting , Morte Celular , Linhagem Celular Tumoral , Cães , Deleção de Genes , Humanos , Microscopia Eletrônica de Transmissão , Myxoma virus/genética , Myxoma virus/metabolismo , Neoplasias/terapia , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Vírus Oncolíticos/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Coelhos , Proteínas Virais/genética
11.
Future Med Chem ; 4(12): 1553-65, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22917245

RESUMO

BACKGROUND: Resonant recognition model-myxoma virus (RRM-MV), a bioactive peptide analogue for myxoma virus MV-T5 protein, was computationally designed by the RRM. In this study, the anticancer effects of RRM-MV were assessed in vitro against four negative control peptides on human skin cancer and normal cells. RESULTS & DISCUSSION: The effects of RRM-MV versus negative control peptides on cells were evaluated by quantitative and qualitative assays. The RRM-MV treatment was able to induce cell death in cancer cells without triggering similar effects on normal cells. However, the negative control peptides produced no toxic effects on skin cancer and normal cells. No effects on human erythrocytes were detected when treated with all peptides. CONCLUSION: It is suggested that the RRM can be applied to design therapeutic anticancer peptides.


Assuntos
Peptídeos/toxicidade , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Humanos , L-Lactato Desidrogenase/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Myxoma virus/metabolismo , Peptídeos/química , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Proteínas Virais/química
12.
PLoS One ; 7(8): e43298, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905251

RESUMO

Graft-versus-host disease (GVHD) is a potentially lethal clinical complication arising from the transfer of alloreactive T lymphocytes into immunocompromised recipients. Despite conventional methods of T cell depletion, GVHD remains a major challenge in allogeneic hematopoietic cell transplant. Here, we demonstrate a novel method of preventing GVHD by ex vivo treatment of primary human hematopoietic cell sources with myxoma virus, a rabbit specific poxvirus currently under development for oncolytic virotherapy. This pretreatment dramatically increases post-transplant survival of immunocompromised mice injected with primary human bone marrow or peripheral blood cells and prevents the expansion of human CD3(+) lymphocytes in major recipient organs. Similar viral treatment also prevents human-human mixed alloreactive T lymphocyte reactions in vitro. Our data suggest that ex vivo virotherapy with myxoma virus can be a simple and effective method for preventing GVHD following infusion of hematopoietic products containing alloreactive T lymphocytes such as: allogeneic hematopoietic stem and progenitor cells, donor leukocyte infusions and blood transfusions.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Células-Tronco Hematopoéticas/citologia , Myxoma virus/metabolismo , Animais , Transfusão de Sangue , Células da Medula Óssea/citologia , Complexo CD3/biossíntese , Separação Celular , Doença Enxerto-Hospedeiro/etiologia , Efeito Enxerto vs Leucemia , Humanos , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Infecções por Poxviridae/metabolismo , Células-Tronco/citologia , Linfócitos T/virologia , Transplante Heterólogo , Viroses/metabolismo
13.
Gynecol Oncol ; 125(2): 441-50, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22306204

RESUMO

OBJECTIVE: We propose that metastatic epithelial ovarian cancer (EOC) is a potential therapeutic target for the oncolytic agent, Myxoma virus (MYXV). METHODS: Primary EOC cells were isolated from patient ascites and cultured as adherent cells or in suspension using Ultra Low-Attachment dishes. MYXV expressing green fluorescent protein was used to infect cells and spheroids. Infection was monitored by fluorescence microscopy, viral titering and immunoblotting for M-T7 and M130 virus protein expression, and cell viability by alamarBlue assay. Akti-1/2 (5 µM) and rapamycin (20 nM) were used to assay the role of PI3K-AKT signaling in mediating MYXV infection. RESULTS: Ascites-derived EOC cells grown in adherent culture are effectively killed by MYXV infection. EOC cells grown in suspension to form three-dimensional EOC spheroids readily permit MYXV entry into cells, yet are protected from the cytopathic effects of late MYXV infection. Upon reattachment (to model secondary metastasis), EOC spheroids are re-sensitized to MYXV-mediated oncolysis. The critical determinant that facilitates efficient MYXV infection is the presence of an activated PI3K-AKT signaling pathway. Treatment with the specific AKT inhibitor Akti-1/2 reduces infection of monolayer EOC cells and spheroids. Direct infection of freshly-collected ascites demonstrated that 54.5% of patient samples were sensitive to MYXV-mediated oncolytic cell killing. We also demonstrate that factor(s) present in ascites may negatively impact MYXV infection and oncolysis of EOC cells, which may be due to a down-regulation in endogenous AKT activity. CONCLUSIONS: Differential activity of AKT serves as the mechanistic basis for regulating MYXV-mediated oncolysis of EOC spheroids during key steps of the metastatic program. In addition, we provide the first evidence that MYXV oncolytic therapy may be efficacious for a significant proportion of ovarian cancer patients with metastatic disease.


Assuntos
Myxoma virus/fisiologia , Neoplasias Epiteliais e Glandulares/terapia , Proteína Oncogênica v-akt/metabolismo , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Ascite/patologia , Carcinoma Epitelial do Ovário , Feminino , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Microscopia de Fluorescência/métodos , Myxoma virus/genética , Myxoma virus/metabolismo , Neoplasias Epiteliais e Glandulares/enzimologia , Neoplasias Epiteliais e Glandulares/virologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/virologia , Fosfatidilinositol 3-Quinases/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas
14.
Methods Enzymol ; 460: 209-28, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19446727

RESUMO

Chemokines are important for activation of a host of cellular immune and inflammatory responses including cell signaling, activation, and communication. M-T7, a myxoma virus protein, inhibits the activity of chemokines by direct binding to chemokines and/or with glycosaminoglycans (GAGs). To study the effects of this chemokine-modulating protein (CMP), we use a variety of in vitro and in vivo techniques to evaluate M-T7 inhibition of inflammatory cells. To quickly analyze the effects of M-T7, changes in cell adhesion and membrane fluidity are measured as well as cell migration in mouse ascites. For more physiological analyses, an aortic transplant model in rodents is used to assess change in inflammatory cell infiltrates and vascular plaque growth (rejection). Utilization of the combination of these in vitro and in vivo techniques allows for a more complete study of the chemokine-modulating activity of M-T7, and can be used to study other immune and inflammation-modulating proteins.


Assuntos
Quimiocinas/farmacologia , Monócitos/efeitos dos fármacos , Myxoma virus/metabolismo , Proteínas Virais/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Humanos , Fluidez de Membrana/efeitos dos fármacos , Camundongos , Ligação Proteica/efeitos dos fármacos , Coelhos , Ratos
15.
J Virol ; 83(11): 5933-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19279088

RESUMO

Myxoma virus (MYXV) is a poxvirus pathogenic only for European rabbits, but its permissiveness in human cancer cells gives it potential as an oncolytic virus. A recombinant MYXV expressing both the tdTomato red fluorescent protein and interleukin-15 (IL-15) (vMyx-IL-15-tdTr) was constructed. Cells infected with vMyx-IL-15-tdTr secreted bioactive IL-15 and had in vitro replication kinetics similar to that of wild-type MYXV. To determine the safety of this virus for future oncolytic studies, we tested its pathogenesis in European rabbits. In vivo, vMyx-IL-15-tdTr no longer causes lethal myxomatosis. Thus, ectopic IL-15 functions as an antiviral cytokine in vivo, and vMyx-IL-15-tdTr is a safe candidate for animal studies of oncolytic virotherapy.


Assuntos
Interleucina-15/metabolismo , Myxoma virus/metabolismo , Mixomatose Infecciosa/patologia , Animais , Vetores Genéticos/genética , Interleucina-15/genética , Cinética , Myxoma virus/genética , Myxoma virus/patogenicidade , Plasmídeos/genética , Coelhos , Taxa de Sobrevida , Replicação Viral
16.
Biochim Biophys Acta ; 1784(1): 228-37, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17905673

RESUMO

Poxviruses demonstrate strict species specificity in vivo that range from narrow to broad, however the fundamental factors that mediate the basis of poxvirus tropism remain poorly understood. It is generally believed that most, if not all, poxviruses can efficiently bind and enter a wide range of mammalian cells and all of the known host anti-viral pathways that block viral replication in nonpremissive cells operate downstream of virus entry. A productive poxvirus infection is heavily dependent upon the production of a vast array of host modulatory products that specifically target and manipulate both extracellular immune response pathways of the host, as well as intracellular signal transduction pathways of the individually infected cells. The unique pathogenesis and host tropism of specific poxviruses can be attributed to the broad diversity of host modulatory proteins they express. Myxoma virus (MV) is a rabbit-specific poxviruses that encodes multiple host range factors, including an ankyrin-repeat protein M-T5, which functions to regulate tropism of MV for rabbit lymphocytes and some human cancer cells. At the molecular level, M-T5 binds and alters at least two distinct cellular proteins: Akt and cullin-1. The direct interaction between M-T5 and Akt was shown to be a key restriction determinant for MV tropism in a spectrum of human cancer cells making MV an excellent oncolytic candidate. Thus, the intricate relationship between viral encoded proteins and components of the host cell signaling networks can have profound impact on poxvirus tropism. The lessons we continue to learn from poxvirus host range factors like M-T5 will provide further insights into the factors that regulate poxvirus tropism and the mechanisms by which poxviruses micromanipulate the signaling pathways of the infected cell.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Culina/metabolismo , Myxoma virus/fisiologia , Transdução de Sinais , Proteínas Virais/metabolismo , Animais , Repetição de Anquirina , Antibióticos Antineoplásicos/farmacologia , Motivos F-Box , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Myxoma virus/genética , Myxoma virus/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Coelhos , Sirolimo/farmacologia , Proteínas Virais/química , Proteínas Virais/genética
17.
Mol Cell ; 25(6): 933-42, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17386268

RESUMO

Many viruses express antiapoptotic proteins to counter host defense mechanisms that would otherwise trigger the rapid clearance of infected cells. For example, adenoviruses and some gamma-herpesviruses express homologs of prosurvival Bcl-2 to subvert the host's apoptotic machinery. Myxoma virus, a double-stranded DNA virus of the pox family, harbors antiapoptotic M11L, its virulence factor. Analysis of its three-dimensional structure reveals that despite lacking any primary sequence similarity to Bcl-2, it adopts a virtually identical protein fold. This allows it to associate with BH3 domains, especially those of Bax and Bak. We found that M11L acts primarily by sequestering Bax and Bak, thereby blocking the killing action of these essential cell-death mediators. These findings expand the family of protein sequences that act like Bcl-2 to block apoptosis and support the conclusion that the prosurvival action of these proteins critically depends on their ability to bind and antagonize Bax and/or Bak.


Assuntos
Proteínas Proto-Oncogênicas c-bcl-2/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Sequência de Aminoácidos , Apoptose , Morte Celular , Modelos Moleculares , Dados de Sequência Molecular , Myxoma virus/metabolismo , Conformação Proteica , Dobramento de Proteína , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/química , Proteína X Associada a bcl-2/química
18.
J Virol ; 81(1): 106-14, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17065210

RESUMO

Myxoma virus (MV) encodes a cell surface protein (M135R) that is predicted to mimic the host alpha/beta interferon receptor (IFN-alpha/beta-R) and thus prevent IFN-alpha/beta from triggering a host antiviral response. This prediction is based on sequence similarity to B18R, the viral IFN-alpha/beta-R from vaccinia virus (VV), which has been demonstrated to bind and inhibit type I interferons. However, M135R is only half the size of VV B18R. All other poxvirus-encoded IFN-alpha/beta-R homologs align only to the amino-terminal half of M135R. Peptide antibodies raised against M135R were used for immunoblotting and immunofluorescence and indicate that M135R is expressed as an early gene and that the product is a cell surface N-linked glycoprotein that is not secreted. In contrast to the predicted properties of M135R as an inhibitor of type I interferon, all binding and inhibition assays designed to demonstrate whether M135R can interact with IFN-alpha/beta have been negative. However, pathogenesis studies with a targeted M135-knockout MV construct (vMyx135KO) indicate that the deletion of M135R severely attenuates MV pathogenesis in the European rabbit. We propose that M135R is an important immunomodulatory virulence factor for myxomatosis but that the target immune ligand is not from the predicted type I interferon family and remains to be identified.


Assuntos
Proteínas de Membrana/metabolismo , Myxoma virus/patogenicidade , Proteínas Virais/metabolismo , Fatores de Virulência/fisiologia , Sequência de Aminoácidos , Animais , Imunofluorescência , Immunoblotting , Proteínas de Membrana/análise , Proteínas de Membrana/química , Dados de Sequência Molecular , Myxoma virus/metabolismo , Myxoma virus/fisiologia , Mixomatose Infecciosa/virologia , Coelhos , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/fisiologia , Alinhamento de Sequência , Vaccinia virus/metabolismo , Proteínas Virais/análise , Proteínas Virais/química , Fatores de Virulência/análise , Fatores de Virulência/química , Replicação Viral
19.
Proc Natl Acad Sci U S A ; 103(12): 4640-5, 2006 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-16537421

RESUMO

We demonstrate that the susceptibility of human cancer cells to be infected and killed by an oncolytic poxvirus, myxoma virus (MV), is related to the basal level of endogenous phosphorylated Akt. We further demonstrate that nonpermissive tumor cells will switch from resistant to susceptible for MV infection after expression of ectopically active Akt (Myr-Akt) and that permissive cancer cells can be rendered nonpermissive by blocking Akt activation with a dominant-negative inhibitor of Akt. Finally, the activation of Akt by MV involves the formation of a complex between the viral host range ankyrin-repeat protein, M-T5, and Akt. We conclude that the Akt pathway is a key restriction determinant for permissiveness of human cancer cells by MV.


Assuntos
Myxoma virus/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Virais/metabolismo , Animais , Repetição de Anquirina , Linhagem Celular Tumoral , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Genes Virais , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores
20.
J Biol Chem ; 281(12): 8041-50, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16407226

RESUMO

The thrombolytic serine protease cascade is intricately involved in activation of innate immune responses. The urokinase-type plasminogen activator and receptor form complexes that aid inflammatory cell invasion at sites of arterial injury. Plasminogen activator inhibitor-1 is a mammalian serpin that binds and regulates the urokinase receptor complex. Serp-1, a myxomaviral serpin, also targets the urokinase receptor, displaying profound anti-inflammatory and anti-atherogenic activity in a wide range of animal models. Serp-1 reactive center site mutations, mimicking known mammalian and viral serpins, were constructed in order to define sequences responsible for regulation of inflammation. Thrombosis, inflammation, and plaque growth were assessed after treatment with Serp-1, Serp-1 chimeras, plasminogen activator inhibitor-1, or unrelated viral serpins in plasminogen activator inhibitor or urokinase receptor-deficient mouse aortic transplants. Altering the P1-P1' Arg-Asn sequence compromised Serp-1 protease-inhibitory activity and anti-inflammatory activity in animal models; P1-P1' Ala-Ala mutants were inactive, P1 Met increased remodeling, and P1' Thr increased thrombosis. Substitution of Serp-1 P2-P7 with Ala6 allowed for inhibition of urokinase but lost plasmin inhibition, unexpectedly inducing a diametrically opposed, proinflammatory response with mononuclear cell activation, thrombosis, and aneurysm formation (p < 0.03). Other serpins did not reproduce Serp-1 activity; plasminogen activator inhibitor-1 increased thrombosis (p < 0.0001), and unrelated viral serpin, CrmA, increased inflammation. Deficiency of urokinase receptor in mouse transplants blocked Serp-1 and chimera activity, in some cases increasing inflammation. In summary, 1) Serp-1 anti-inflammatory activity is highly dependent upon the reactive center loop sequence, and 2) plasmin inhibition is central to anti-inflammatory activity.


Assuntos
Myxoma virus/metabolismo , Serpinas/química , Animais , Aorta/metabolismo , Aorta/transplante , Sequência de Bases , Sítios de Ligação , Coagulação Sanguínea , Células CHO , Adesão Celular , Movimento Celular , Separação Celular , Cricetinae , Modelos Animais de Doenças , Citometria de Fluxo , Sistema Imunitário , Inflamação , Cinética , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Estatísticos , Dados de Sequência Molecular , Peritônio/citologia , Peritônio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativação Plaquetária , Ligação Proteica , Isoformas de Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Trombose , Fatores de Tempo , Tempo de Coagulação do Sangue Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA