Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(3): e59633, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527234

RESUMEN

General cellular functions of proteasomes occur through protein degradation, whereas the specific function of immunoproteasomes is the optimization of antigen processing associated with MHC class I. We and others previously reported that deficiency in subunits of immunoproteasomes impaired the activation of antigen-specific CD8(+) T cells, resulting in higher susceptibility to tumor and infections. We demonstrated that CD8(+) T cells contributed to protection against malaria parasites. In this study, we evaluated the role of immunoproteasomes in the course of infection with rodent malaria parasites. Unexpectedly, Plasmodium yoelii infection of mice deficient in LMP7, a catalytic subunit of immunoproteasomes, showed lower parasite growth in the early phase of infection and lower lethality compared with control mice. The protective characteristics of LMP7-deficient mice were not associated with enhanced immune responses, as the mutant mice showed comparable or diminished activation of innate and acquired immunity. The remarkable difference was observed in erythrocytes instead of immune responses. Parasitized red blood cells (pRBCs) purified from LMP7-deficient mice were more susceptible to phagocytosis by macrophages compared with those from wild-type mice. The susceptibility of pRBC to phagocytosis appeared to correlate with deformity of the membrane structures that were only observed after infection. Our results suggest that RBCs of LMP7-deficient mice were more likely to deform in response to infection with malaria parasites, presumably resulting in higher susceptibility to phagocytosis and in the partial resistance to malaria.


Asunto(s)
Resistencia a la Enfermedad/fisiología , Eritrocitos/parasitología , Malaria/fisiopatología , Fagocitosis/fisiología , Plasmodium yoelii , Complejo de la Endopetidasa Proteasomal/fisiología , Animales , Cartilla de ADN/genética , Eritrocitos/ultraestructura , Citometría de Flujo , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Complejo de la Endopetidasa Proteasomal/deficiencia , Complejo de la Endopetidasa Proteasomal/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
J Immunol ; 189(4): 1618-26, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22802414

RESUMEN

Aquaporin-1 (AQP-1) is a water channel protein highly expressed in the vascular endothelial cells of proliferating tissues including malignant cancers. Given that in APC ubiquitinated peptides are effectively introduced into proteasomes from which CD8 epitopes are excised, we fused ubiquitin with AQP-1 (pUB-AQP-1) to produce a DNA vaccine. In C57BL/6J mice immunized with pUB-AQP-1, the growth of B16F10 melanoma was profoundly inhibited. The antitumor effect of the pUB-AQP-1 DNA vaccine was largely mediated by CD8 T cells, which secrete IFN-γ, perforin, and granzyme-B in the presence of APCs transfected with pUB-AQP-1. AQP-1-specific CD8 T cells possessed cytotoxic activity both in vivo and in vitro. After tumor challenge, the microvessel density decreased and the ratio of total blood vessel area to tumor area was significantly reduced as compared with control mice, resulting in a dramatic suppression of tumor growth. The immunization effect was completely abrogated in immunoproteasome-deficient mice. Strikingly this pUB-AQP-1 DNA vaccine was also effective against Colon 26 colon tumors (BALB/c) and MBT/2 bladder tumors (C3H/HeN). Thus, this ubiquitin-conjugated DNA immunization-targeting tumor vasculature is a valid and promising antitumor therapy. This vaccine works across the barriers of tumor species and MHC class I differences in host mice.


Asunto(s)
Acuaporina 1/inmunología , Vacunas contra el Cáncer/farmacología , Neoplasias Experimentales/terapia , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/farmacología , Animales , Western Blotting , Vacunas contra el Cáncer/inmunología , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/inmunología , Neovascularización Patológica/terapia , Complejo de la Endopetidasa Proteasomal/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ubiquitina/inmunología , Vacunas de ADN/inmunología
3.
Korean J Parasitol ; 49(1): 85-90, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21461275

RESUMEN

Relatively little has been studied on the AMA-1 vaccine against Plasmodium vivax and on the plasmid DNA vaccine encoding P. vivax AMA-1 (PvAMA-1). In the present study, a plasmid DNA vaccine encoding AMA-1 of the reemerging Korean P. vivax has been constructed and a preliminary study was done on its cellular immunogenicity to recipient BALB/c mice. The PvAMA-1 gene was cloned and expressed in the plasmid vector UBpcAMA-1, and a protein band of approximately 56.8 kDa was obtained from the transfected COS7 cells. BALB/c mice were immunized intramuscularly or using a gene gun 4 times with the vaccine, and the proportions of splenic T-cell subsets were examined by fluorocytometry at week 2 after the last injection. The spleen cells from intramuscularly injected mice revealed no significant changes in the proportions of CD8(+) T-cells and CD4(+) T-cells. However, in mice immunized using a gene gun, significantly higher (P<0.05) proportions of CD8(+) cells were observed compared to UB vector-injected control mice. The results indicated that cellular immunogenicity of the plasmid DNA vaccine encoding AMA-1 of the reemerging Korean P. vivax was weak when it was injected intramuscularly; however, a promising effect was observed using the gene gun injection technique.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Malaria Vivax/inmunología , Proteínas de la Membrana/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Vacunas Antiprotozoos/inmunología , Vacunas de ADN/inmunología , Animales , Antígenos de Protozoos/administración & dosificación , Antígenos de Protozoos/genética , Células COS , Chlorocebus aethiops , Humanos , Activación de Linfocitos , Malaria Vivax/parasitología , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium vivax/genética , Proteínas Protozoarias/administración & dosificación , Proteínas Protozoarias/genética , Vacunas Antiprotozoos/administración & dosificación , Vacunas Antiprotozoos/genética , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
4.
Eur J Immunol ; 40(4): 1053-61, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20101613

RESUMEN

When developing malaria vaccines, the most crucial step is to elucidate the mechanisms involved in protective immunity against the parasites. We found that CD8(+) T cells contribute to protective immunity against infection with blood-stage parasites of Plasmodium yoelii. Infection of C57BL/6 mice with P. yoelii 17XL was lethal, while all mice infected with a low-virulence strain of the parasite 17XNL acquired complete resistance against re-infection with P. yoelii 17XL. However, the host mice transferred with CD8(+) T cells from mice primed only with P. yoelii 17XNL failed to acquire protective immunity. On the other hand, the irradiated host mice were completely resistant to P. yoelii 17XL infection, showing no grade of parasitemia when adoptively transferred with CD8(+) T cells from immune mice that survived infection with both P. yoelii XNL and, subsequently, P. yoelii 17XL. These protective CD8(+) T cells from immune WT mice had the potential to generate IFN-gamma, perforin (PFN) and granzyme B. When mice deficient in IFN-gamma were used as donor mice for CD8(+) T cells, protective immunity in the host mice was fully abrogated, and the immunity was profoundly attenuated in PFN-deficient mice. Thus, CD8(+) T cells producing IFN-gamma and PFN appear to be involved in protective immunity against infection with blood-stage malaria.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Malaria/inmunología , Parasitemia/inmunología , Plasmodium yoelii/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Convalecencia , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Granzimas/biosíntesis , Interferón gamma/biosíntesis , Interferón gamma/deficiencia , Interferón gamma/genética , Macrófagos/inmunología , Malaria/prevención & control , Vacunas contra la Malaria/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parasitemia/prevención & control , Plasmodium yoelii/patogenicidad , Proteínas Citotóxicas Formadoras de Poros/biosíntesis , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Proteínas Citotóxicas Formadoras de Poros/genética , Quimera por Radiación , Virulencia
5.
Biochem Biophys Res Commun ; 392(3): 277-82, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20059980

RESUMEN

Cytotoxic CD8(+) T cells are particularly important to the development of protective immunity against the intracellular protozoan parasite, Trypanosoma cruzi, the etiological agent of Chagas disease. We have developed a new effective strategy of genetic immunization by activating CD8(+) T cells through the ubiquitin-fusion degradation (UFD) pathway. We constructed expression plasmids encoding the amastigote surface protein-2 (ASP-2) of T. cruzi. To induce the UFD pathway, a chimeric gene encoding ubiquitin fused to ASP-2 (pUB-ASP-2) was constructed. Mice immunized with pUB-ASP-2 presented lower parasitemia and longer survival period, compared with mice immunized with pASP-2 alone. Depletion of CD8(+) T cells abolished protection against T. cruzi in mice immunized with pUB-ASP-2 while depletion of CD4(+) T cells did not influence the effective immunity. Mice deficient in LMP2 or LMP7, subunits of immunoproteasomes, were not able to develop protective immunity induced. These results suggest that ubiquitin-fused antigens expressed in antigen-presenting cells were effectively degraded via the UFD pathway, and subsequently activated CD8(+) T cells. Consequently, immunization with pUB-ASP-2 was able to induce potent protective immunity against infection of T. cruzi.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad de Chagas/prevención & control , Neuraminidasa/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Vacunas Antiprotozoos/inmunología , Trypanosoma cruzi/inmunología , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Antígenos/inmunología , Antígenos/metabolismo , Enfermedad de Chagas/inmunología , Cisteína Endopeptidasas/genética , Femenino , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Complejos Multienzimáticos/genética , Neuraminidasa/genética , Neuraminidasa/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Vacunas Antiprotozoos/genética , Vacunas Antiprotozoos/metabolismo , Ubiquitina/metabolismo , Vacunación , Vacunas de ADN/genética , Vacunas de ADN/metabolismo
6.
Eur J Immunol ; 39(12): 3385-94, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19830724

RESUMEN

Proteasome-mediated proteolysis is responsible for the generation of immunogenic epitopes presented by MHC class I molecules, which activate antigen-specific CD8+ T cells. Immunoproteasomes, defined by the presence of the three catalytic subunits LMP2, MECL-1, and LMP7, have been hypothesized to optimize MHC class I antigen processing. In this study, we demonstrate that the infection of mice with a protozoan parasite, Toxoplasma gondii, induced the expression of LMP7 mRNA in APC and increased the capacity of APC to induce the production of IFN-gamma by antigen-specific CD8+ T cells. In vitro infection of a DC cell line with T. gondii also induced the expression of LMP7 and resulted in enhanced proteasome proteolytic activity. Finally, mice lacking LMP7 were highly susceptible to infection with T. gondii and showed a reduced number of functional CD8+ T cells. These results demonstrate that proteasomes containing LMP7 play an indispensable role in the survival of mice infected with T. gondii, presumably due to the efficient generation of CTL epitopes required for the functional development of CD8+ T cells.


Asunto(s)
Complejos Multienzimáticos/genética , Toxoplasma/inmunología , Toxoplasmosis/genética , Toxoplasmosis/inmunología , Animales , Western Blotting , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/parasitología , Células Cultivadas , Cisteína Endopeptidasas/biosíntesis , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/parasitología , Inducción Enzimática , Expresión Génica , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/biosíntesis , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Toxoplasma/fisiología , Toxoplasmosis/parasitología
7.
Eur J Immunol ; 39(10): 2822-30, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19728313

RESUMEN

Malaria and intestinal nematode infection are widespread and co-infection frequently occurs. We investigated whether co-infected intestinal nematodes modulate immunity against co-existing malaria parasites. Infection of C57BL/6 mice with Plasmodium yoelii 17XNL (Py) was transient and self-limiting, but preceding infection with Heligmosomoides polygyrus (Hp), a mouse intestinal nematode, exacerbated malaria resulting in higher parasite burdens and poor survival of the mice. Co-infection with Hp led to reduced Py-responsive proliferation and IFN-gamma production of spleen cells, and higher activation of CD4(+)CD25(+)Foxp3(+) Treg. In vivo depletion of Treg recovered anti-Py immunity and rescued co-infected mice from exacerbated malaria. However, we did not observe any obvious ex vivo activation of Treg by either Hp products or living worms. Our results suggest that intestinal nematodes moderate host immune responses during acute malaria infection by aggressive activation of Treg. Elucidation of the mechanisms of Treg activation in situ is a target for future analyses.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Malaria/inmunología , Nematospiroides dubius/inmunología , Plasmodium yoelii/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos Helmínticos/inmunología , Recuento de Células , Concanavalina A/farmacología , Células Dendríticas/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Tolerancia Inmunológica/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Depleción Linfocítica , Malaria/complicaciones , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Parasitemia , Bazo/citología , Bazo/inmunología , Infecciones por Strongylida/complicaciones , Análisis de Supervivencia , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
8.
Vaccine ; 27(44): 6154-9, 2009 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-19712768

RESUMEN

Chagas' disease, caused by infection with the protozoan parasite Trypanosoma cruzi (T. cruzi), is intractable showing a high mortality rate, and the development of effective vaccines is much desired. To examine the efficacy of a new mode of recombinant viral vaccine, we constructed two non-transmissible Sendai viruses (rSeV/dF) encoding the full-length parasite antigen amastigote surface protein-2 (ASP2) or ASP2 fused with a mono-ubiquitin on its N-terminus (UASP2). C57BL/6 mice immunized intranasally with rSeV/dF expressing either ASP2 or UASP2 showed significantly suppressed parasitemia and could be protected from lethal T. cruzi challenge. Depletion of CD8(+) T cells around the time of infection with T. cruzi completely abolished this protection, confirming that acquired immunity against the infection of T. cruzi is dependent on CD8(+) T cells. We also demonstrated that the protective immunity correlated with higher secretion of interferon-gamma (IFN-gamma) by spleen cells on in vitro-specific or non-specific stimulation. Increased CTL activity was also confirmed by degranulation or CTL assays. Interestingly, the control virus, rSeV/dF-GFP, induced even a higher IFN-gamma production from spleen cells following non-specific but not specific stimulation in vitro, suggesting that SeV may also be a good adjuvant when used as a vaccine vehicle. Taking together, the current findings indicate that recombinant Sendai virus expressing the ASP2 or UASP2 antigens of T. cruzi are interesting candidates for the development of a new mode of recombinant viral vaccine against Chagas' disease.


Asunto(s)
Enfermedad de Chagas/prevención & control , Neuraminidasa/inmunología , Vacunas Antiprotozoos/inmunología , Trypanosoma cruzi/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Enfermedad de Chagas/inmunología , Femenino , Interferón gamma/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Vacunas Antiprotozoos/genética , Virus Sendai/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
9.
Am J Trop Med Hyg ; 79(6): 819-22, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19052285

RESUMEN

We investigated whether concurrent infection with Heligmosomoides polygyrus, an intestinal nematode, modulated anti-malaria parasite immunity and development of experimental cerebral malaria (ECM) in mice. The C57BL/6 mice infected with Plasmodium berghei ANKA showed typical symptoms of ECM. Interestingly, preceding H. polygyrus infection did not alter ECM development, despite accelerated P. berghei growth in vivo. Our observation provides a new insight that ECM can be induced in a fashion independent of the immune responses affected by concurrent H. polygyrus. Differentiation between protective immunity and infection-associated host-damaging inflammatory response is urgently required for understanding the pathogenesis of cerebral malaria.


Asunto(s)
Malaria Cerebral/complicaciones , Malaria Cerebral/inmunología , Nematospiroides dubius , Plasmodium berghei , Infecciones por Strongylida/complicaciones , Animales , Heces/parasitología , Ratones , Ratones Endogámicos C57BL , Recuento de Huevos de Parásitos , Parasitemia , Factores de Tiempo
10.
Microbes Infect ; 10(3): 241-50, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18321749

RESUMEN

Acquired immunity against infection with Trypanosoma cruzi is dependent on CD8(+)T cells. Here, to develop a vaccine strategy taking advantage of activated CD8(+)T cells, we constructed a DNA vaccine, designated pGFP-TSA1, encoding a fusion protein linking GFP to a single CTL epitope of TSA1, a leading candidate for vaccine against T. cruzi. C57BL/6 mice vaccinated with this plasmid showed suppressed parasitemia and prolonged survival. Vaccination with pGFP-TSA1 enhanced epitope-specific cytotoxicity and IFN-gamma secretion by CD8(+)T cells. Furthermore, the depletion of CD8(+)T cells prior to challenge infection with T. cruzi completely abolished this protection, indicating that CD8(+)T cells are the principal effector T cells involved. When mice deficient in the proteasome activator PA28alpha/beta or the immunoproteasome subunits LMP2 and LMP7 were used, the protective immunity against infection was profoundly attenuated. Our findings clearly demonstrate that vaccination with pGFP-TSA1 successfully induces protection dependent on CD8(+)T cell activation, in which immunoproteasomes play a crucial role. It is noteworthy to document that physical binding of the epitope and GFP is required for induction of this protection, since mice vaccinated with pTSA1-IRES-GFP failed to acquire resistance, probably because the epitope and GFP are separately expressed in the antigen-presenting cells.


Asunto(s)
Enfermedad de Chagas/prevención & control , Cisteína Endopeptidasas/inmunología , Vacunas Antiprotozoos/administración & dosificación , Trypanosoma cruzi/inmunología , Vacunación , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Esquemas de Inmunización , Inyecciones Intradérmicas , Interferón gamma/biosíntesis , Activación de Linfocitos , Complejo Mayor de Histocompatibilidad/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Complejos Multienzimáticos/inmunología , Plásmidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Subgrupos de Linfocitos T , Linfocitos T Citotóxicos , Vacunas de ADN/administración & dosificación
11.
J Immunol ; 180(4): 2496-503, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18250459

RESUMEN

Malaria is still a life-threatening infectious disease that continues to produce 2 million deaths annually. Malaria parasites have acquired immune escape mechanisms and prevent the development of sterile immunity. Regulatory T cells (Tregs) have been reported to contribute to immune evasion during malaria in mice and humans, suggesting that activating Tregs is one of the mechanisms by which malaria parasites subvert host immune systems. However, little is known about how these parasites activate Tregs. We herein show that TLR9 signaling to dendritic cells (DCs) is crucial for activation of Tregs. Infection of mice with the rodent malaria parasite Plasmodium yoelii activates Tregs, leading to enhancement of their suppressive function. In vitro activation of Tregs requires the interaction of DCs with parasites in a TLR9-dependent manner. Furthermore, TLR9(-/-) mice are partially resistant to lethal infection, and this is associated with impaired activation of Tregs and subsequent development of effector T cells. Thus, malaria parasites require TLR9 to activate Tregs for immune escape.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Activación de Linfocitos/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/parasitología , Receptor Toll-Like 9/fisiología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/parasitología , Malaria/metabolismo , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasmodium yoelii/patogenicidad , Transducción de Señal/genética , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética
12.
Biochem Biophys Res Commun ; 365(4): 621-7, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17999919

RESUMEN

The ubiquitin-proteasome system (UPS) plays an indispensable role in inducing MHC class I-restricted CD8(+) T cells. In this study, we exploited UPS to induce CD8(+) T cells specific for mycobacterial HSP65 (mHSP65), one of the leading vaccine candidates against infection with Mycobacterium tuberculosis. A chimeric DNA termed pU-HSP65 encoding a fusion protein between murine ubiquitin and mHSP65 was constructed, and C57BL/6 (B6) mice were immunized with the DNA using gene gun bombardment. Mice immunized with the chimeric DNA acquired potent resistance against challenge with the syngeneic B16F1 melanoma cells transfected with the mHSP65 gene (HSP65/B16F1), compared with those immunized with DNA encoding only mHSP65. Splenocytes from the former group of mice showed a higher grade of cytotoxic activity against HSP65/B16F1 cells and contained a larger number of granzyme B- or IFN-gamma-producing CD8(+) T cells compared with those from the latter group of mice.


Asunto(s)
Proteínas Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Chaperoninas/inmunología , Mycobacterium tuberculosis/fisiología , Transducción de Señal/inmunología , Ubiquitina/inmunología , Animales , Células Cultivadas , Chaperonina 60 , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/inmunología , Ubiquitina/genética
13.
Biochem Biophys Res Commun ; 365(4): 758-63, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18029260

RESUMEN

We have developed a DNA vaccine encoding a fusion protein of ubiquitin (Ub) and target proteins at the N-terminus for effective induction of antigen-specific CD8(+) T cells. A series of expression plasmids encoding a model antigen, ovalbumin (OVA), fused with mutated Ub, was constructed. Western blotting analyses using COS7 cells transfected with these plasmids revealed that there were three types of amino acid causing different binding capacities between Ub and OVA. Natural Ub with a C-terminal glycine readily dissociated from OVA; on the other hand, artificially mutated Ub, the C-terminal amino acid of which had been exchanged to valine or arginine, stably united with the polypeptide, while Ub with a C-terminal alanine partially dissociated. The ability of DNA vaccination to induce OVA-specific CD8(+) T cells closely correlated with the stability of Ub fusion to OVA. Our strategy could be used to optimize the effect of genetic vaccines on the induction of CD8(+) T cells.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Ubiquitina/genética , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Ratones , Proteínas Recombinantes de Fusión/metabolismo
15.
Nat Immunol ; 8(10): 1067-75, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17767160

RESUMEN

The lineage commitment of CD4+ T cells is coordinately regulated by signals through the T cell receptor and cytokine receptors, yet how these signals are integrated remains elusive. Here we find that mice lacking Dock2, a Rac activator in lymphocytes, developed allergic disease through a mechanism dependent on CD4+ T cells and the interleukin 4 receptor (IL-4R). Dock2-deficient CD4+ T cells showed impaired antigen-driven downregulation of IL-4Ralpha surface expression, resulting in sustained IL-4R signaling and excessive T helper type 2 responses. Dock2 was required for T cell receptor-mediated phosphorylation of the microtubule-destabilizing protein stathmin and for lysosomal trafficking and the degradation of IL-4Ralpha. Thus, Dock2 links T cell receptor signals to downregulation of IL-4Ralpha to control the lineage commitment of CD4+ T cells.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Receptores de Superficie Celular/metabolismo , Células Th2/inmunología , Secuencia de Aminoácidos , Animales , Factores de Intercambio de Guanina Nucleótido , Interleucina-4/biosíntesis , Ratones , Ratones Endogámicos C57BL , Microtúbulos/fisiología , Datos de Secuencia Molecular , Transporte de Proteínas , Células Th2/citología
16.
Parasitol Res ; 100(5): 1023-31, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17171567

RESUMEN

Fifty-eight stocks of Trypanosoma cruzi from Latin America were genetically characterized using the methods of the polymerase chain reaction (PCR) and the single-stranded conformation DNA polymorphism (SSCP) with four genes, mini-exon, 24Salpha rRNA, 18Sr RNA, cruzipain, and a RAPD fragment DNA region, P7-P8. All the isolates examined were assigned to T. cruzi I or subgroups of T. cruzi II by these methods. From these results, the SSCP analysis, which was simple to perform and highly sensitive to sequence variation, seemed to be a good modality for characterizing T. cruzi, particularly for subgroups of T. cruzi II. However, in several isolates of T. cruzi II, the subgroups determined with the SSCP of 24Salpha rRNA were not consistent with those determined with other genes, the SSCP of 18S rRNA and cruzipain, and the PCR of P7-P8, possibly because of the occurrence of rare genetic exchanges or mutations or both in natural populations of this parasite. The SSCP patterns of 24Salpha rRNA and 18S rRNA were highly variable in the T. cruzi I isolates; therefore, analyses using both genes are considered to be one possible method for the characterization of isolates within T. cruzi I.


Asunto(s)
Enfermedad de Chagas/parasitología , ADN Protozoario/genética , Polimorfismo Conformacional Retorcido-Simple , Trypanosoma cruzi/clasificación , Animales , Animales Salvajes/parasitología , Cisteína Endopeptidasas/genética , ADN Ribosómico/genética , Exones/genética , Genes Protozoarios , Humanos , Proteínas Protozoarias , ARN Ribosómico 18S/genética , Triatoma/parasitología , Trypanosoma cruzi/genética
17.
J Immunol ; 177(10): 7059-66, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17082622

RESUMEN

Host resistance to the intracellular protozoan parasite Trypanosoma cruzi depends on IFN-gamma production by T cells and NK cells. However, the involvement of innate immunity in host resistance to T. cruzi remains unclear. In the present study, we investigated host defense against T. cruzi by focusing on innate immunity. Macrophages and dendritic cells (DCs) from MyD88(-/-)TRIF(-/-) mice, in which TLR-dependent activation of innate immunity was abolished, were defective in the clearance of T. cruzi and showed impaired induction of IFN-beta during T. cruzi infection. Neutralization of IFN-beta in MyD88(-/-) macrophages led to enhanced T. cruzi growth. Cells from MyD88(-/-)IFNAR1(-/-) mice also showed impaired T. cruzi clearance. Furthermore, both MyD88(-/-)TRIF(-/-) and MyD88(-/-)IFNAR1(-/-) mice were highly susceptible to in vivo T. cruzi infection, highlighting the involvement of innate immune responses in T. cruzi infection. We further analyzed the molecular mechanisms for the IFN-beta-mediated antitrypanosomal innate immune responses. MyD88(-/-)TRIF(-/-) and MyD88(-/-)IFNAR1(-/-) macrophages and DCs exhibited defective induction of the GTPase IFN-inducible p47 (IRG47) after T. cruzi infection. RNA interference-mediated reduction of IRG47 expression in MyD88(-/-) macrophages resulted in increased intracellular growth of T. cruzi. These findings suggest that TLR-dependent expression of IFN-beta is involved in resistance to T. cruzi infection through the induction of IRG47.


Asunto(s)
Enfermedad de Chagas/inmunología , Interferón beta/biosíntesis , Receptores Toll-Like/fisiología , Trypanosoma cruzi/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Células Cultivadas , Enfermedad de Chagas/genética , Enfermedad de Chagas/parasitología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/parasitología , GTP Fosfohidrolasas/biosíntesis , GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP/biosíntesis , Proteínas de Unión al GTP/fisiología , Regulación de la Expresión Génica/inmunología , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/fisiología , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interferón beta/fisiología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Trypanosoma cruzi/crecimiento & desarrollo
18.
J Exp Med ; 203(4): 1021-31, 2006 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-16606674

RESUMEN

Suppressor of cytokine signaling (SOCS)3 is a major negative feedback regulator of signal transducer and activator of transcription (STAT)3-activating cytokines. Transgenic mouse studies indicate that high levels of SOCS3 in T cells result in type 2 T helper cell (Th2) skewing and lead to hypersensitivity to allergic diseases. To define the physiological roles of SOCS3 in T cells, we generated T cell-specific SOCS3 conditional knockout mice. We found that the mice lacking SOCS3 in T cells showed reduced immune responses not only to ovalbumin-induced airway hyperresponsiveness but also to Leishmania major infection. In vitro, SOCS3-deficient CD4+ T cells produced more transforming growth factor (TGF)-beta1 and interleukin (IL)-10, but less IL-4 than control T cells, suggesting preferential Th3-like differentiation. We found that STAT3 positively regulates TGF-beta1 promoter activity depending on the potential STAT3 binding sites. Furthermore, chromatin immunoprecipitation assay revealed that more STAT3 was recruited to the TGF-beta1 promoter in SOCS3-deficient T cells than in control T cells. The activated STAT3 enhanced TGF-beta1 and IL-10 expression in T cells, whereas the dominant-negative form of STAT3 suppressed these. From these findings, we propose that SOCS3 regulates the production of the immunoregulatory cytokines TGF-beta1 and IL-10 through modulating STAT3 activation.


Asunto(s)
Regulación hacia Abajo/inmunología , Interleucina-10/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética , Linfocitos T Colaboradores-Inductores/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Diferenciación Celular/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/fisiología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1 , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
19.
Int Immunol ; 18(5): 679-87, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16569681

RESUMEN

MUT1 is an H-2Kb-restricted 8-mer CTL epitope expressed in Lewis lung carcinoma (3LL) tumor cells derived from C57BL/6 (B6) mice. We constructed a chimeric gene encoding ubiquitin-fused MUT1 (pUB-MUT1). By using a gene gun, B6 mice were immunized with the gene prior to challenge with 3LL tumor cells. Tumor growth and lung metastasis were prominently suppressed in mice immunized with pUB-MUT1 but only slightly in those immunized with the MUT1 gene (pMUT) alone. CD8+ T cells were confirmed to be the final effector by in vitro experiments and in vivo removal of the cells with a corresponding antibody. Anti-tumor immunity was profoundly suppressed in mice deficient in an immuno-subunit of proteasome, LMP7. Furthermore, mice deficient in a proteasome regulator, PA28alpha/beta, failed to acquire protective immunity. Thus, application of the ubiquitin-fusion degradation pathway was useful even in immunization with genes encoding a single CTL epitope for induction of specific and active CD8+ T cells.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Epítopos de Linfocito T/inmunología , Oligopéptidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T Citotóxicos/inmunología , Ubiquitina/inmunología , Animales , Presentación de Antígeno/inmunología , Biolística , Células COS , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/prevención & control , Chlorocebus aethiops , Epítopos de Linfocito T/biosíntesis , Femenino , Interferón gamma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/inmunología , Oligopéptidos/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/inmunología , Proteínas/metabolismo , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/metabolismo , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología
20.
Microbes Infect ; 8(4): 1045-53, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16515877

RESUMEN

The ubiquitin-proteasome system (UPS) plays an indispensable role in inducing MHC class I-restricted CD8+ T cells and was exploited in the development of a DNA vaccine against the intracellular protozoan Toxoplasma gondii by constructing a chimeric DNA encoding a fusion protein between murine ubiquitin and the toxoplasma antigen SAG1. The SAG1 peptide was promptly degraded in antigen-presenting cells (APCs) transfected with the chimeric DNA. Degradation, however, was hampered by incubating the APCs with the proteasome inhibitor epoxomicin. Mice vaccinated with the DNA acquired potent protective immunity mediated by MHC class I-restricted CD8+ T cells against infection by the highly virulent Toxoplasma. The accelerated degradation and induction of immunity were dependent on the UPS since mice lacking an immuno-subunit of 20S proteasome, LMP7, lost these functions, although they were independent of the proteasome regulator PA28alpha/beta complex.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Complejos Multienzimáticos/inmunología , Proteínas Protozoarias/inmunología , Vacunas Antiprotozoos/administración & dosificación , Toxoplasma/inmunología , Toxoplasmosis/prevención & control , Ubiquitina/inmunología , Animales , Antígenos de Protozoos/metabolismo , Biolística , Femenino , Esquemas de Inmunización , Complejo Mayor de Histocompatibilidad/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Proteínas/genética , Proteínas/inmunología , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Especificidad del Receptor de Antígeno de Linfocitos T , Toxoplasmosis/inmunología , Ubiquitina/metabolismo , Vacunas de ADN/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...