Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Med Microbiol Immunol ; 204(3): 355-66, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25805565

RESUMEN

A murine model of B and T cell lymphomas in recipients after hematoablative conditioning for hematopoietic cell transplantation (HCT) has previously revealed a tumor-repressive, metastasis-inhibiting function of murine cytomegalovirus (mCMV). More recently, this prediction from the experimental model was put on trial in several clinical studies that indeed gave evidence for a lower incidence of tumor relapse associated with early reactivation of latent human cytomegalovirus (hCMV) after allogeneic HCT in patients treated against different types of hematopoietic malignancies, including lymphoma and acute as well as chronic leukemias. Due to the limitations inherent to clinical studies, the tumor-repressive role of hCMV remained observational with no approach to clarify mechanisms. Although the tumor-repressive mechanisms of mCMV and hCMV may differ and depend on the type of tumor, experimental approaches in the murine model might give valuable hints for concepts to follow in clinical research. We have previously shown for the liver-adapted A20-derived B cell lymphoma E12E that mCMV does not infect the lymphoma cells for causing cell death by viral cytopathogenicity but triggers tumor-selective apoptosis at a tissue site of tumor metastasis distant from a local site of infection. This finding suggested involvement of a cytokine that triggers apoptosis, directly or indirectly. Here we used a series of differential high-density microarray analyses to identify cellular genes whose expression is specifically upregulated at the site of virus entry only by viruses capable of triggering lymphoma cell apoptosis. This strategy identified interleukin-15 (IL-15) as most promising candidate, eventually confirmed by lymphoma repression with recombinant IL-15.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Interleucina-15/metabolismo , Linfoma/inmunología , Linfoma/patología , Muromegalovirus/inmunología , Aloinjertos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Humanos , Interleucina-15/genética , Interleucina-15/farmacología , Subunidad alfa del Receptor de Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Linfoma/genética , Linfoma/mortalidad , Linfoma/virología , Ratones , Acondicionamiento Pretrasplante , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Carga Tumoral/inmunología , Irradiación Corporal Total
2.
J Virol ; 84(12): 6254-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375164

RESUMEN

Major immediate-early transcriptional enhancers are genetic control elements that act, through docking with host transcription factors, as a decisive regulatory unit for efficient initiation of the productive virus cycle. Animal models are required for studying the function of enhancers paradigmatically in host organs. Here, we have sought to quantitatively assess the establishment, maintenance, and level of in vivo growth of enhancerless mutants of murine cytomegalovirus in comparison with those of an enhancer-bearing counterpart in models of the immunocompromised or immunologically immature host. Evidence is presented showing that enhancerless viruses are capable of forming restricted foci of infection but fail to grow exponentially.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/crecimiento & desarrollo , Elementos de Facilitación Genéticos , Animales , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos BALB C , Ratones SCID
3.
Med Microbiol Immunol ; 197(2): 167-78, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18309517

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is a promising therapeutic option against hematopoietic malignancies. Infection with cytomegalovirus (CMV) and tumor relapse are complications that limit the success of HSCT. In theory, CMV infection can facilitate tumor relapse and growth by inhibiting "graft take" and reconstitution of the immune system or by inducing the secretion of tumor cell growth-promoting cytokines. Conversely, one can also envisage an anti-tumoral effect of CMV by cytopathic/oncolytic infection of tumor cells, by inducing the secretion of death ligands for tumor cell apoptosis, and by the activation of systemic innate and adaptive immunity. Here we will briefly review the current knowledge about tumor control in a murine model of CMV infection and liver-adapted B cell lymphoma, with a focus on a putative implication of CD49(+)NKG2D(+) hepatic natural killer cells.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Linfoma/inmunología , Animales , Infecciones por Citomegalovirus/complicaciones , Huésped Inmunocomprometido , Linfoma/complicaciones , Ratones , Ratones Endogámicos BALB C
4.
J Virol ; 80(10): 4801-19, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641273

RESUMEN

Cytomegalovirus (CMV) poses a threat to the therapy of hematopoietic malignancies by hematopoietic stem cell transplantation, but efficient reconstitution of antiviral immunity prevents CMV organ disease. Tumor relapse originating from a minimal residual leukemia poses another threat. Although a combination of risk factors was supposed to enhance the incidence and severity of transplantation-associated disease, a murine model of a liver-adapted B-cell lymphoma has previously shown a survival benefit and tumor growth inhibition by nonlethal subcutaneous infection with murine CMV. Here we have investigated the underlying antitumoral mechanism. Virus replication proved to be required, since inactivated virions or the highly attenuated enhancerless mutant mCMV-DeltaMIEenh did not impact the lymphoma in the liver. Surprisingly, the dissemination-deficient mutant mCMV-DeltaM36 inhibited tumor growth, even though this virus fails to infect the liver. On the other hand, various strains of herpes simplex viruses consistently failed to control the lymphoma, even though they infect the liver. A quantitative analysis of the tumor growth kinetics identified a transient tumor remission by apoptosis as the antitumoral effector mechanism. Tumor cell colonies with cells surviving the CMV-induced "apoptotic crisis" lead to tumor relapse even in the presence of full-blown tissue infection. Serial transfer of surviving tumor cells did not indicate a selection of apoptosis-resistant genetic variants. NK cell activity of CD49b-expressing cells failed to control the lymphoma upon adoptive transfer. We propose the existence of an innate antitumoral mechanism that is triggered by CMV infection and involves an apoptotic signal effective at a distant site of tumor growth.


Asunto(s)
Apoptosis , Hígado/patología , Linfoma/patología , Muromegalovirus/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Hígado/virología , Linfoma/virología , Linfoma de Células B/mortalidad , Linfoma de Células B/patología , Linfoma de Células T/mortalidad , Linfoma de Células T/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Muromegalovirus/genética , Células 3T3 NIH , Mutación Puntual
5.
J Virol ; 76(6): 2857-70, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11861853

RESUMEN

Tumor relapse and cytomegalovirus (CMV) infection are major concerns in the therapy of hematopoietic malignancies by bone marrow transplantation (BMT). Little attention so far has been given to a possible pathogenetic interplay between CMV and lymphomas. CMV inhibits stem cell engraftment and hematopoietic reconstitution. Thus, by causing maintenance of bone marrow aplasia and immunodeficiency, CMV could promote tumor relapse. Alternatively, CMV could aid tumor remission. One might think of cytopathogenic infection of tumor cells, induction of apoptosis or inhibitory cytokines, interference with tumor cell extravasation or tumor vascularization, or bystander stimulation of an antitumoral immune response. To approach these questions, the established model of experimental BMT and murine CMV infection was extended by the introduction of liver-infiltrating, highly tumorigenic variant clone E12E of BALB/c-derived B-cell lymphoma A20. We document a remarkable retardation of lymphoma progression. First-guess explanations were ruled out: (i) lymphoma cells were not infected; (ii) lymphoma cells located next to infected hepatocytes did not express executioner caspase 3 but were viable and proliferated; (iii) an inhibitory effect of virus on the formation of tumor nodules in the liver became apparent by day 7 after BMT, long before the reconstitution of immune cells; and (iv) recombinant tumor necrosis factor alpha (TNF-alpha) did not substitute for virus; accordingly anti-TNF-alpha did not prevent the inhibition. Notably, while the antitumoral effect required replicative virus, prevention of cytopathogenic infection of the liver by antiviral CD8 T cells did not abolish lymphoma control. These findings are paradigmatic for a novel virus-associated antitumoral mechanism distinct from oncolysis.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Infecciones por Citomegalovirus/inmunología , Linfoma de Células B/prevención & control , Muromegalovirus/inmunología , Animales , Modelos Animales de Enfermedad , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA