Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunity ; 41(6): 960-72, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526309

RESUMEN

Granzyme B (GzmB) is a protease with a well-characterized intracellular role in targeted destruction of compromised cells by cytotoxic lymphocytes. However, GzmB also cleaves extracellular matrix components, suggesting that it influences the interplay between cytotoxic lymphocytes and their environment. Here, we show that GzmB-null effector T cells and natural killer (NK) cells exhibited a cell-autonomous homing deficit in mouse models of inflammation and Ectromelia virus infection. Intravital imaging of effector T cells in inflamed cremaster muscle venules revealed that GzmB-null cells adhered normally to the vessel wall and could extend lamellipodia through it but did not cross it efficiently. In vitro migration assays showed that active GzmB was released from migrating cytotoxic lymphocytes and enabled chemokine-driven movement through basement membranes. Finally, proteomic analysis demonstrated that GzmB cleaved basement membrane constituents. Our results highlight an important role for GzmB in expediting cytotoxic lymphocyte diapedesis via basement membrane remodeling.


Asunto(s)
Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/inmunología , Granzimas/metabolismo , Células Asesinas Naturales/fisiología , Linfocitos T Citotóxicos/fisiología , Animales , Membrana Basal/metabolismo , Movimiento Celular/genética , Células Cultivadas , Quimiocinas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Granzimas/genética , Células Asesinas Naturales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , Linfocitos T Citotóxicos/virología , Migración Transendotelial y Transepitelial/genética
2.
Immunol Cell Biol ; 95(10): 884-894, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28722018

RESUMEN

Intracellular serpins are proposed to inactivate proteases released from lysosome-related organelles into the host cell interior, preventing cell death. Serpinb9 opposes the immune cytotoxic protease, granzyme B, and in a number of settings protects cells against granzyme B-mediated cell death. Using a knockout mouse line engineered to express green fluorescent protein under the serpbinb9 promoter, we demonstrate that serpinb9 is vital for host survival during Ectromelia virus infection by maintaining both mature natural killer NK) cells, and activated CD8+ T cells. Serpinb9 expression parallels granzyme B expression within both populations during infection. Maturing serpinb9-null NK cells exhibit higher levels of granzyme B-mediated apoptosis during infection; hence there are fewer mature NK cells, and these cells also have lower cytotoxic potential. Thus the serpinb9-granzyme B axis is important for homeostasis of both major cytotoxic effector cell populations.


Asunto(s)
Granzimas/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/farmacología , Infecciones por Poxviridae/inmunología , Poxviridae/inmunología , Serpinas/farmacología , Animales , Muerte Celular , Supervivencia Celular , Homeostasis , Humanos , Espacio Intracelular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
J Virol ; 85(21): 11170-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21849445

RESUMEN

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Asunto(s)
Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/mortalidad , Interacciones Huésped-Patógeno , Células Asesinas Naturales/inmunología , Serpinas/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Virus de la Ectromelia/genética , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/virología , Eliminación de Gen , Interferón gamma/metabolismo , Interleucina-18/metabolismo , Hígado/virología , Subgrupos Linfocitarios/química , Subgrupos Linfocitarios/inmunología , Ratones , Serpinas/genética , Bazo/patología , Análisis de Supervivencia , Carga Viral , Proteínas Virales/genética , Replicación Viral
4.
J Cell Biol ; 176(4): 425-33, 2007 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-17283185

RESUMEN

Cathepsin C activates serine proteases expressed in hematopoietic cells by cleaving an N-terminal dipeptide from the proenzyme upon granule packaging. The lymphocytes of cathepsin C-null mice are therefore proposed to totally lack granzyme B activity and perforin-dependent cytotoxicity. Surprisingly, we show, using live cell microscopy and other methodologies, that cells targeted by allogenic CD8(+) cytotoxic T lymphocyte (CTL) raised in cathepsin C-null mice die through perforin-dependent apoptosis indistinguishable from that induced by wild-type CTL. The cathepsin C-null CTL expressed reduced but still appreciable granzyme B activity, but minimal granzyme A activity. Also, in contrast to mice with inactivation of both their granzyme A/B genes, cathepsin C deficiency did not confer susceptibility to ectromelia virus infection in vivo. Overall, our results indicate that although cathepsin C clearly generates the majority of granzyme B activity, some is still generated in its absence, pointing to alternative mechanisms for granzyme B processing and activation. Cathepsin C deficiency also results in considerably milder immune deficiency than perforin or granzyme A/B deficiency.


Asunto(s)
Apoptosis/fisiología , Catepsina C/genética , Activación Enzimática/inmunología , Granzimas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Linfocitos T Citotóxicos/enzimología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Virus de la Ectromelia/inmunología , Activación Enzimática/genética , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Perforina , Linfocitos T Citotóxicos/inmunología
5.
J Virol ; 84(9): 4212-21, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20164231

RESUMEN

We previously demonstrated that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A virus can provide robust protection in mice against both homologous and heterosubtypic challenges, including challenge with an H5N1 avian virus strain. We investigated the mechanism behind the observed cross-protection to define which arms of the adaptive immune response are involved in mediating this protection. Studies with gene knockout mice showed the cross-protective immunity to be mediated mainly by T cells and to be dependent on the cytolytic effector molecule perforin. Adoptive transfer of memory T cells from immunized mice, but not of memory B cells, protected naïve recipients against lethal heterosubtypic influenza virus challenge. Furthermore, gamma-irradiated influenza viruses induced cross-reactive Tc-cell responses but not cross-neutralizing or cross-protective antibodies. In addition, histological analysis showed reduced lung inflammation in vaccinated mice compared to that in unvaccinated controls following heterosubtypic challenge. This reduced inflammation was associated with enhanced early recruitment of T cells, both CD4(+) and CD8(+), and with early influenza virus-specific cytotoxic T-cell responses. Therefore, cross-protective immunity induced by vaccination with gamma-irradiated influenza A virus is mediated mainly by Tc-cell responses.


Asunto(s)
Protección Cruzada , Rayos gamma , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antivirales/sangre , Peso Corporal , Subtipo H1N1 del Virus de la Influenza A/efectos de la radiación , Subtipo H3N2 del Virus de la Influenza A/efectos de la radiación , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia , Vacunas de Productos Inactivados/inmunología
6.
J Immunol ; 183(1): 37-40, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19525394

RESUMEN

Cytotoxic T (Tc) cells lyse target cells via exocytosis of granules containing perforin (perf) and granzymes (gzm). In vitro, gzm delivery into the target cell cytosol results in apoptosis, and in the absence of gzm A and B the induction of apoptosis is severely impaired. However, using in vivo Tc cell killing assays, we find that virus-immune, gzm A x B-deficient (gzmAxB(-/-)) mice are competent to eliminate adoptively transferred target cells pulsed with an immunodominant Tc cell determinant as rapidly and completely as their wild-type counterparts. Specific target cell elimination occurred with similar kinetics in both spleen and lymph nodes. Thus, neither gzmA nor gzmB are required for rapid and efficient in vivo cytotoxicity by Tc cells.


Asunto(s)
Citotoxicidad Inmunológica , Granzimas/fisiología , Linfocitos T Citotóxicos/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Citotoxicidad Inmunológica/genética , Virus de la Ectromelia/inmunología , Ectromelia Infecciosa/enzimología , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/patología , Granzimas/deficiencia , Granzimas/genética , Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Linfocitos T Citotóxicos/patología , Linfocitos T Citotóxicos/virología , Factores de Tiempo
7.
J Exp Med ; 195(11): 1433-44, 2002 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-12045241

RESUMEN

Interleukin (IL)-5 and IL-13 are thought to play key roles in the pathogenesis of asthma. Although both cytokines use eotaxin to regulate eosinophilia, IL-13 is thought to operate a separate pathway to IL-5 to induce airways hyperreactivity (AHR) in the allergic lung. However, identification of the key pathway(s) used by IL-5 and IL-13 in the disease process is confounded by the failure of anti-IL-5 or anti-IL-13 treatments to completely inhibit the accumulation of eosinophils in lung tissue. By using mice deficient in both IL-5 and eotaxin (IL-5/eotaxin(-/-)) we have abolished tissue eosinophilia and the induction of AHR in the allergic lung. Notably, in mice deficient in IL-5/eotaxin the ability of CD4(+) T helper cell (Th)2 lymphocytes to produce IL-13, a critical regulator of airways smooth muscle constriction and obstruction, was significantly impaired. Moreover, the transfer of eosinophils to IL-5/eotaxin(-/-) mice overcame the intrinsic defect in T cell IL-13 production. Thus, factors produced by eosinophils may either directly or indirectly modulate the production of IL-13 during Th2 cell development. Our data show that IL-5 and eotaxin intrinsically modulate IL-13 production from Th2 cells and that these signaling systems are not necessarily independent effector pathways and may also be integrated to regulate aspects of allergic disease.


Asunto(s)
Asma/complicaciones , Hiperreactividad Bronquial/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Quimiocinas CC/metabolismo , Eosinofilia/metabolismo , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Traslado Adoptivo , Animales , Asma/inmunología , Asma/metabolismo , Hiperreactividad Bronquial/complicaciones , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Linfocitos T CD4-Positivos/inmunología , Quimiocina CCL11 , Quimiocinas CC/sangre , Quimiocinas CC/genética , Modelos Animales de Enfermedad , Eosinofilia/complicaciones , Eosinofilia/inmunología , Eosinofilia/patología , Eosinófilos/metabolismo , Eosinófilos/trasplante , Eliminación de Gen , Humanos , Interleucina-13/biosíntesis , Interleucina-18/metabolismo , Interleucina-5/sangre , Interleucina-5/genética , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Esputo/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
8.
J Gen Virol ; 91(Pt 6): 1450-60, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20147516

RESUMEN

We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.


Asunto(s)
Protección Cruzada , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Inactivación de Virus , Animales , Peso Corporal , Femenino , Formaldehído/toxicidad , Rayos gamma , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de la radiación , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Análisis de Supervivencia , Linfocitos T Citotóxicos/inmunología , Rayos Ultravioleta , Vacunas de Productos Inactivados/inmunología , Carga Viral
9.
Virus Res ; 228: 61-65, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27865865

RESUMEN

Mousepox is caused by the orthopoxvirus ectromelia virus (ECTV), and is thought to be transmitted via skin abrasions. We studied the ECTV virulence factor N1 following subcutaneous infection of mousepox-susceptible BALB/c mice. In this model, ECTV lacking N1L gene was attenuated more than 1000-fold compared with wild-type virus and replication was profoundly reduced as early as four days after infection. However, in contrast to data from an intranasal model, N1 protein was not required for virus dissemination. Further, neither T cell nor cytokine responses were enhanced in the absence of N1. Together with the early timing of reduced virus titres, this suggests that in a cutaneous model, N1 exerts its function at the level of infected cells or in the inhibition of the very earliest effectors of innate immunity.


Asunto(s)
Virus de la Ectromelia/fisiología , Ectromelia Infecciosa/virología , Proteínas Virales/genética , Animales , Interacciones Huésped-Patógeno , Ratones , Carga Viral , Proteínas Virales/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Replicación Viral
10.
J Leukoc Biol ; 75(6): 1001-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15020648

RESUMEN

Eosinophil degranulation is thought to play a pivotal role in the pathogenesis of allergic disorders. Although mouse models of allergic disorders have been used extensively to identify the contribution of eosinophils to disease, ultrastructural evidence of active granule disassembly has not been reported. In this investigation, we characterized the degree of eosinophil activation in the bone marrow, blood, lung tissue, and airways lumen [bronchoalveolar lavage fluid (BALF)] of ovalbumin-sensitized and aero-challenged wild-type and interleukin-5 transgenic mice. Degranulation was most prominent in and primarily compartmentalized to the airways lumen. Eosinophils released granule proteins by the process of piecemeal degranulation (PMD). Accordingly, recruitment and activation of eosinophils in the lung correlated with the detection of cell-free eosinophil peroxidase in BALF and with the induction of airways hyper-reactivity. As in previous studies with human eosinophils, degranulation of isolated mouse cells did not occur until after adherence to extracellular matrix. However, higher concentrations of exogenous stimuli appear to be required to trigger adherence and degranulation (piecemeal) of mouse eosinophils when compared with values reported for studies of human eosinophils. Thus, mouse eosinophils undergo PMD during allergic inflammation, and in turn, this process may contribute to pathogenesis. However, the degranulation process in the allergic lung of mice is primarily compartmentalized to the airway lumen. Understanding the mechanism of eosinophil degranulation in the airway lumen may provide important insights into how this process occurs in human respiratory diseases.


