Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Oncotarget ; 8(6): 8999-9000, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28129638
2.
Cell Mol Life Sci ; 73(20): 3897-916, 2016 10.
Article in English | MEDLINE | ID: mdl-27117550

ABSTRACT

Alphavirus budding is driven by interactions between nucleocapsids assembled in the cytoplasm and envelope proteins present at the plasma membrane. So far, the expression of capsid and envelope proteins in infected cells has been considered an absolute requirement for alphavirus budding and propagation. In the present study, we show that Semliki Forest virus and Sindbis virus lacking the capsid gene can propagate in mammalian and insect cells. This propagation is mediated by the release of infectious microvesicles (iMVs), which are pleomorphic and have a larger size and density than wild-type virus. iMVs, which contain viral RNA inside and viral envelope proteins on their surface, are released at the plasma membrane and infect cells using the endocytic pathway in a similar way to wild-type virus. iMVs are not pathogenic in immunocompetent mice when injected intravenously, but can infect different organs like lungs and heart. Finally, we also show that alphavirus genomes without capsid can mediate the propagation of heterologous genes, making these vectors potentially interesting for gene therapy or vaccination studies. The minimalist infectious system described in this study shows that a self-replicating RNA able to express membrane proteins with binding and fusion properties is able to propagate, providing some insights into virus evolution.


Subject(s)
Alphavirus/pathogenicity , Capsid/metabolism , Cell Membrane/virology , Cell-Derived Microparticles/virology , Alphavirus/genetics , Animals , Cell Fusion , Cell Line , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/ultrastructure , Female , Genome, Viral , Green Fluorescent Proteins/metabolism , Humans , Mice, Inbred C57BL , Neutralization Tests , RNA, Viral/metabolism , Semliki forest virus/pathogenicity , Transfection , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism
3.
Cancer Res ; 75(3): 497-507, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25527611

ABSTRACT

Host responses are increasingly considered important for the efficacious response to experimental cancer therapies that employ viral vectors, but little is known about the specific nature of host responses required. In this study, we investigated the role of host type I interferons (IFN-I) in the efficacy of virally delivered therapeutic genes. Specifically, we used a Semliki Forest virus encoding IL12 (SFV-IL12) based on its promise as an RNA viral vector for cancer treatment. Intratumoral injection of SFV-IL12 induced production of IFN-I as detected in serum. IFN-I production was abolished in mice deficient for the IFNß transcriptional regulator IPS-1 and partially attenuated in mice deficient for the IFNß signaling protein TRIF. Use of bone marrow chimeric hosts established that both hematopoietic and stromal cells were involved in IFN-I production. Macrophages, plasmacytoid, and conventional dendritic cells were each implicated based on cell depletion experiments. Further, mice deficient in the IFN-I receptor (IFNAR) abolished the therapeutic activity of SFV-IL12, as did a specific antibody-mediated blockade of IFNAR signaling. Reduced efficacy was not caused by an impairment in IL12 expression, because IFNAR-deficient mice expressed the viral IL12 transgene even more strongly than wild-type (WT) hosts. Chimeric host analysis for the IFNAR involvement established a strict requirement in hematopoietic cells. Notably, although tumor-specific CD8 T lymphocytes expanded robustly after intratumoral injection of WT mice with SFV-IL12, this did not occur in mice where IFNAR was inactivated genetically or pharmacologically. Overall, our results argued that the antitumor efficacy of a virally based transgene therapeutic relied strongly on a vector-induced IFN-I response, revealing an unexpected mechanism of action that is relevant to a broad array of current translational products in cancer research.


Subject(s)
Antineoplastic Agents/chemistry , Genetic Therapy/methods , Genetic Vectors , Interleukin-12/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Crosses, Genetic , Dendritic Cells/cytology , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Immunotherapy , Interferon-alpha/metabolism , Interferon-beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA, Viral/genetics , Recombinant Proteins/metabolism , Transgenes
4.
J Immunol ; 190(6): 2994-3004, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23401594

ABSTRACT

Semliki Forest virus vectors expressing IL-12 (SFV-IL-12) were shown to induce potent antitumor responses against s.c. MC38 colon adenocarcinomas in immunocompetent mice. However, when MC38 tumors were implanted in liver, where colon tumors usually metastasize, SFV-IL-12 efficacy was significantly reduced. We reasoned that characterization of immune responses against intrahepatic tumors in responder and nonresponder animals could provide useful information for designing more potent antitumor strategies. Remarkably, SFV-IL-12 induced a high percentage of circulating tumor-specific CD8 T cells in all treated animals. Depletion studies showed that these cells were essential for SFV-IL-12 antitumor activity. However, in comparison with nonresponders, tumor-specific cells from responder mice acquired an effector-like phenotype significantly earlier, were recruited more efficiently to the liver, and, importantly, persisted for a longer period of time. All treated mice had high levels of functional specific CD8 T cells at 8 d posttreatment reflected by both in vivo killing and IFN-γ-production assays, but responder animals showed a more avid and persistent IFN-γ response. Interestingly, differences in immune responses between responders and nonresponders seemed to correlate with the immune status of the animals before treatment and were not due to the treatment itself. Mice that rejected tumors were protected against tumor rechallenge, indicating that sustained memory responses are required for an efficacious therapy. Interestingly, tumor-specific CD8 T cells of responder animals showed upregulation of IL-15Rα expression compared with nonresponders. These results suggest that SFV-IL-12 therapy could benefit from the use of strategies that could either upregulate IL-15Rα expression or activate this receptor.


Subject(s)
Interleukin-12/biosynthesis , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/prevention & control , Semliki forest virus/immunology , Semliki forest virus/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/prevention & control , Adenocarcinoma/virology , Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Colorectal Neoplasms/immunology , Colorectal Neoplasms/prevention & control , Colorectal Neoplasms/virology , Cricetinae , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/metabolism , Interleukin-12/genetics , Liver Neoplasms, Experimental/virology , Mice , Mice, Inbred C57BL
5.
Recent Pat Biotechnol ; 5(3): 212-26, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22360469

ABSTRACT

Alphaviruses contain a single-strand RNA genome that can be modified to express heterologous genes at high levels. Alphavirus vectors can be packaged within viral particles (VPs) or used as DNA/RNA layered systems. The broad tropism and high expression levels of alphavirus vectors have made them very attractive for applications like recombinant protein expression, vaccination or gene therapy. Expression mediated by alphavirus vectors is generally transient due to induction of apoptosis. However, during the last years several non-cytopathic mutations have been identified within the replicase sequence of different alphaviruses, allowing prolonged protein expression in culture cells. Some of these mutants, which have been patented, have allowed the generation of stable cell lines able to express recombinant proteins for extended periods of time in a constitutive or inducible manner. Production of alphavirus VPs usually requires cotransfection of cells with vector and helper RNAs providing viral structural proteins in trans. During this process full-length wild type (wt) genomes can be generated through recombination between different RNAs. Several new strategies to reduce wt virus generation during packaging, optimize VP production, increase packaging capacity, and provide VPs with specific targeting have been recently patented. Finally, hybrid vectors between alphavirus and other types of viruses have led to a number of patents with applications in vaccination, cancer therapy or retrovirus production.


Subject(s)
Alphavirus/genetics , Genetic Vectors/metabolism , Recombinant Proteins/biosynthesis , Alphavirus/metabolism , Animals , Patents as Topic , Recombinant Proteins/genetics , Virus Replication
6.
Virus Res ; 153(2): 179-96, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20692305

ABSTRACT

Alphaviruses contain a single strand RNA genome that can be easily modified to express heterologous genes at very high levels in a broad variety of cells, including tumor cells. Alphavirus vectors can be used as viral particles containing a packaged vector RNA, or directly as nucleic acids in the form of RNA or DNA. In the latter case alphavirus RNA is cloned within a DNA vector downstream of a eukaryotic promoter. Expression mediated by these vectors is generally transient due to the induction of apoptosis. The high expression levels, induction of apoptosis, and activation of type I IFN response are the key features that have made alphavirus vectors very attractive for cancer treatment and vaccination. Alphavirus vectors have been successfully used as vaccines to induce protective and therapeutic immune responses against many tumor-associated antigens in animal models of mastocytoma, melanoma, mammary, prostate, and virally induced tumors. Alphavirus vectors have also shown a high antitumoral efficacy by expressing antitumoral molecules in tumor cells, which include cytokines, antiangiogenic factors or toxic proteins. In these studies induction of apoptosis in tumor cells contributed to the antitumoral efficacy by the release of tumor antigens that can be uptaken by antigen presenting cells, enhancing immune responses against tumors. The potential use of alphaviruses as oncolytic agents has also been evaluated for avirulent strains of Semliki Forest virus and Sindbis virus. The fact that this latter virus has a natural tropism for tumor cells has led to many studies in which this vector was able to reach metastatic tumors when administered systemically. Other "artificial" strategies to increase the tropism of alphavirus for tumors have also been evaluated and will be discussed.


Subject(s)
Alphavirus/genetics , Genetic Vectors , Neoplasms/therapy , Animals , Apoptosis , Female , Genetic Therapy/methods , Humans , Interferon Type I/immunology , Male , Mammary Neoplasms, Animal/therapy , Mastocytoma/therapy , Melanoma/therapy , Models, Animal , Neoplasms/immunology , Neoplasms/virology , Oncolytic Viruses , Prostatic Neoplasms/therapy
7.
N Biotechnol ; 27(2): 138-48, 2010 May 31.
Article in English | MEDLINE | ID: mdl-20188220

ABSTRACT

Semliki Forest virus (SFV) vectors lead to high protein expression in mammalian cells, but expression is transient due to vector cytopathic effects, inhibition of host cell proteins and RNA-based expression. We have used a noncytopathic SFV mutant (ncSFV) RNA vector to generate stable cell lines expressing two human therapeutic proteins: insulin-like growth factor I (IGF-I) and cardiotrophin-1 (CT-1). Therapeutic genes were fused at the carboxy-terminal end of Puromycin N-acetyl-transferase gene by using as a linker the sequence coding for foot-and-mouth disease virus (FMDV) 2A autoprotease. These cassettes were cloned into the ncSFV vector. Recombinant ncSFV vectors allowed rapid and efficient selection of stable BHK cell lines with puromycin. These cells expressed IGF-I and CT-1 in supernatants at levels reaching 1.4 and 8.6 microg/10(6)cells/24 hours, respectively. Two cell lines generated with each vector were passaged ten times during 30 days, showing constant levels of protein expression. Recombinant proteins expressed at different passages were functional by in vitro signaling assays. Stability at RNA level was unexpectedly high, showing a very low mutation rate in the CT-1 sequence, which did not increase at high passages. CT-1 was efficiently purified from supernatants of ncSFV cell lines, obtaining a yield of approximately 2mg/L/24 hours. These results indicate that the ncSFV vector has a great potential for the production of recombinant proteins in mammalian cells.


Subject(s)
Cytokines/metabolism , Genetic Vectors/genetics , Insulin-Like Growth Factor I/metabolism , Protein Engineering/methods , Semliki forest virus/genetics , Transfection/methods , Animals , Cell Line , Cytokines/genetics , Cytokines/therapeutic use , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/therapeutic use , Mice , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use
8.
J Virol ; 83(23): 12266-78, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19740992

ABSTRACT

A vector based on Semliki Forest virus (SFV) expressing high levels of interleukin-12 (SFV-enhIL-12) has previously demonstrated potent antitumoral efficacy in small rodents with hepatocellular carcinoma (HCC) induced by transplantation of tumor cells. In the present study, the infectivity and antitumoral/antiviral effects of SFV vectors were evaluated in the clinically more relevant woodchuck model, in which primary HCC is induced by chronic infection with woodchuck hepatitis virus (WHV). Intratumoral injection of SFV vectors expressing luciferase or IL-12 resulted in high reporter gene activity within tumors and cytokine secretion into serum, respectively, demonstrating that SFV vectors infect woodchuck tumor cells. For evaluating antitumoral efficacy, woodchuck tumors were injected with increasing doses of SFV-enhIL-12, and tumor size was measured by ultrasonography following treatment. In five (83%) of six woodchucks, a dose-dependent, partial tumor remission was observed, with reductions in tumor volume of up to 80%, but tumor growth was restored thereafter. Intratumoral treatment further produced transient changes in WHV viremia and antigenemia, with >or=1.5-log(10) reductions in serum WHV DNA in half of the woodchucks. Antitumoral and antiviral effects were associated with T-cell responses to tumor and WHV antigens and with expression of CD4 and CD8 markers, gamma interferon, and tumor necrosis factor alpha in peripheral blood mononuclear cells, suggesting that immune responses against WHV and HCC had been induced. These experimental observations suggest that intratumoral administration of SFV-enhIL-12 may represent a strategy for treatment of chronic HBV infection and associated HCC in humans but indicate that this approach could benefit from further improvements.


Subject(s)
Carcinoma, Hepatocellular/veterinary , Hepatitis B Vaccines/immunology , Hepatitis B, Chronic/veterinary , Interleukin-12/immunology , Semliki forest virus/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Female , Hepatitis B Virus, Woodchuck/immunology , Hepatitis B Virus, Woodchuck/isolation & purification , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/therapy , Immunotherapy/methods , Interferon-gamma/biosynthesis , Interleukin-12/genetics , Liver/diagnostic imaging , Liver/pathology , Male , Marmota , Semliki forest virus/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Ultrasonography
9.
Virology ; 376(1): 242-51, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18442838

ABSTRACT

Alphavirus vectors express high levels of recombinant proteins in mammalian cells, but their cytopathic nature makes this expression transient. In order to generate a Semliki Forest virus (SFV) noncytopathic vector we introduced mutations previously described to turn Sindbis virus noncytopathic into a conserved position in an SFV vector expressing LacZ. Interestingly, mutant P718T in replicase nsp2 subunit was able to replicate in only a small percentage of BHK cells, producing beta-gal-expressing colonies without selection. Puromycin N-acetyl-transferase (pac) gene was used to replace LacZ in this mutant allowing selection of an SFV noncytopathic replicon containing a second mutation in nsp2 nuclear localization signal (R649H). This latter mutation did not confer a noncytopathic phenotype by itself and did not alter nsp2 nuclear translocation. Replicase synthesis was diminished in the SFV double mutant, leading to genomic and subgenomic RNA levels that were 125-fold and 66-fold lower than in wild-type vector, respectively. Interestingly, this mutant expressed beta-gal levels similar to parental vector. By coexpressing pac and LacZ from independent subgenomic promoters this vector was able to generate stable cell lines maintaining high expression levels during at least 10 passages, indicating that it could be used as a powerful system for protein production in mammalian cells.


Subject(s)
Cytopathogenic Effect, Viral , Gene Expression , Genetic Vectors , Molecular Biology/methods , Semliki forest virus/genetics , Semliki forest virus/pathogenicity , Virology/methods , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Amino Acid Substitution/genetics , Animals , Artificial Gene Fusion , Cell Line , Cricetinae , Cysteine Endopeptidases/genetics , Genes, Reporter , Mutation, Missense , Nuclear Localization Signals/genetics , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Serial Passage , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL