Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 21427, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34728659

RESUMEN

A promising therapy for patients with B-cell lymphoma is based on vaccination with idiotype monoclonal antibodies (mAbs). Since idiotypes are different in each tumor, a personalized vaccine has to be produced for each patient. Expression of immunoglobulins with appropriate post-translational modifications for human use often requires the use of stable mammalian cells that can be scaled-up to reach the desired level of production. We have used a noncytopathic self-amplifying RNA vector derived from Semliki Forest virus (ncSFV) to generate BHK cell lines expressing murine follicular lymphoma-derived idiotype A20 mAb. ncSFV/BHK cell lines expressed approximately 2 mg/L/24 h of A20 mAb with proper quaternary structure and a glycosylation pattern similar to that of A20 mAb produced by hybridoma cells. A20 mAb purified from the supernatant of a ncSFV cell line, or from the hybridoma, was conjugated to keyhole limpet hemocyanin and used to immunize Balb/c mice by administration of four weekly doses of 25 µg of mAb. Both idiotype mAbs were able to induce a similar antitumor protection and longer survival compared to non-immunized mice. These results indicate that the ncSFV RNA vector could represent a quick and efficient system to produce patient-specific idiotypes with potential application as lymphoma vaccines.


Asunto(s)
Alphavirus/genética , Anticuerpos Monoclonales/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Vectores Genéticos/administración & dosificación , Idiotipos de Inmunoglobulinas/inmunología , Linfoma de Células B/terapia , Vacunación/métodos , Animales , Anticuerpos Monoclonales/inmunología , Apoptosis , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Proliferación Celular , Femenino , Vectores Genéticos/genética , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Ther ; 27(11): 1892-1905, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31563534

RESUMEN

Immune checkpoint blockade has shown anti-cancer efficacy, but requires systemic administration of monoclonal antibodies (mAbs), often leading to adverse effects. To avoid toxicity, mAbs could be expressed locally in tumors. We developed adeno-associated virus (AAV) and Semliki Forest virus (SFV) vectors expressing anti-programmed death ligand 1 (aPDL1) mAb. When injected intratumorally in MC38 tumors, both viral vectors led to similar local mAb expression at 24 h, diminishing quickly in SFV-aPDL1-treated tumors. However, SFV-aPDL1 induced >40% complete regressions and was superior to AAV-aPDL1, as well as to aPDL1 mAb given systemically or locally. SFV-aPDL1 induced abscopal effects and was also efficacious against B16-ovalbumin (OVA). The higher SFV-aPDL1 antitumor activity could be related to local upregulation of interferon-stimulated genes because of SFV RNA replication. This was confirmed by combining local SFV-LacZ administration and systemic aPDL1 mAb, which provided higher antitumor effects than each separated agent. SFV-aPDL1 promoted tumor-specific CD8 T cells infiltration in both tumor models. In MC38, SFV-aPDL1 upregulated co-stimulatory markers (CD137/OX40) in tumor CD8 T cells, and its combination with anti-CD137 mAb showed more pronounced antitumor effects than each single agent. These results indicate that local transient expression of immunomodulatory mAbs using non-propagative RNA vectors inducing type I interferon (IFN-I) responses represents a potent and safe approach for cancer treatment.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Expresión Génica , Vectores Genéticos/genética , Neoplasias/genética , Neoplasias/inmunología , Virus ARN/genética , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Inmunomodulación/efectos de los fármacos , Inmunofenotipificación , Inyecciones Intralesiones , Ratones , Neoplasias/patología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/genética , Virus de los Bosques Semliki/genética , Tasa de Supervivencia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Carga Tumoral
3.
Cell Mol Life Sci ; 73(20): 3897-916, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27117550

RESUMEN

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.


Asunto(s)
Alphavirus/patogenicidad , Cápside/metabolismo , Membrana Celular/virología , Micropartículas Derivadas de Células/virología , Alphavirus/genética , Animales , Fusión Celular , Línea Celular , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/ultraestructura , Femenino , Genoma Viral , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones Endogámicos C57BL , Pruebas de Neutralización , ARN Viral/metabolismo , Virus de los Bosques Semliki/patogenicidad , Transfección , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo
4.
BMC Cancer ; 15: 620, 2015 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-26347489

RESUMEN

BACKGROUND: Metastatic breast cancer is a major cause of death among women worldwide; therefore efficient therapeutic strategies are extremely needed. In this work we have developed a gene therapy- and bacteria-based combined neoadjuvant approach and evaluated its antitumor effect in a clinically relevant animal model of metastatic breast cancer. METHODS: 2×10(8) particles of a Semliki Forest virus vector expressing interleukin-12 (SFV-IL-12) and/or 2×10(7) units of an aroC (-) Samonella Typhimurium strain (LVR01) were injected into 4T1 tumor nodules orthotopically implanted in mice. Tumors were surgically resected and long-term survival was determined. IL-12 and interferon-γ were quantified by Enzyme-Linked ImmunoSorbent Assay, bacteria was visualized by inmunohistochemistry and the number of lung metastasis was calculated with a clonogenic assay. RESULTS: SFV-IL-12 and LVR01 timely inoculated and followed by surgical resection of tumors succeeded in complete inhibition of lethal lung metastasis and long-term survival in 90% of treated mice. The combined therapy was markedly synergistic compared to each treatment alone, since SFV-IL-12 monotherapy showed a potent antiangiogenic effect, being able to inhibit tumor growth and extend survival, but could not prevent establishment of distant metastasis and death of tumor-excised animals. On the other hand, LVR01 alone also showed a significant, although limited, antitumor potential, despite its ability to invade breast cancer cells and induce granulocyte recruitment. The efficacy of the combined therapy depended on the order in which both factors were administered; inasmuch the therapeutic effect was only observed when SFV-IL-12 was administered previous to LVR01, whereas administration of LVR01 before SFV-IL-12 had negligible antitumor activity. Moreover, pre-treatment with LVR01 seemed to suppress SFV-IL-12 antiangiogenic effects associated to lower IL-12 expression in this group. Re-challenged mice were unable to reject a second 4T1 tumor; however 100% of them could be totally cured by applying the same neoadjuvant combined regimen. To our knowledge, these are the most encouraging results obtained to date in a post-operatory setting using the highly aggressive 4T1 animal model. CONCLUSIONS: SFV-IL-12-based gene therapy combined with Salmonella LVR01 neoadjuvant administration has a synergic antitumor effect and may be a promising therapeutic option to prevent and/or eradicate pre-operatory metastasis in locally advanced breast cancer.


Asunto(s)
Neoplasias de la Mama/terapia , Regulación de la Expresión Génica/fisiología , Terapia Genética , Interleucina-12/genética , Neoplasias Pulmonares/terapia , Salmonella typhimurium/genética , Virus de los Bosques Semliki/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Terapia Combinada , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Inmunocompetencia , Inmunohistoquímica , Interferón gamma/sangre , Interleucina-12/sangre , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Terapia Neoadyuvante , Trasplante de Neoplasias , Fragmentos de Péptidos/sangre , Tasa de Supervivencia
5.
Cancer Immunol Res ; 3(5): 449-54, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25691326

RESUMEN

Virotherapy and checkpoint inhibitors can be combined for the treatment of cancer with complementarity and potential for synergistic effects. We have developed a cytolytic but nonreplicative viral vector system based on Semliki Forest virus that encodes IL12 (SFV-IL12). Following direct intratumoral injection, infected cells release transgenic IL12, die, and elicit an inflammatory response triggered by both abundantly copied viral RNA and IL12. In difficult-to-treat mouse cancer models, such as those derived from MC38 and bilateral B16-OVA, SFV-IL12 synergized with an anti-PD-1 monoclonal antibody (mAb) to induce tumor regression and prolong survival. Similar synergistic effects were attained upon PD-L1 blockade. Combined SFV-IL12 + anti-PD-1 mAb treatment only marginally increased the elicited cytotoxic T-lymphocyte response over SFV-IL12 as a single agent, at least when measured by in vivo killing assays. In contrast, we observed that SFV-IL12 treatment induced expression of PD-L1 on tumor cells in an IFNγ-dependent fashion. PD-L1-mediated adaptive resistance thereby provides a mechanistic explanation of the observed synergistic effects achieved by the SFV-IL12 + anti-PD-1 mAb combination.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Interleucina-12/genética , Neoplasias/terapia , Viroterapia Oncolítica , Receptor de Muerte Celular Programada 1/inmunología , Virus de los Bosques Semliki/genética , Animales , Línea Celular Tumoral , Femenino , Vectores Genéticos , Interleucina-12/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Bazo/citología
6.
Cancer Res ; 75(3): 497-507, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25527611

RESUMEN

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.


Asunto(s)
Antineoplásicos/química , Terapia Genética/métodos , Vectores Genéticos , Interleucina-12/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Cruzamientos Genéticos , Células Dendríticas/citología , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Inmunoterapia , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Viral/genética , Proteínas Recombinantes/metabolismo , Transgenes
7.
Cell Mol Life Sci ; 71(23): 4637-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24794511

RESUMEN

We report a new method to generate high-expressing mammalian cell lines in a quick and efficient way. For that purpose, we developed a master cell line (MCL) containing an inducible alphavirus vector expressing GFP integrated into the genome. In the MCL, recombinant RNA levels increased >4,600-fold after induction, due to a doxycycline-dependent RNA amplification loop. The MCL maintained inducibility and expression during 50 passages, being more efficient for protein expression than a conventional cell line. To generate new cell lines, mutant LoxP sites were inserted into the MCL, allowing transgene and selection gene exchange by Cre-directed recombination, leading to quick generation of inducible cell lines expressing proteins of therapeutic interest, like human cardiotrophin-1 and oncostatin-M at several mg/l/24 h. These proteins contained posttranslational modifications, showed bioactivity, and were efficiently purified. Remarkably, this system allowed production of toxic proteins, like oncostatin-M, since cells able to express it could be grown to the desired amount before induction. These cell lines were easily adapted to growth in suspension, making this methodology very attractive for therapeutic protein production.


Asunto(s)
Alphavirus/genética , Línea Celular/metabolismo , Clonación Molecular/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Transgenes , Animales , Técnicas de Cultivo de Célula , Línea Celular/citología , Línea Celular/virología , Cricetinae , Citocinas/genética , ADN/genética , Genoma , Células Hep G2 , Humanos , Oncostatina M/genética , ARN/genética , Proteínas Recombinantes/genética
8.
J Immunol ; 190(6): 2994-3004, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23401594

RESUMEN

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.


Asunto(s)
Interleucina-12/biosíntesis , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/prevención & control , Virus de los Bosques Semliki/inmunología , Virus de los Bosques Semliki/metabolismo , Adenocarcinoma/inmunología , Adenocarcinoma/prevención & control , Adenocarcinoma/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/virología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , Células Cultivadas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/prevención & control , Neoplasias Colorrectales/virología , Cricetinae , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Interleucina-12/genética , Neoplasias Hepáticas Experimentales/virología , Ratones , Ratones Endogámicos C57BL
9.
Int J Pharm ; 440(1): 19-26, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22580212

RESUMEN

Human glial cell line-derived neurotrophic factor (hGDNF) is a very promising protein for the treatment of Parkinson's disease and other neurodegenerative disorders. The present work describes a quick and simple method to obtain a high amount of purified hGDNF using a mammalian cell-derived system. The method is based on the high expression level provided by a Semliki Forest virus vector and its ability to induce a strong shut-off of host-cell protein synthesis in mammalian cells. As a result, hGDNF is the only protein present in the supernatant and can be efficiently purified by a single chromatographic step. Using this system it was possible to eliminate other secreted proteins from the culture medium, like insulin-like growth factor-5, which are hard to remove using other hGDNF production methods. Purified hGDNF presents a complex glycosylation pattern typical of mammalian expression systems and is biologically active. This protocol could be extended to other secreted proteins and could be easily scaled up for industrial purposes.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Virus de los Bosques Semliki/genética , Animales , Línea Celular , Cricetinae , Fibroblastos , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Glicosilación , Humanos , Células PC12 , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Recent Pat Biotechnol ; 5(3): 212-26, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22360469

RESUMEN

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.


Asunto(s)
Alphavirus/genética , Vectores Genéticos/metabolismo , Proteínas Recombinantes/biosíntesis , Alphavirus/metabolismo , Animales , Patentes como Asunto , Proteínas Recombinantes/genética , Replicación Viral
11.
Virus Res ; 153(2): 179-96, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20692305

RESUMEN

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.


Asunto(s)
Alphavirus/genética , Vectores Genéticos , Neoplasias/terapia , Animales , Apoptosis , Femenino , Terapia Genética/métodos , Humanos , Interferón Tipo I/inmunología , Masculino , Neoplasias Mamarias Animales/terapia , Mastocitoma/terapia , Melanoma/terapia , Modelos Animales , Neoplasias/inmunología , Neoplasias/virología , Virus Oncolíticos , Neoplasias de la Próstata/terapia
12.
N Biotechnol ; 27(2): 138-48, 2010 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-20188220

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Vectores Genéticos/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ingeniería de Proteínas/métodos , Virus de los Bosques Semliki/genética , Transfección/métodos , Animales , Línea Celular , Citocinas/genética , Citocinas/uso terapéutico , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Ratones , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico
13.
Virology ; 376(1): 242-51, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18442838

RESUMEN

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.


Asunto(s)
Efecto Citopatogénico Viral , Expresión Génica , Vectores Genéticos , Biología Molecular/métodos , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/patogenicidad , Virología/métodos , Acetiltransferasas/biosíntesis , Acetiltransferasas/genética , Sustitución de Aminoácidos/genética , Animales , Fusión Artificial Génica , Línea Celular , Cricetinae , Cisteína Endopeptidasas/genética , Genes Reporteros , Mutación Missense , Señales de Localización Nuclear/genética , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Pase Seriado , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...