Asunto(s)
Degranulación de la Célula/fisiología , Gránulos Citoplasmáticos/metabolismo , Eosinófilos/fisiología , Pulmón/inmunología , Hipersensibilidad Respiratoria/fisiopatología , Mucosa Respiratoria/fisiología , Animales , Médula Ósea/química , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Adhesión Celular , Peroxidasa del Eosinófilo , Eosinófilos/efectos de los fármacos , Eosinófilos/ultraestructura , Matriz Extracelular , Femenino , Humanos , Interleucina-5/genética , Interleucina-5/fisiología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/farmacología , Peroxidasas/metabolismo
11.
PLoS One ; 8(6): e68458, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840854

RESUMEN

MHCII molecules are heterodimeric cell surface proteins composed of an α and ß chain. These molecules are almost exclusively expressed on thymic epithelium and antigen presenting cells (APCs) and play a central role in the development and function of CD4 T cells. Various MHC-II knockout mice have been generated including MHC-IIAα(-/-) (I-Aα(-/-)), MHC-IIAß(-/-) (I-ß(-/-)) and the double knockout (I-Aαxß(-/-)). Here we report a very striking observation, namely that alphaviruses including the avirulent strain of Semliki Forest virus (aSFV), which causes asymptomatic infection in wild-type C57BL6/J (B6) mice, causes a very acute and lethal infection in I-Aα(-/-), but not in I-ß(-/-) or I-Aαxß(-/-), mice. This susceptibility to aSFV is associated with high virus titres in muscle, spleen, liver, and brain compared to B6 mice. In addition, I-Aα(-/-) mice show intact IFN-I responses in terms of IFN-I serum levels and IFN-I receptor expression and function. Radiation bone marrow chimeras of B6 mice reconstituted with I-Aα(-/-) bone marrow expressed B6 phenotype, whereas radiation chimeras of I-Aα(-/-) mice reconstituted with B6 bone marrow expressed the phenotype of high viral susceptibility. Virus replication experiments both in vivo and in vitro showed enhanced virus growth in tissues and cell cultures derived form I-Aα(-/-) compared to B6 mice. This enhanced virus replication is evident for other alpha-, flavi- and poxviruses and may be of great benefit to producers of viral vaccines. In conclusion, I-Aα(-/-) mice exhibit a striking susceptibility to virus infections independent of their defective MHC-II expression. Detailed genetic analysis will be carried out to characterise the underlining genetic defects responsible for the observed phenomenon.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Síndromes de Inmunodeficiencia/inmunología , Replicación Viral/inmunología , Animales , Médula Ósea/inmunología , Médula Ósea/virología , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de los Bosques Semliki/inmunología , Células Vero , Replicación Viral/genética
12.
PLoS One ; 6(10): e25765, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21998693

RESUMEN

BACKGROUND: We have shown previously in mice, that infection with live viruses, including influenza/A and Semliki Forest virus (SFV), induces systemic partial activation of lymphocytes, characterized by cell surface expression of CD69 and CD86, but not CD25. This partial lymphocytes activation is mediated by type-I interferons (IFN-I). Importantly, we have shown that γ-irradiated SFV does not induce IFN-I and the associated lymphocyte activation. PRINCIPAL FINDINGS: Here we report that, in contrast to SFV, γ-irradiated influenza A virus elicits partial lymphocyte activation in vivo. Furthermore, we show that when using influenza viruses inactivated by a variety of methods (UV, ionising radiation and formalin treatment), as well as commercially available influenza vaccines, only γ-irradiated influenza virus is able to trigger IFN-I-dependent partial lymphocyte activation in the absence of the TLR7/MyD88 signalling pathways. CONCLUSIONS: Our data suggest an important mechanism for the recognition of γ-irradiated influenza vaccine by cytosolic receptors, which correspond with the ability of γ-irradiated influenza virus to induce cross-reactive and cross-protective cytotoxic T cell responses.


Asunto(s)
Rayos gamma , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H1N1 del Virus de la Influenza A/efectos de la radiación , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/efectos de la radiación , Interferón Tipo I/metabolismo , Linfocitos/virología , Animales , Línea Celular , Cricetinae , Perros , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Interferón Tipo I/biosíntesis , Linfocitos/citología , Linfocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Neuraminidasa/metabolismo , Transducción de Señal , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología , Receptor Toll-Like 7/metabolismo , Activación Viral/efectos de la radiación
13.
PLoS One ; 4(10): e7512, 2009 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-19838298

RESUMEN

BACKGROUND: Ectromelia virus is a natural mouse pathogen, causing mousepox. The cytotoxic T (Tc) cell granule serine-protease, granzyme B, is important for its control, but the underlying mechanism is unknown. Using ex vivo virus immune Tc cells, we have previously shown that granzyme B is able to activate several independent pro-apoptotic pathways, including those mediated by Bid/Bak/Bax and caspases-3/-7, in target cells pulsed with Tc cell determinants. METHODS AND FINDINGS: Here we analysed the physiological relevance of those pro-apoptotic pathways in ectromelia infection, by incubating ectromelia-immune ex vivo Tc cells from granzyme A deficient (GzmB(+) Tc cells) or granzyme A and granzyme B deficient (GzmAxB(-/-) Tc cell) mice with ectromelia-infected target cells. We found that gzmB-induced apoptosis was totally blocked in ectromelia infected or peptide pulsed cells lacking caspases-3/-7. However ectromelia inhibited only partially apoptosis in cells deficient for Bid/Bak/Bax and not at all when both pathways were operative suggesting that the virus is able to interfere with apoptosis induced by gzmB in case not all pathways are activated. Importantly, inhibition of viral replication in vitro, as seen with wild type cells, was not affected by the lack of Bid/Bak/Bax but was significantly reduced in caspase-3/-7-deficient cells. Both caspase dependent processes were strictly dependent on gzmB, since Tc cells, lacking both gzms, neither induced apoptosis nor reduced viral titers. SIGNIFICANCE: Out findings present the first evidence on the biological importance of the independent gzmB-inducible pro-apoptotic pathways in a physiological relevant virus infection model.


Asunto(s)
Caspasas/metabolismo , Virus de la Ectromelia/genética , Ectromelia Infecciosa/virología , Fibroblastos/metabolismo , Granzimas/metabolismo , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología , Animales , Apoptosis , Calreticulina/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína X Asociada a bcl-2/metabolismo
14.
PLoS One ; 4(4): e5336, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19401775

RESUMEN

BACKGROUND: Influenza A (flu) virus causes significant morbidity and mortality worldwide, and current vaccines require annual updating to protect against the rapidly arising antigenic variations due to antigenic shift and drift. In fact, current subunit or split flu vaccines rely exclusively on antibody responses for protection and do not induce cytotoxic T (Tc) cell responses, which are broadly cross-reactive between virus strains. We have previously reported that gamma-ray inactivated flu virus can induce cross-reactive Tc cell responses. METHODOLOGY/PRINCIPAL FINDING: Here, we report that intranasal administration of purified gamma-ray inactivated human influenza A virus preparations (gamma-Flu) effectively induces heterotypic and cross-protective immunity. A single intranasal administration of gamma-A/PR8[H1N1] protects mice against lethal H5N1 and other heterotypic infections. CONCLUSIONS/SIGNIFICANCE: Intranasal gamma-Flu represents a unique approach for a cross-protective vaccine against both seasonal as well as possible future pandemic influenza A virus infections.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Gripe Humana/prevención & control , Administración Intranasal , Animales , Reacciones Cruzadas , Femenino , Rayos gamma , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , ARN Viral/análisis , ARN Viral/genética , Linfocitos T Citotóxicos/inmunología , Vacunación/métodos , Vacunas de Productos Inactivados/administración & dosificación
15.
J Gen Virol ; 89(Pt 8): 1942-1944, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18632965

RESUMEN

Previously, we have shown that mice defective in granule exocytosis and/or Fas.L/Fas-mediated cytolytic pathways are significantly more resistant to alphavirus, Semliki Forest virus (SFV), infection compared with wild-type mice. Here, we evaluated SFV replication in different tissues of mice defective in both cytolytic pathways (perf(-/-)xgld) relative to that in wild-type counterparts and found that viral replication in perf(-/-)xgld mice is remarkably restricted. Although the mechanism responsible for this observation is yet to be established, the lower virus titres found in these mice indicate that the role of cytolytic effector molecules in antiviral immunity needs to be re-evaluated.


Asunto(s)
Infecciones por Alphavirus/inmunología , Proteína Ligando Fas/deficiencia , Perforina/deficiencia , Virus de los Bosques Semliki/fisiología , Virus de los Bosques Semliki/patogenicidad , Replicación Viral , Infecciones por Alphavirus/virología , Animales , Proteína Ligando Fas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Perforina/genética
16.
Antimicrob Agents Chemother ; 51(8): 2965-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17562808

RESUMEN

Gemfibrozil, an agent that inhibits production of proinflammatory cytokines in addition to its clinically useful lipid-lowering activity, increased survival in BALB/c mice that were already ill from infection by influenza virus A/Japan/305/57 (H2N2). Gemfibrozil was administered intraperitoneally once daily from days 4 to 10 after intranasal exposure to the virus. Survival increased from 26% in vehicle-treated mice (n = 50) to 52% in mice given gemfibrozil at 60 mg/kg/day (n = 46) (P = 0.0026). If this principle translates to patients, a drug already approved for human use, albeit by a different route for another purpose, might be adapted relatively fast for use against influenza, conceivably including human infection with a derivative of the avian H5N1 strain.


Asunto(s)
Antiinflamatorios/uso terapéutico , Gemfibrozilo/uso terapéutico , Hipolipemiantes/uso terapéutico , Subtipo H2N2 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/mortalidad , Animales , Antiinflamatorios/administración & dosificación , Gemfibrozilo/administración & dosificación , Hipolipemiantes/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/mortalidad , Inflamación/virología , Subtipo H2N2 del Virus de la Influenza A/patogenicidad , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Resultado del Tratamiento
17.
J Gen Virol ; 87(Pt 12): 3599-3609, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17098975

RESUMEN

The flavivirus West Nile virus (WNV) can cause fatal encephalitis in humans and mice. It has recently been demonstrated, in an experimental model using WNV strain Sarafend and C57BL/6 mice, that both virus- and immune-mediated pathology is involved in WNV encephalitis, with CD8(+) T cells being the dominant subpopulation of lymphocyte infiltrates in the brain. Here, the role of activated WNV-immune CD8(+) T cells in mouse WNV encephalitis was investigated further. Passive transfer of WNV-immune CD8(+) T cells reduced mortality significantly and prolonged survival times of mice infected with WNV. Early infiltration of WNV-immune CD8(+) T cells into infected brains is shown, suggesting a beneficial contribution of these lymphocytes to recovery from encephalitis. This antiviral function was not markedly mediated by gamma interferon (IFN-gamma), as a deficiency in IFN-gamma did not affect mortality to two strains of WNV (Sarafend and Kunjin) or brain virus titres significantly. The cytolytic potential, as well as precursor frequency, of WNV-immune CD8(+) T cells were not altered by the absence of IFN-gamma. This was reflected in transfer experiments of WNV-immune CD8(+) T cells from IFN-gamma(-/-) mice into WNV-infected wild-type mice, which showed that IFN-gamma-deficient T cells were as effective as those from WNV-immune wild-type mice in ameliorating disease outcome. It is speculated here that one of the pleiotropic functions of IFN-gamma is mimicked by WNV-Sarafend-mediated upregulation of cell-surface expression of major histocompatibility complex antigens, which may explain the lack of phenotype of IFN-gamma(-/-) mice in response to WNV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Traslado Adoptivo , Animales , Encéfalo/patología , Encéfalo/virología , Pruebas Inmunológicas de Citotoxicidad , Modelos Animales de Enfermedad , Interferón gamma/genética , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología
18.
J Immunol ; 177(5): 3235-41, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920963

RESUMEN

Viral infections often cause a period of heightened susceptibility to a secondary infection but the cause of this phenomenon is unknown. We found that a primary viral infection in mice rapidly triggers an IFN-I-dependent partial activation state in the majority of B and T lymphocytes, which reverts to a resting phenotype within 5 days. When a secondary infection with an unrelated virus occurred 5 to 9 days after the primary infection, no recurrence of marked activation of lymphocytes was observed. This was not due to an inherent inability of the previously activated cells to undergo renewed partial activation, because they responded when challenged with virus after transfer into "naive" recipients. Instead, the failure to respond optimally resided in the original host's incapacity to mount an IFN-I response to the secondary infection during this time period. Thus, transient immunosuppression through exhaustion of IFN-I production during an acute viral infection creates a time period of enhanced susceptibility to secondary infection.


Asunto(s)
Interferón Tipo I/biosíntesis , Virosis/inmunología , Virosis/metabolismo , Enfermedad Aguda , Adenoviridae/fisiología , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/metabolismo , Infecciones por Alphavirus/virología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Biomarcadores , Línea Celular , Chlorocebus aethiops , Cricetinae , Células Dendríticas/citología , Células Dendríticas/inmunología , Virus de la Ectromelia/patogenicidad , Ectromelia Infecciosa/inmunología , Ectromelia Infecciosa/virología , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Virus de los Bosques Semliki/fisiología , Linfocitos T/citología , Linfocitos T/inmunología , Virosis/virología
19.
Eur J Immunol ; 36(4): 887-96, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16541469

RESUMEN

Natural killer (NK) and cytotoxic T (Tc) cells are prime effector populations in the antiviral response of the host. Tc cells are essential for recovery from many viral diseases but may also be responsible for immunopathology. The role of NK cells in recovery from viral infections is less well established. We have studied acute virulent Semliki Forest virus (vSFV) infection of the central nervous system in C57BL/6J mice, which was mainly controlled by NK cells without marked Tc cell involvement. We show that mice with defects in the Fas and/or granule exocytosis pathways of cytotoxicity are more resistant to lethal vSFV infection than wild-type mice. On the other hand, mice defective in the IFN-gamma response are more sensitive than wild-type mice, whereas mice lacking the Tc cell compartment (beta-2 microglobulin-deficient mice) exhibit susceptibility similar to wild-type mice. The additional finding that depletion of NK cells significantly delayed the mean time to death but did not prevent mortality in SFV-infected B6 mice suggests that cytolytic activity of NK cells is detrimental, while IFN-gamma production is beneficial for recovery from SFV infection. This is the first study illustrating an NK cell-mediated immunopathological outcome to an acute viral infection.


Asunto(s)
Infecciones por Alphavirus/inmunología , Enfermedades del Sistema Nervioso Central/virología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Animales , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/patología , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Virus de los Bosques Semliki/inmunología , Linfocitos T Citotóxicos/inmunología
20.
J Immunol ; 175(7): 4635-40, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16177109

RESUMEN

The vast majority of both T and B cells in mice were found to up-regulate cell surface expression of the early activation markers CD69 and CD86, but not CD25, within 24 h of infection with Semliki Forest virus. Kinetics and magnitude of activation marker expression was dependent on live virus, dose, and correlated with strain virulence. Activation marker expression declined to baseline levels over the next 96 h. This very early "activation" of such a high percentage of lymphocytes required the presence of type I IFN receptor genes, was inducible with poly(I:C), and correlated with IFN-I levels in serum. We conclude that virus-induced IFN-I release systemically affects most of the hosts T and B cells by triggering them rapidly and independently of Ag-reactivity into a semiactivated state.


Asunto(s)
Infecciones por Alphavirus/inmunología , Interferón Tipo I/fisiología , Activación de Linfocitos/inmunología , Linfocitos/fisiología , Infecciones por Alphavirus/sangre , Infecciones por Alphavirus/genética , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Biomarcadores , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Citometría de Flujo , Interferón Tipo I/sangre , Cinética , Lectinas Tipo C , Activación de Linfocitos/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de los Bosques Semliki/inmunología , Bazo/citología , Bazo/metabolismo , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA