Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Exp Med ; 203(5): 1259-71, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16636135

RESUMEN

Radiotherapy is one of the most successful cancer therapies. Here the effect of irradiation on antigen presentation by MHC class I molecules was studied. Cell surface expression of MHC class I molecules was increased for many days in a radiation dose-dependent manner as a consequence of three responses. Initially, enhanced degradation of existing proteins occurred which resulted in an increased intracellular peptide pool. Subsequently, enhanced translation due to activation of the mammalian target of rapamycin pathway resulted in increased peptide production, antigen presentation, as well as cytotoxic T lymphocyte recognition of irradiated cells. In addition, novel proteins were made in response to gamma-irradiation, resulting in new peptides presented by MHC class I molecules, which were recognized by cytotoxic T cells. We show that immunotherapy is successful in eradicating a murine colon adenocarcinoma only when preceded by radiotherapy of the tumor tissue. Our findings indicate that directed radiotherapy can improve the efficacy of tumor immunotherapy.


Asunto(s)
Adenocarcinoma/inmunología , Presentación de Antígeno/efectos de la radiación , Neoplasias del Colon/inmunología , Rayos gamma , Antígeno HLA-A2/inmunología , Inmunoterapia , Adenocarcinoma/genética , Adenocarcinoma/terapia , Animales , Presentación de Antígeno/inmunología , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Relación Dosis-Respuesta en la Radiación , Regulación Neoplásica de la Expresión Génica/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Antígeno HLA-A2/genética , Humanos , Ratones , Ratones Transgénicos , Péptidos/inmunología , Biosíntesis de Proteínas/inmunología , Biosíntesis de Proteínas/efectos de la radiación , Proteínas Quinasas/inmunología , Radioterapia , Linfocitos T Citotóxicos/inmunología , Serina-Treonina Quinasas TOR
2.
J Virol ; 84(15): 7713-25, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20504925

RESUMEN

Alphavirus-based replicon vector systems (family Togaviridae) have been developed as expression vectors with demonstrated potential in vaccine development against both infectious diseases and cancer. The single-cycle nature of virus-like replicon particles (VRP), generated by supplying the structural proteins from separate replicable helper RNAs, is an attractive safety component of these systems. MicroRNAs (miRNAs) have emerged as important cellular RNA regulation elements. Recently, miRNAs have been employed as a mechanism to attenuate or restrict cellular tropism of replication-competent viruses, such as oncolytic adenoviruses, vesicular stomatitis virus, and picornaviruses as well as nonreplicating lentiviral and adenoviral vectors. Here, we describe the incorporation of miRNA-specific target sequences into replicable alphavirus helper RNAs that are used in trans to provide the structural proteins required for VRP production. VRP were found to be efficiently produced using miRNA-targeted helper RNAs if miRNA-specific inhibitors were introduced into cells during VRP production. In the absence of such inhibitors, cellular miRNAs were capable of downregulating helper RNA replication in vitro. When miRNA targets were incorporated into a replicon RNA, cellular miRNAs were capable of downregulating replicon RNA replication upon delivery of VRP into animals, demonstrating activity in vivo. These data provide the first example of miRNA-specific repression of alphavirus replicon and helper RNA replication and demonstrate the feasibility of miRNA targeting of expression vector helper functions that are provided in trans.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/genética , Marcación de Gen , Vectores Genéticos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Animales , Chlorocebus aethiops , Femenino , Ratones , Ratones Endogámicos BALB C , ARN Viral/genética , ARN Viral/metabolismo , Células Vero
3.
Clin Cancer Res ; 14(13): 4241-9, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18594006

RESUMEN

PURPOSE: Exposing human tumor cells to sublethal doses of external beam radiation up-regulates expression of tumor antigen and accessory molecules, rendering tumor cells more susceptible to killing by antigen-specific CTLs. This study explored the possibility that exposure to palliative doses of a radiopharmaceutical agent could alter the phenotype of tumor cells to render them more susceptible to T cell-mediated killing. EXPERIMENTAL DESIGN: Here, 10 human tumor cell lines (4 prostate, 2 breast, and 4 lung) were exposed to increasing doses of the radiopharmaceutical samarium-153-ethylenediaminetetramethylenephosphonate ((153)Sm-EDTMP) used in cancer patients to treat pain due to bone metastasis. Fluorescence-activated cell sorting analysis and quantitative real-time PCR analysis for expression of five surface molecules and several tumor-associated antigens involved in prostate cancer were done. LNCaP human prostate cancer cells were exposed to (153)Sm-EDTMP and incubated with tumor-associated antigen-specific CTL in a CTL killing assay to determine whether exposure to (153)Sm-EDTMP rendered LNCaP cells more susceptible to T cell-mediated killing. RESULTS: Tumor cells up-regulated the surface molecules Fas (100% of cell lines up-regulated Fas), carcinoembryonic antigen (90%), mucin-1 (60%), MHC class I (50%), and intercellular adhesion molecule-1 (40%) in response to (153)Sm-EDTMP. Quantitative real-time PCR analysis revealed additional up-regulated tumor antigens. Exposure to (153)Sm-EDTMP rendered LNCaP cells more susceptible to killing by CTLs specific for prostate-specific antigen, carcinoembryonic antigen, and mucin-1. CONCLUSIONS: Doses of (153)Sm-EDTMP equivalent to palliative doses delivered to bone alter the phenotype of tumor cells, suggesting that (153)Sm-EDTMP may work synergistically with immunotherapy to increase the susceptibility of tumor cells to CTL killing.


Asunto(s)
Neoplasias de la Mama/terapia , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/terapia , Compuestos Organometálicos/uso terapéutico , Compuestos Organofosforados/uso terapéutico , Neoplasias de la Próstata/terapia , Radioisótopos/farmacología , Samario/farmacología , Linfocitos T Citotóxicos/inmunología , Línea Celular Tumoral , Separación Celular , Femenino , Humanos , Masculino , Fenotipo , Linfocitos T Citotóxicos/metabolismo , Resultado del Tratamiento
4.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18594015

RESUMEN

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Asunto(s)
Antígenos de Neoplasias/química , Antineoplásicos/farmacología , Antígeno Carcinoembrionario/química , Regulación de la Expresión Génica , Inmunoterapia/métodos , Saccharomyces cerevisiae/metabolismo , Vacunas de ADN/química , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer , Antígeno Carcinoembrionario/metabolismo , Proliferación Celular , Femenino , Humanos , Linfocitos/citología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/química , Vacunas de ADN/metabolismo
5.
Clin Cancer Res ; 13(6): 1936-46, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363550

RESUMEN

PURPOSE: Interleukin (IL)-2 therapy is currently used for therapy of renal cell carcinoma (RCC). However, it is only effective in approximately 10% to 15% of patients, showing a need for additional therapies. We have previously described a replication-defective fowlpox vector encoding three costimulatory molecules (B7-1, ICAM-1, and LFA-3), designated rF-TRICOM. Here, we show that intratumoral administration of rF-TRICOM in an orthotopic RCC model effectively enhances tumor immunogenicity and reduces tumor burden in mice and the combination of rF-TRICOM and IL-2 is more effective than either therapy alone. EXPERIMENTAL DESIGN: RCC cells were implanted under the capsule of the kidney, and mice were given rF-TRICOM intratumorally 14 days later. We compared the effect of rF-TRICOM, rF-granulocyte macrophage colony-stimulating factor (GM-CSF), and two doses of IL-2 and combinations of the above on antitumor efficacy and survival. Host CD4(+) and CD8(+) T-cell responses were also evaluated. RESULTS: The results show that (a) systemic IL-2 therapy was moderately effective in the reduction of tumor burden in an orthotopic RCC model; (b) a single intratumoral injection of rF-TRICOM and rF-GM-CSF significantly reduced tumor burden; (c) the addition of systemic IL-2 to intratumoral rF-TRICOM/rF-GM-CSF administration resulted in further reduction of tumor burden, decrease in the incidence of metastasis, and extended survival in tumor-bearing mice above that seen with either treatment alone; and (d) CD8(+) T cells played a critical role in the antitumor effect seen with rF-TRICOM/rF-GM-CSF + IL-2 therapy. Finally, the addition of systemic recombinant IL-15 or intratumoral vector-delivered IL-15 to intratumoral rF-TRICOM/rF-GM-CSF administration resulted in substantially more tumor-free mice than either therapy alone. CONCLUSIONS: These studies show that intratumoral administration of rF-TRICOM admixed with rF-GM-CSF is effective at reducing tumor burden in mice and the addition of IL-2 further contributes to this effect. These studies thus form the rationale for combination immunotherapy clinical trials in patients with RCC.


Asunto(s)
Carcinoma de Células Renales/terapia , Terapia Genética/métodos , Inmunoterapia/métodos , Interleucina-2/uso terapéutico , Neoplasias Renales/terapia , Animales , Antígeno B7-1/genética , Antígenos CD58/genética , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Terapia Combinada , Femenino , Vectores Genéticos/administración & dosificación , Inyecciones Intralesiones , Molécula 1 de Adhesión Intercelular/genética , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/mortalidad , Neoplasias Experimentales/terapia , Carga Tumoral , Células Tumorales Cultivadas
6.
Front Biosci ; 12: 4900-10, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17569618

RESUMEN

Since its discovery more than a hundred years ago, radiation has been used to treat cancer. In recent decades, advances in radiation technology have expanded the role and value of radiation in imaging and treating many forms of cancer. Currently, there is a growing interest in combining radiation with other modalities, such as immunotherapy, to treat a broad range of malignancies. This article reviews the use of standard and novel combinations of radiation therapy and immunotherapy to eradicate tumor cells. The combination of radiation therapy and immunotherapy holds particular promise as a strategy for cancer therapeutics for a variety of reasons. First, there is evidence that immunotherapy is most beneficial when employed early in the disease process and in combination with standard therapies. In addition, radiation may act synergistically with immunotherapy to enhance immune responses, inhibit immunosuppression, and/or alter the phenotype of tumor cells, thus rendering them more susceptible to immune-mediated killing. Finally, as monotherapies, both immunotherapy and radiation may be insufficient to eliminate tumor masses. However, following immunization with a cancer vaccine, the destruction of even a small percentage of tumor cells by radiation could result in cross-priming and presentation of tumor antigens to the immune system, thereby potentiating antitumor responses.


Asunto(s)
Inmunoterapia , Neoplasias/terapia , Braquiterapia , Humanos , Metástasis de la Neoplasia , Neoplasias/radioterapia , Radioinmunodetección , Radioinmunoterapia , Radioterapia Adyuvante
7.
Vaccine ; 26(4): 509-21, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18155327

RESUMEN

Recombinant Saccharomyces cerevisiae (yeast) represents a unique and attractive vehicle to deliver antigens in vaccine immunotherapy protocols for cancer or infectious disease, in that it has been shown to be extremely safe and can be administered multiple times to hosts. In the studies reported here, we describe the effects of treatment with recombinant yeast on murine immature dendritic cells (DCs). Yeast expressing human carcinoembryonic antigen (CEA) as a model antigen was studied. Injection of mice subcutaneously with yeast-CEA resulted in rapid increases in MHC class II(+) cells and total antigen-presenting cells in draining lymph nodes. Post-treatment with yeast-CEA, DCs rapidly elevated both MHC class I and class II, numerous costimulatory molecules and other DC maturation markers, and secreted a range of Type I inflammatory cytokines. Gene expression arrays also revealed the rapid up-regulation of numerous cytokine and chemokine mRNAs, as well as genes involved in signal transduction and antigen uptake. Functional studies demonstrated enhanced allospecific reactivity of DCs following treatment with yeast-CEA or control yeast. Additionally, treatment of DCs with yeast-CEA resulted in specific activation of CEA-specific CD8(+) T cells in an MHC-restricted manner in vitro. Lastly, vaccination of CEA-transgenic mice with yeast-CEA elicited antigen-specific CD4(+) and CD8(+) immune responses in vivo. Thus, these studies taken together form a scientific rationale for the use of recombinant yeast in vaccination protocols for cancer or infectious diseases.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Vectores Genéticos/inmunología , Saccharomyces cerevisiae/inmunología , Vacunación , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Citocinas/biosíntesis , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inyecciones Subcutáneas , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Saccharomyces cerevisiae/genética
8.
Cancer Immunol Immunother ; 56(12): 1897-910, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17503041

RESUMEN

Systemic IL-2 is currently employed in the therapy of several tumor types, but at the price of often severe toxicities. Local vector mediated delivery of IL-2 at the tumor site may enhance local effector cell activity while reducing toxicity. To examine this, a model using CEA-transgenic mice bearing established CEA expressing tumors was employed. The vaccine regimen was a s.c. prime vaccination with recombinant vaccinia (rV) expressing transgenes for CEA and a triad of costimulatory molecules (TRICOM) followed by i.t. boosting with rF-CEA/TRICOM. The addition of intratumoral (i.t.) delivery of IL-2 via a recombinant fowlpox (rF) IL-2 vector greatly enhanced anti-tumor activity of a recombinant vaccine, resulting in complete tumor regression in 70-80% of mice. The anti-tumor activity was shown to be dependent on CD8(+) cells and NK1.1(+). Cellular immune assays revealed that the addition of rF-IL-2 to the vaccination therapy enhanced CEA-specific tetramer(+) cell numbers, cytokine release and CTL lysis of CEA(+) targets. Moreover, tumor-bearing mice vaccinated with the CEA/TRICOM displayed an antigen cascade, i.e., CD8(+) T cell responses to two other antigens expressed on the tumor and not the vaccine: wild-type p53 and endogenous retroviral antigen gp70. Mice receiving rF-IL-2 during vaccination demonstrated higher avidity CEA-specific, as well as higher avidity gp70-specific, CD8(+) T cells when compared with mice vaccinated without rF-IL-2. These studies demonstrate for the first time that the level and avidity of antigen specific CTL, as well as the therapeutic outcome can be improved with the use of i.t. rF-IL-2 with vaccine regimens.


Asunto(s)
Terapia Genética/métodos , Inmunoterapia/métodos , Interleucina-2/uso terapéutico , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Citometría de Flujo , Viruela Aviar/microbiología , Vectores Genéticos , Sistema Inmunológico , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL
9.
J Gen Virol ; 87(Pt 12): 3643-3648, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17098980

RESUMEN

The paramyxovirus Simian virus 5 (SV5) is largely non-cytopathic in human epithelial and fibroblast cells. WF-PIV has been described previously as a naturally occurring SV5 variant that encodes P and V proteins differing from the wild-type (WT) SV5 proteins in eight and five amino acid positions, respectively. In this study, it is shown that WF-PIV is like WT SV5 by being largely non-cytopathic in A549 lung epithelial cells. However, substitution of the WF-PIV P/V gene into the background of WT SV5 resulted in a hybrid virus (P/V-WF) that induced apoptotic cell death not seen with either of the parental viruses. The kinetics of HeLa cell killing and induction of apoptosis by the P/V-WF chimera differed from those of the previously described P/V-CPI- chimera by being slower and less extensive. HeLa cell killing by the P/V-WF chimera was effectively reduced by inhibitors of caspase-9, but not of caspase-8. These results demonstrate that an exchange of P/V genes from two non-cytopathic SV5 variants can produce apoptosis-inducing chimeras, and that the role of the SV5 P/V gene products in limiting apoptosis can be dependent on expression in the context of a native viral genome.


Asunto(s)
Inhibidores de Caspasas , Supervivencia Celular , Células Epiteliales/virología , Virus de la Parainfluenza 5/patogenicidad , Fosfoproteínas/genética , Recombinación Genética , Proteínas Virales/genética , Proteínas Estructurales Virales/genética , Apoptosis , Línea Celular , Línea Celular Tumoral , Efecto Citopatogénico Viral/genética , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Virus de la Parainfluenza 5/genética , Proteínas de Unión al ARN
10.
Virology ; 335(1): 131-44, 2005 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-15823612

RESUMEN

A paramyxovirus SV5 mutant (rSV5-P/V-CPI-) that encodes 6 naturally-occurring P/V gene substitutions is a potent inducer of type I interferon (IFN) and is restricted for low moi growth, two phenotypes not seen with WT SV5. In this study, we have compared the IFN sensitivity of WT SV5 and the rSV5-P/V-CPI- mutant in tumor cell lines and in cultures of normal primary cells. We have tested the hypothesis that differences in IFN induction elicited by WT rSV5 and rSV5-P/V-CPI- are responsible for differences in low moi growth and spread. In contrast to WT SV5, low moi infection of A549 lung carcinoma cells with rSV5-P/V-CPI- resulted in a plateau of virus production by 24-48 h pi when secreted IFN levels were between approximately 100 and 1000 U/ml. Gene microarray and RT-PCR analyses identified IFN genes and IFN-stimulated genes whose expression were increased by infection of A549 cells with WT and P/V mutant viruses. Restricted low moi growth and spread of rSV5-P/V-CPI- in A549 cells was relieved in the presence of neutralizing antibodies to IFN-beta but not TNF-alpha. When A549 or MDA-MB-435 breast tumor cells were pretreated with IFN, both WT and P/V mutant viruses showed delayed spread and approximately 10-fold reduction in virus yield, but infections were not eliminated. Using normal primary human epithelial cells that have undergone limited passage in culture, WT rSV5 and rSV5-P/V-CPI- displayed high moi growth properties that were similar to that seen in A549 cells. However, IFN pretreatment of these primary cells as well as normal human lung cells eliminated low moi spread of both mutant and WT rSV5 infections. Together, these data demonstrate that SV5 growth in normal primary human cells is highly sensitive to IFN compared to growth in some tumor cell lines, regardless of whether the P/V gene is WT or mutant. These results suggest a model in which spread of WT SV5 in normal human cells is dependent on the ability of the virus to prevent IFN synthesis. The implications of these results for the use of recombinant paramyxoviruses as vectors are discussed.


Asunto(s)
Interferón Tipo I/farmacología , Mutación , Virus de la Parainfluenza 5/crecimiento & desarrollo , Fosfoproteínas/genética , Recombinación Genética , Proteínas Virales/genética , Proteínas Estructurales Virales/genética , Línea Celular Tumoral , Células Cultivadas , Humanos , Interferón Tipo I/metabolismo , Virus de la Parainfluenza 5/efectos de los fármacos , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/patogenicidad , Proteínas de Unión al ARN
11.
J Virol ; 76(20): 10109-21, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12239285

RESUMEN

The V protein of the paramyxovirus simian virus 5 (SV5) is responsible for targeted degradation of STAT1 and the block in alpha/beta interferon (IFN-alpha/beta) signaling that occurs after SV5 infection of human cells. We have analyzed the growth properties of a recombinant SV5 that was engineered to be defective in targeting STAT1 degradation. A recombinant SV5 (rSV5-P/V-CPI-) was engineered to contain six naturally occurring P/V protein mutations, three of which have been shown in previous transfection experiments to disrupt the V-mediated block in IFN-alpha/beta signaling. In contrast to wild-type (WT) SV5, human cells infected with rSV5-P/V-CPI- had STAT1 levels similar to those in mock-infected cells. Cells infected with rSV5-P/V-CPI- were found to express higher-than-WT levels of viral proteins and mRNA, suggesting that the P/V mutations had disrupted the regulation of viral RNA synthesis. Despite the inability to target STAT1 for degradation, single-step growth assays showed that the rSV5-P/V-CPI- mutant virus grew better than WT SV5 in all cell lines tested. Unexpectedly, cells infected with rSV5-P/V-CPI- but not WT SV5 showed an activation of a reporter gene that was under control of the IFN-beta promoter. The secretion of IFN from cells infected with rSV5-P/V-CPI- but not WT SV5 was confirmed by a bioassay for IFN. The rSV5-P/V-CPI- mutant grew to higher titers than did WT rSV5 at early times in multistep growth assays. However, rSV5-P/V-CPI- growth quickly reached a final plateau while WT rSV5 continued to grow and produced a final titer higher than that of rSV5-P/V-CPI- by late times postinfection. In contrast to WT rSV5, infection of a variety of cell lines with rSV5-P/V-CPI- induced cell death pathways with characteristics of apoptosis. Our results confirm a role for the SV5 V protein in blocking IFN signaling but also suggest new roles for the P/V gene products in controlling viral gene expression, the induction of IFN-alpha/beta synthesis, and virus-induced apoptosis.


Asunto(s)
Apoptosis , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Paramyxoviridae/patogenicidad , Fosfoproteínas/fisiología , Proteínas Virales/fisiología , Proteínas Estructurales Virales/fisiología , Sustitución de Aminoácidos , Animales , Bovinos , Línea Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral , Proteínas de Unión al ADN/metabolismo , Genes Virales , Proteínas Fluorescentes Verdes , Humanos , Interferón-alfa/genética , Interferón beta/genética , Proteínas Luminiscentes/genética , Mutagénesis , Paramyxoviridae/genética , Paramyxoviridae/crecimiento & desarrollo , Fosfoproteínas/genética , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , Proteínas de Unión al ARN , Factor de Transcripción STAT1 , Transactivadores/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Estructurales Virales/genética
12.
Virology ; 316(1): 41-54, 2003 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-14599789

RESUMEN

Infection of human cells with the paramyxovirus simian virus 5 (SV5) results in minimal cytopathic effect, and host interferon (IFN) and apoptotic pathways are not activated. We have previously shown that an rSV5 containing six naturally occurring P/V gene substitutions (rSV5-P/V-CPI-) displays premature and elevated expression of viral RNA and protein. In addition, cells infected with rSV5-P/V-CPI- show induction of the IFN-beta promoter as well as activation of IFN signaling and apoptotic pathways. In this article, we have tested the hypothesis that rSV5-WT can supply trans-acting factors that prevent host cell antiviral responses induced by rSV5-P/V-CPI-. During coinfection of human A549 cells, rSV5-WT blocked cell rounding, loss of cell volume, and DNA fragmentation induced by rSV5-P/V-CPI-, three later events in the apoptotic pathway, but was not able to block the loss of mitochondrial membrane potential (DeltaPsi(m)), an early event in the cell death process. As expected, IFN signaling was blocked during coinfections, and this was attributed to the loss of STAT1 induced by the rSV5-WT V protein. Surprisingly, simultaneous infection with rSV5-WT could not suppress the activation of the IFN-beta promoter by rSV5-P/V-CPI- infection. However, the IFN-beta promoter was not activated in cells that were first preinfected for 1 h with rSV5-WT and then subsequently infected with rSV5-P/V-CPI-. A model is proposed for activation of host responses to infection with the rSV5-P/V-CPI- mutant and the steps that are blocked by rSV5-WT.


Asunto(s)
Apoptosis , Interferón beta/metabolismo , Mutación , Virus de la Parainfluenza 5/patogenicidad , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Proteínas Virales/genética , Proteínas Estructurales Virales/genética , Línea Celular , Efecto Citopatogénico Viral , Humanos , Interferón-alfa/metabolismo , Interferón beta/genética , Potenciales de la Membrana , Mitocondrias , Virus de la Parainfluenza 5/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN , Transducción de Señal , Activación Transcripcional , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/metabolismo
13.
Virology ; 293(1): 192-203, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11853412

RESUMEN

In most tissue culture cell lines tested, infection with the paramyxovirus simian virus 5 (SV5) results in very little cell death. To determine if SV5 could be used as a vector for controlled killing of tumor cells, a recombinant SV5 (rSV5-TK) was constructed to encode the herpes simplex virus thymidine kinase (TK) gene. MDBK cells infected with rSV5-TK showed a time-dependent loss of viability when infected cells were cultured in the presence of the prodrug acyclovir (ACV) or ganciclovir (GCV) while no significant toxicity was observed in the absence of prodrug. Cells infected with a control rSV5 expressing GFP and cultured with prodrug showed only a slight reduction in growth rate and little cell death. Time-lapse video microscopy of rSV5-TK-infected MDBK cells that were cultured in the presence of ACV showed an accumulation of cells with morphological effects characteristic of apoptotic cell death. An MDBK cell line persistently infected with rSV5-TK retained long-term expression of TK and sensitivity to prodrug-mediated cell killing that were comparable to those found in an acute infection. Titration experiments showed that the rSV5-TK plus GCV combination resulted in cell death for all mouse and human cell lines tested, although the kinetics and efficiency of cell death varied between cell types. Our results demonstrating controlled cell killing by a recombinant paramyxovirus support the use of negative-strand RNA viruses as therapeutic vectors for targeted killing of cancer cells.


Asunto(s)
Terapia Genética , Neoplasias/terapia , Respirovirus/genética , Timidina Quinasa/genética , Aciclovir/uso terapéutico , Animales , Muerte Celular , Línea Celular , Relación Dosis-Respuesta a Droga , Ganciclovir/uso terapéutico , Vectores Genéticos , Proteínas Recombinantes/uso terapéutico , Simplexvirus/enzimología , Timidina Quinasa/uso terapéutico , Proteína p53 Supresora de Tumor/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA