Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 92(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29514905

RESUMEN

Several viruses induce intestinal epithelial cell death during enteric infection. However, it is unclear whether proapoptotic capacity promotes or inhibits replication in this tissue. We infected mice with two reovirus strains that infect the intestine but differ in the capacity to alter immunological tolerance to new food antigen. Infection with reovirus strain T1L, which induces an inflammatory immune response to fed antigen, is prolonged in the intestine, whereas T3D-RV, which does not induce this response, is rapidly cleared from the intestine. Compared with T1L, T3D-RV infection triggered apoptosis of intestinal epithelial cells and subsequent sloughing of dead cells into the intestinal lumen. We conclude that the infection advantage of T1L derives from its capacity to subvert host restriction by epithelial cell apoptosis, providing a possible mechanism by which T1L enhances inflammatory signals during antigen feeding. Using a panel of T1L × T3D-RV reassortant viruses, we identified the viral M1 and M2 gene segments as determinants of reovirus-induced apoptosis in the intestine. Expression of the T1L M1 and M2 genes in a T3D-RV background was sufficient to limit epithelial cell apoptosis and enhance viral infection to levels displayed by T1L. These findings define additional reovirus gene segments required for enteric infection of mice and illuminate the antiviral effect of intestinal epithelial cell apoptosis in limiting enteric viral infection. Viral strain-specific differences in the capacity to infect the intestine may be useful in identifying viruses capable of ameliorating tolerance to fed antigen in autoimmune conditions like celiac disease.IMPORTANCE Acute viral infections are thought to be cleared by the host with few lasting consequences. However, there may be much broader and long-lasting effects of viruses on immune homeostasis. Infection with reovirus, a common, nonpathogenic virus, triggers inflammation against innocuous food antigens, implicating this virus in the development of celiac disease, an autoimmune intestinal disorder triggered by exposure to dietary gluten. Using two reovirus strains that differ in the capacity to abrogate oral tolerance, we found that strain-specific differences in the capacity to replicate in the intestine inversely correlate with the capacity to induce apoptotic death of intestinal epithelial cells, providing a host-mediated process to restrict intestinal infection. This work contributes new knowledge about virus-host interactions in the intestine and establishes a foundation for future studies to define mechanisms by which viruses break oral tolerance in celiac disease.


Asunto(s)
Apoptosis/inmunología , Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Orthoreovirus Mamífero 3/inmunología , Orthoreovirus de los Mamíferos/inmunología , Infecciones por Reoviridae/inmunología , Animales , Antígenos Virales/inmunología , Línea Celular , Cricetinae , Células Epiteliales/patología , Células Epiteliales/virología , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Ratones , Infecciones por Reoviridae/patología
2.
PLoS Pathog ; 11(4): e1004782, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849543

RESUMEN

Epithelial cells are a major port of entry for many viruses, but the molecular networks which protect barrier surfaces against viral infections are incompletely understood. Viral infections induce simultaneous production of type I (IFN-α/ß) and type III (IFN-λ) interferons. All nucleated cells are believed to respond to IFN-α/ß, whereas IFN-λ responses are largely confined to epithelial cells. We observed that intestinal epithelial cells, unlike hematopoietic cells of this organ, express only very low levels of functional IFN-α/ß receptors. Accordingly, after oral infection of IFN-α/ß receptor-deficient mice, human reovirus type 3 specifically infected cells in the lamina propria but, strikingly, did not productively replicate in gut epithelial cells. By contrast, reovirus replicated almost exclusively in gut epithelial cells of IFN-λ receptor-deficient mice, suggesting that the gut mucosa is equipped with a compartmentalized IFN system in which epithelial cells mainly respond to IFN-λ that they produce after viral infection, whereas other cells of the gut mostly rely on IFN-α/ß for antiviral defense. In suckling mice with IFN-λ receptor deficiency, reovirus replicated in the gut epithelium and additionally infected epithelial cells lining the bile ducts, indicating that infants may use IFN-λ for the control of virus infections in various epithelia-rich tissues. Thus, IFN-λ should be regarded as an autonomous virus defense system of the gut mucosa and other epithelial barriers that may have evolved to avoid unnecessarily frequent triggering of the IFN-α/ß system which would induce exacerbated inflammation.


Asunto(s)
Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Leucocitos/inmunología , Infecciones por Reoviridae/inmunología , Animales , Separación Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Interferón-alfa/inmunología , Interferón beta/inmunología , Interferón gamma/inmunología , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
3.
J Immunol ; 194(9): 4397-412, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825443

RESUMEN

Tumor-associated immunosuppression aids cancer cells to escape immune-mediated attack and subsequent elimination. Recently, however, many oncolytic viruses, including reovirus, have been reported to overturn such immunosuppression and promote the development of a clinically desired antitumor immunity, which is known to promote favorable patient outcomes. Contrary to this existing paradigm, in this article we demonstrate that reovirus augments tumor-associated immunosuppression immediately following its therapeutic administration. Our data show that reovirus induces preferential differentiation of highly suppressive CD11b(+), Gr-1(+), Ly6C(high) myeloid cells from bone marrow hematopoietic progenitor cells. Furthermore, reovirus administration in tumor-bearing hosts drives time-dependent recruitment of CD11b(+), Gr-1(+), Ly6C(high) myeloid cells in the tumor milieu, which is further supported by virus-induced increased expression of numerous immune factors involved in myeloid-derived suppressor cell survival and trafficking. Most importantly, CD11b(+), Gr-1(+), Ly6C(high) myeloid cells specifically potentiate the suppression of T cell proliferation and are associated with the absence of IFN-γ response in the tumor microenvironment early during oncotherapy. Considering that the qualitative traits of a specific antitumor immunity are largely dictated by the immunological events that precede its development, our findings are of critical importance and must be considered while devising complementary interventions aimed at promoting the optimum efficacy of oncolytic virus-based anticancer immunotherapies.


Asunto(s)
Vectores Genéticos , Inmunomodulación , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/inmunología , Virus Oncolíticos , Fenotipo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular , Quimiotaxis/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/inmunología , Ratones , Células Mieloides/citología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Receptores de Quimiocina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Microambiente Tumoral/inmunología
4.
Mol Ther ; 22(10): 1851-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24957982

RESUMEN

Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system. We show that preconditioning with immune modulators, or loading virus onto carrier cells ex vivo, enhances virus-mediated antitumor activity. Our early trials of systemic reovirus delivery showed that after infusion reovirus could be recovered from blood cells--but not from plasma--suggesting that rapid association with blood cells may protect virus from neutralizing antibody. We therefore postulated that stimulation of potential carrier cells directly in vivo before intravenous viral delivery would enhance delivery of cell-associated virus to tumor. We show that mobilization of the CD11b(+) cell compartment by granulocyte macrophage-colony stimulating factor immediately before intravenous reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning therapy was most effective in the presence of preexisting neutralizing antibody. Consistent with this, reovirus bound by neutralizing antibody effectively accessed monocytes/macrophages and was handed off to tumor cells. Thus, preconditioning with cytokine stimulated recipient cells in vivo for enhanced viral delivery to tumors. Moreover, preexisting neutralizing antibody to an oncolytic virus may, therefore, even be exploited for systemic delivery to tumors in the clinic.


Asunto(s)
Citocinas/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Virus Oncolíticos/genética , Transducción Genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales , Antígeno CD11b/metabolismo , Citocinas/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunidad/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Receptores Fc/genética , Receptores Fc/metabolismo , Carga Tumoral
5.
J Virol ; 87(6): 3447-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23302889

RESUMEN

Reovirus infection is a well-characterized experimental system for the study of viral pathogenesis and antiviral immunity within the central nervous system (CNS). We have previously shown that c-Jun N-terminal kinase (JNK) and the Fas death receptor each play a role in neuronal apoptosis occurring in reovirus-infected brains. Death-associated protein 6 (Daxx) is a cellular protein that mechanistically links Fas signaling to JNK signaling in several models of apoptosis. In the present study, we demonstrate that Daxx is upregulated in reovirus-infected brain tissue through a type I interferon-mediated mechanism. Daxx upregulation is limited to brain regions that undergo reovirus-induced apoptosis and occurs in the cytoplasm and nucleus of neurons. Cytoplasmic Daxx is present in Fas-expressing cells during reovirus encephalitis, suggesting a role for Daxx in Fas-mediated apoptosis following reovirus infection. Further, in vitro expression of a dominant negative form of Daxx (DN-Daxx), which binds to Fas but which does not transmit downstream signaling, inhibits apoptosis of reovirus-infected cells. In contrast, in vitro depletion of Daxx results in increased expression of caspase 3 and apoptosis, suggesting that Daxx plays an antiapoptotic role in the nucleus. Overall, these data imply a regulatory role for Daxx in reovirus-induced apoptosis, depending on its location in the nucleus or cytoplasm.


Asunto(s)
Apoptosis , Proteínas Portadoras/biosíntesis , Citoplasma/virología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Orthoreovirus Mamífero 3/patogenicidad , Neuronas/virología , Proteínas Nucleares/biosíntesis , Animales , Proteínas Co-Represoras , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares , Regulación hacia Arriba
6.
J Virol ; 86(15): 8107-18, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623770

RESUMEN

Reovirus infection of the murine spinal cord (SC) was used as a model system to investigate innate immune responses during viral myelitis, including the activation of glia (microglia and astrocytes) and interferon (IFN) signaling and increased expression of inflammatory mediators. Reovirus myelitis was associated with the pronounced activation of SC glia, as evidenced by characteristic changes in cellular morphology and increased expression of astrocyte and microglia-specific proteins. Expression of inflammatory mediators known to be released by activated glia, including interleukin-1ß (IL-1ß), tumor necrosis factor alpha (TNF-α), chemokine (C-C motif) ligand 5 (CCL 5), chemokine (C-X-C motif) ligand 10 (CXCL10), and gamma interferon (IFN-γ), was also significantly upregulated in the SC of reovirus-infected animals compared to mock-infected controls. Reovirus infection of the mouse SC was also associated with increased expression of genes involved in IFN signaling, including IFN-stimulated genes (ISG). Further, reovirus infection of mice deficient in the expression of the IFN-α/ß receptor (IFNAR(-/-)) resulted in accelerated mortality, demonstrating that IFN signaling is protective during reovirus myelitis. Experiments performed in ex vivo SC slice cultures (SCSC) confirmed that resident SC cells contribute to the production of at least some of these inflammatory mediators and ISG during reovirus infection. Microglia, but not astrocytes, were still activated, and glia-associated inflammatory mediators were still produced in reovirus-infected INFAR(-/-) mice, demonstrating that IFN signaling is not absolutely required for these neuroinflammatory responses. Our results suggest that activated glia and inflammatory mediators contribute to a local microenvironment that is deleterious to neuronal survival.


Asunto(s)
Inmunidad Innata , Orthoreovirus Mamífero 3/inmunología , Mielitis/inmunología , Infecciones por Reoviridae/inmunología , Transducción de Señal/inmunología , Médula Espinal/inmunología , Animales , Citocinas , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/virología , Mediadores de Inflamación/inmunología , Ratones , Ratones Noqueados , Mielitis/genética , Mielitis/patología , Mielitis/virología , Neuroglía/inmunología , Neuroglía/patología , Neuroglía/virología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/patología , Transducción de Señal/genética , Médula Espinal/patología , Médula Espinal/virología
7.
FASEB J ; 26(3): 1280-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22125314

RESUMEN

The trafficking of effector cells to sites of infection is crucial for antiviral responses. However, the mechanisms of recruitment of the interferon-γ-producing and cytotoxic CD56(+) T cells are poorly understood. Human mast cells are sentinel cells found in the skin and airway and produce selected proinflammatory mediators in response to multiple pathogen-associated signals. The role of human mast cell-derived chemokines in T-cell recruitment to virus infection was examined. Supernatants from primary human cord blood-derived mast cells (CBMCs) infected with mammalian reovirus were examined for chemokine production and utilized in chemotaxis assays. Virus-infected CBMCs produced several chemokines, including CCL3, CCL4, and CCL5. Supernatants from reovirus-infected CBMCs selectively induced the chemotaxis of CD8(+) T cells (10±1%) and CD3(+)CD56(+) T cells (19±5%). CD56(+) T-cell migration was inhibited by pertussis toxin (65±9%) and met-RANTES (56±7%), a CCR1/CCR5 antagonist. CD56(+) T cells expressed CCR5, but little CCR1. The depletion of CCL3, CCL4, and CCL5 from reovirus-infected CBMC supernatants significantly (41±10%) inhibited CD56(+) T-cell chemotaxis. This study demonstrates a novel role for mast cells and CCR5 in CD56(+) T-cell trafficking and suggests that human mast cells enhance immunity to viruses through the selective recruitment of cytotoxic effector cells to virus infection sites. These findings could be exploited to enhance local T-cell responses in chronic viral infection and malignancies at mast cell-rich sites.


Asunto(s)
Antígeno CD56/inmunología , Orthoreovirus Mamífero 3/inmunología , Mastocitos/inmunología , Linfocitos T/inmunología , Antígeno CD56/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Quimiocina CCL3/inmunología , Quimiocina CCL3/metabolismo , Quimiocina CCL3/farmacología , Quimiocina CCL4/inmunología , Quimiocina CCL4/metabolismo , Quimiocina CCL4/farmacología , Quimiocina CCL5/inmunología , Quimiocina CCL5/metabolismo , Quimiocina CCL5/farmacología , Quimiocinas/inmunología , Quimiocinas/metabolismo , Quimiocinas/farmacología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Sangre Fetal/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Patógeno/inmunología , Humanos , Ligandos , Orthoreovirus Mamífero 3/fisiología , Mastocitos/metabolismo , Mastocitos/virología , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Linfocitos T/metabolismo
8.
Methods ; 52(4): 301-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20801221

RESUMEN

An oncolytic virus is considered a targeted cancer therapy due to its ability to specifically target, replicate in and lyse cancer cells while leaving normal cells unharmed. Over the last few years several tumor selective oncolytic viruses have been developed. These include certain DNA viruses such as adenovirus that have been genetically manipulated to target specific cancer cells by exerting restrictions on the virus at the level of cell entry, viral gene transcription and viral protein translation. There are a variety of RNA viruses being studied as possible cancer therapies including reovirus. Reovirus is intrinsically oncolytic without the need for any genetic manipulation. The inherent oncolytic properties of this virus are derived from the fact that it specifically targets cells with an activated Ras pathway found in many cancer cells. REOLYSIN® is a proprietary formulation of human reovirus type 3 Dearing developed by Oncolytics Biotech Inc. and is the only therapeutic reovirus in clinical development. This review provides an overview of the development of REOLYSIN as a potential cancer therapeutic and the growing role of in situ based molecular pathology methods in providing clinical proof of concept and in guiding clinical development.


Asunto(s)
Inmunohistoquímica , Orthoreovirus Mamífero 3/genética , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Replicación Viral , Animales , Anticuerpos Antivirales/análisis , Ensayos Clínicos como Asunto , Humanos , Masculino , Orthoreovirus Mamífero 3/inmunología , Ratones , Neoplasias/virología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/virología
9.
J Exp Med ; 170(3): 887-900, 1989 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2549167

RESUMEN

The mammalian reoviruses provide a model for studying specific aspects of the immunopathogenesis of viral infection. We have used two serotype 3 reoviruses to define stages in the pathogenesis of central nervous system (CNS) infection at which a mAb specific for the reoviral cell attachment protein sigma 1 (sigma 1mAbG5) acts to protect mice against lethal disease. sigma 1mAbG5 administered either before or at the time of footpad inoculation with reovirus T3D prevented entry of T3D into the CNS. sigma 1mAbG5 also inhibited the spread of reovirus T3C9 from the gastrointestinal tract to the CNS after peroral inoculation with T3C9. These effects occurred in the absence of a significant effect of sigma 1mAbG5 on primary replication in skeletal muscle (T3D) or the gastrointestinal tract (T3C9). sigma 1mAbG5 administered after T3D had reached the spinal cord inhibited subsequent spread of infectious virus from spinal cord to brain. Even after direct intracerebral inoculation of T3D, sigma 1mAbG5 prevented both growth in the brain and spread of infectious virus from brain to eye, spinal cord, and muscle. Treatment with sigma 1mAbG5 after intracerebral inoculation with T3D prevented neuronal necrosis and resulted in a delayed and topographically restricted inflammatory response. We detected no antibody-resistant T3D variants in vivo after treatment with sigma 1mAbG5. We conclude that systemic IgG does not play a significant role at the primary site of infection with reoviruses, while it clearly acts to prevent infection of the CNS and extension of infection with the CNS. Further study will be directed to defining what components of the immune system do act at primary sites of infection, and to defining the mechanisms by which antibody acts at defined stages in pathogenesis.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Encefalopatías/inmunología , Orthoreovirus Mamífero 3/inmunología , Infecciones por Reoviridae/inmunología , Reoviridae/inmunología , Animales , Encéfalo/microbiología , Encéfalo/patología , Encefalopatías/etiología , Ratones , Músculos/microbiología , Necrosis , Infecciones por Reoviridae/etiología , Replicación Viral
10.
J Exp Med ; 155(1): 155-67, 1982 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-6172539

RESUMEN

A xenogeneic antiserum raised to antireovirus immunoglobulin was used to define an idiotypic determinant present on antibodies to reovirus type 3 hemagglutinin. The same idiotype was identified on nonimmune lymphoid cells and on neuronal cells that specifically bind the hemagglutinin of type 3 reovirus. This idiotypic determinant, called Id3, is shared by (a) a monoclonal antibody to the neutralization site of hemagglutinin from type 3 reovirus; (b) BALB/c serum antibodies to the hemagglutinin of reovirus type 3; (c) R1.1, a murine thymoma cell line that binds reovirus type 3; (d) primary cultures of murine neuronal cells. The presence of an idiotype shared by antihemagglutinin antibodies and by structures on nonlymphoid cells suggests a general relationship between disparate receptors that recognize a common determinant. Furthermore, this suggests a novel approach for the study of viral receptor interactions and for analysis of mechanisms of autoimmune responses.


Asunto(s)
Hemaglutininas/inmunología , Idiotipos de Inmunoglobulinas/inmunología , Linfocitos/inmunología , Reoviridae/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Epítopos , Ligamiento Genético , Hemaglutininas/genética , Sueros Inmunes/farmacología , Idiotipos de Inmunoglobulinas/genética , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos BALB C , Neuronas/inmunología , Conejos , Reoviridae/genética
11.
Int J Exp Pathol ; 91(3): 276-80, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20353422

RESUMEN

Soluble gp130 (sgp130) has been shown to suppress the inflammatory response of autoimmune pathologies; however, its effects on virus infection are not known. Here, we report that intraperitoneal treatment of mice with sgp130-Fc fusion protein at the time of oral reovirus serotype 3 infection resulted in altered morphopathological changes that were evident by less shortening of intestinal villi length and crypt depth after infection. That the effect mediated by sgp130 treatment was due to an increase in intestinal crypt cell proliferation was demonstrated by an increase in the number of crypt mitotic figures. This was further confirmed by increased immunoreactivity to the Cdc47 proliferation-associated antigen in crypts of sgp130-treated virus-infected mice compared to infected non-treated mice. These findings suggest that sgp130 may have a beneficial effect during intestinal virus infection by disrupting interleukin-6 trans-signalling, thereby reducing the local inflammatory response.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Infecciones por Reoviridae/patología , Animales , Receptor gp130 de Citocinas/inmunología , Femenino , Hiperplasia , Inflamación/inmunología , Inflamación/virología , Mucosa Intestinal/metabolismo , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología
12.
Science ; 220(4596): 505-7, 1983 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-6301010

RESUMEN

Variants of the Dearing strain of reovirus type 3 with antigenically altered hemagglutinin proteins are much less neurovirulent than the parental virus. When injected intracerebrally into mice these variants infected a subset of the brain neurons that were infected by the parental virus. When injected intraperitoneally, the variants did not spread to the brain. These results indicate that minor modifications of the reovirus hemagglutinin dramatically alter the ability of the virus to spread into and injure the central nervous system.


Asunto(s)
Encefalopatías/microbiología , Orthoreovirus Mamífero 3/patogenicidad , Reoviridae/patogenicidad , Animales , Anticuerpos Monoclonales/inmunología , Encéfalo/patología , Encefalopatías/patología , Pruebas de Hemaglutinación , Orthoreovirus Mamífero 3/inmunología , Ratones , Infecciones por Reoviridae/microbiología , Infecciones por Reoviridae/patología
13.
Science ; 201(4355): 529-31, 1978 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-208156

RESUMEN

Reovirus type 3, passaged in pancreatic beta-cell cultures, produced an insulitis when inoculated into 1- to 2-week-old mice. By means of a double-label antibody technique, in which we used fluorescein-labeled antibody to reovirus and rhodamine-labeled antibody to insulin, reovirus antigens were found in beta cells. By electron microscopy, viral particles in different stages of morphogenesis were observed in insulin-containing beta cells but not glucagon-containing alpha cells. The infection resulted in destruction of beta cells, reduction in the insulin content of the pancreas, and alteration in the host's capacity to respond normally to a glucose tolerance test.


Asunto(s)
Diabetes Mellitus Experimental/microbiología , Islotes Pancreáticos/microbiología , Infecciones por Reoviridae/complicaciones , Animales , Antígenos Virales/análisis , Células Cultivadas , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Orthoreovirus Mamífero 3/inmunología , Ratones , Replicación Viral
14.
Science ; 282(5392): 1332-4, 1998 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-9812900

RESUMEN

Human reovirus requires an activated Ras signaling pathway for infection of cultured cells. To investigate whether this property can be exploited for cancer therapy, severe combined immune deficient mice bearing tumors established from v-erbB-transformed murine NIH 3T3 cells or human U87 glioblastoma cells were treated with the virus. A single intratumoral injection of virus resulted in regression of tumors in 65 to 80 percent of the mice. Treatment of immune-competent C3H mice bearing tumors established from ras-transformed C3H-10T1/2 cells also resulted in tumor regression, although a series of injections were required. These results suggest that, with further work, reovirus may have applicability in the treatment of cancer.


Asunto(s)
Genes ras , Orthoreovirus Mamífero 3/fisiología , Neoplasias Experimentales/terapia , Proteínas ras/metabolismo , Células 3T3 , Animales , Anticuerpos Antivirales/inmunología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular Transformada , Genes erbB , Humanos , Masculino , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos C3H , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/virología , Transducción de Señal , Células Tumorales Cultivadas , Replicación Viral
15.
Clin Cancer Res ; 14(21): 7127-37, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18981012

RESUMEN

PURPOSE: To determine the safety and feasibility of daily i.v. administration of wild-type oncolytic reovirus (type 3 Dearing) to patients with advanced cancer, assess viral excretion kinetics and antiviral immune responses, identify tumor localization and replication, and describe antitumor activity. EXPERIMENTAL DESIGN: Patients received escalating doses of reovirus up to 3 x 10(10) TCID(50) for 5 consecutive days every 4 weeks. Viral excretion was assessed by reverse transcription-PCR and antibody response by cytotoxicity neutralization assay. Pretreatment and post-treatment tumor biopsies were obtained to measure viral uptake and replication. RESULTS: Thirty-three patients received 76 courses of reovirus from 1 x 10(8) for 1 day up to 3 x 10(10) TCID(50) for 5 days, repeated every four weeks. Dose-limiting toxicity was not seen. Common grade 1 to 2 toxicities included fever, fatigue, and headache, which were dose and cycle independent. Viral excretion at day 15 was not detected by reverse transcription-PCR at 25 cycles and only in 5 patients at 35 cycles. Neutralizing antibodies were detected in all patients and peaked at 4 weeks. Viral localization and replication in tumor biopsies were confirmed in 3 patients. Antitumor activity was seen by radiologic and tumor marker (carcinoembryonic antigen, CA19.9, and prostate-specific antigen) evaluation. CONCLUSIONS: Oncolytic reovirus can be safely and repeatedly administered by i.v. injection at doses up to 3 x 10(10) TCID(50) for 5 days every 4 weeks without evidence of severe toxicities. Productive reoviral infection of metastatic tumor deposits was confirmed. Reovirus is a safe agent that warrants further evaluation in phase II studies.


Asunto(s)
Orthoreovirus Mamífero 3 , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/análisis , Estudios de Factibilidad , Femenino , Humanos , Inyecciones Intravenosas , Masculino , Orthoreovirus Mamífero 3/inmunología , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/inmunología , Viroterapia Oncolítica/efectos adversos
16.
Clin Cancer Res ; 14(22): 7358-66, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010851

RESUMEN

PURPOSE: Early clinical trials are under way exploring the direct oncolytic potential of reovirus. This study addresses whether tumor infection by reovirus is also able to generate bystander, adaptive antitumor immunity. EXPERIMENTAL DESIGN: Reovirus was delivered intravenously to C57BL/6 mice bearing lymph node metastases from the murine melanoma, B16-tk, with assessment of nodal metastatic clearance, priming of antitumor immunity against the tumor-associated antigen tyrosinase-related protein-2, and cytokine responses. In an in vitro human system, the effect of reovirus infection on the ability of Mel888 melanoma cells to activate and load dendritic cells for cytotoxic lymphocyte (CTL) priming was investigated. RESULTS: In the murine model, a single intravenous dose of reovirus reduced metastatic lymph node burden and induced antitumor immunity (splenocyte response to tyrosinase-related protein-2 and interleukin-12 production in disaggregated lymph nodes). In vitro human assays revealed that uninfected Mel888 cells failed to induce dendritic cell maturation or support priming of an anti-Mel888 CTL response. In contrast, reovirus-infected Mel888 cells (reo-Mel) matured dendritic cells in a reovirus dose-dependent manner. When cultured with autologous peripheral blood lymphocytes, dendritic cells loaded with reo-Mel induced lymphocyte expansion, IFN-gamma production, specific anti-Mel888 cell cytotoxicity, and cross-primed CD8+ T cells specific against the human tumor-associated antigen MART-1. CONCLUSION: Reovirus infection of tumor cells reduces metastatic disease burden and primes antitumor immunity. Future clinical trials should be designed to explore both direct cytotoxic and immunotherapeutic effects of reovirus.


Asunto(s)
Antineoplásicos/uso terapéutico , Citotoxicidad Inmunológica/inmunología , Neoplasias Experimentales/terapia , Viroterapia Oncolítica/métodos , Infecciones por Reoviridae/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interferón gamma/biosíntesis , Oxidorreductasas Intramoleculares/inmunología , Metástasis Linfática/patología , Activación de Linfocitos/inmunología , Antígeno MART-1 , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/virología , Reacción en Cadena de la Polimerasa , Linfocitos T Citotóxicos/inmunología
17.
Gene Ther ; 15(12): 911-20, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18323793

RESUMEN

There is an emerging realization from animal models that the immune response may have both detrimental and beneficial therapeutic effects during cancer virotherapy. However, there is a dearth of clinical data on the immune response to viral agents in patients. During a recently completed phase I trial of intravenous reovirus type 3 Dearing (RT3D), heavily pretreated patients with advanced cancers received RT3D at doses escalating from 1 x 10(8) tissue culture infectious dose-50 (TCID(50)) on day 1 to 3 x 10(10) TCID(50) on 5 consecutive days of a 4 weekly cycle. A detailed analysis of the immune effects was conducted by collecting serial clinical samples for analysis of neutralizing anti-reoviral antibodies (NARA), peripheral blood mononuclear cells (PBMC) and cytokines. Significant increases in NARA were seen with peak endpoint titres >1/10 000 in all but one patient. The median fold increase was 250, with a range of 9-6437. PBMC subset analysis showed marked heterogeneity. At baseline, CD3+CD4+ T cells were reduced in most patients, but after RT3D therapy their numbers increased in 47.6% of patients. In contrast, most patients had high baseline CD3+CD8+ T-cell levels, with 33% showing incremental increases after therapy. In some patients, there was increased cytotoxic T-cell activation post-therapy, as shown by increased CD8+perforin/granzyme+ T-cell numbers. Most patients had high numbers of circulating CD3-CD56+ NK cells before therapy and in 28.6% this increased with treatment. Regulatory (CD3+CD4+CD25+) T cells were largely unaffected by the therapy. Combined Th1 and Th2 cytokine expression increased in 38% of patients. These data confirm that even heavily pretreated patients are capable of mounting dynamic immune responses during treatment with RT3D, although these responses are not clearly related to the administered virus dose. These data will provide the basis for future studies aiming to modulate the immune response during virotherapy.


Asunto(s)
Terapia Genética/métodos , Orthoreovirus Mamífero 3/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Citocinas/sangre , Femenino , Humanos , Inmunidad Innata , Inyecciones Intravenosas , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Subgrupos de Linfocitos T/inmunología , Resultado del Tratamiento
18.
Lung Cancer ; 120: 142-148, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29748010

RESUMEN

OBJECTIVES: Pelareorep (reolysin), a Dearing strain of reovirus serotype 3, has demonstrated oncolytic activity as single agent and synergy with chemotherapy. We evaluated pelareorep, combined with standard second-line chemotherapy in patients with non-small cell lung cancer (NSCLC). MATERIALS AND METHODS: This randomized phase II trial enrolled patients with advanced or metastatic NSCLC after first line chemotherapy. After a safety run-in, patients were randomized 1:1 to chemotherapy (pemetrexed [500 mg/m2, non-squamous], or docetaxel [75 mg/m2], day 1 every 21 days]) +/- pelareorep (4.5 × 1010 TCID50, days 1-3 every 21 days), stratified by EGFR mutation status. The primary outcome was progression free survival (PFS) of patients randomized to chemotherapy + pelareorep vs. chemotherapy alone. Secondary outcomes included overall survival, objective response rate and exploratory translational analyses. RESULTS: Between October 2012 and August 2015, 166 patients were enrolled (14 to the safety run in). Pelareorep did not improve the PFS vs. single agent chemotherapy (median PFS 3.0 months, 95% confidence interval [CI] 2.6-4.1) vs. 2.8 months (95% CI 2.5-4.0), hazard ratio (HR) 0.90 (95% CI 0.65-1.25), P = 0.53). Neither KRAS or EGFR mutation was associated with improved PFS, but STK11 mutations did appear to have an association with improved PFS (HR 0.29 [0.12-0.67); as did PIK3CA mutation (HR 0.45 [0.22-0.93]). The combination was tolerable, although associated with increased rates of neutropenic fever. CONCLUSION: The addition of pelareorep to second-line chemotherapy did not improve the PFS of patients with NSCLC. The three-day pelareorep schedule was tolerable. Further research is needed to evaluate the potential benefit in molecular subtypes of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Orthoreovirus Mamífero 3/inmunología , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Infecciones por Reoviridae/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Canadá , Terapia Combinada , Docetaxel/uso terapéutico , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pemetrexed/uso terapéutico , Recurrencia , Terapia Recuperativa , Adulto Joven
19.
J Clin Invest ; 72(2): 617-21, 1983 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6308058

RESUMEN

Specific cellular and host tropism is a characteristic property of many viruses mediated by the interaction of viral attachment proteins with components of the plasma membrane of the cell. We have studied the binding of virus to cells quantitatively by using type 3 reovirus labeled with 125I and GH4C1 pituitary cells in culture. Binding was rapid at both 4 degrees and 15 degrees C and was stable over a 9-h period. Unlabeled virus inhibited binding of the labeled virus in a dose-dependent manner. Scatchard analysis revealed 4,200 viral binding sites/cell with an apparent affinity of 1.2 X 10(-11) M. Also, binding of type 3 reovirus was inhibited by antibodies directed against the viral hemagglutinin and partially inhibited by type 2 reovirus, but was unaffected by type 1 reovirus or a variety of other ligands that bind to receptors on GH4C1 cells. These data indicate that reovirus binds to a high affinity, specific receptor on target cells, which may control its tropism and ultimate disease expression.


Asunto(s)
Receptores Virales/análisis , Infecciones por Reoviridae/microbiología , Animales , Anticuerpos Monoclonales/fisiología , Anticuerpos Antivirales/fisiología , Unión Competitiva , Línea Celular , Hemaglutininas Virales/inmunología , Cinética , Orthoreovirus Mamífero 3/inmunología , Orthoreovirus Mamífero 3/metabolismo , Ratones , Ratas , Infecciones por Reoviridae/inmunología
20.
J Clin Invest ; 90(6): 2536-42, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1281838

RESUMEN

The recognition of cellular receptors by the mammalian reoviruses is an important determinant of cell and tissue tropism exhibited by reovirus strains of different serotypes. To extend our knowledge of the role of reovirus-receptor interactions in reovirus tropism, we determined whether type 1 and type 3 reovirus strains can infect cells derived from erythrocyte precursors. We found that reovirus type 3 Dearing (T3D), but not type 1 Lang, can grow in murine erythroleukemia (MEL) cells. This difference in growth was investigated by using reassortant viruses and we found that the capacity of T3D to infect MEL cells is determined by the viral cell-attachment protein, sigma 1. In experiments using murine monoclonal antibodies (mAbs) that bind to different sigma 1 regions, we show that T3D binding to MEL cells is inhibited by a mAb that identifies a domain important for hemagglutination (HA). We also determined that type 3 strains that can infect murine L cells but do not produce HA do not infect MEL cells. These results suggest that type 3 reovirus binds to and infects erythrocyte precursor cells via a sigma 1 domain important for HA. Moreover, this study suggests that different domains of some viral cell-attachment proteins are used to initiate productive infections of different types of cells.


Asunto(s)
Proteínas de la Cápside , Hemaglutinación , Leucemia Eritroblástica Aguda/metabolismo , Orthoreovirus Mamífero 3/metabolismo , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Epítopos , Técnicas In Vitro , Células L , Orthoreovirus Mamífero 3/crecimiento & desarrollo , Orthoreovirus Mamífero 3/inmunología , Ratones , Células Tumorales Cultivadas , Proteínas Virales/inmunología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA