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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Nat Med ; 10(1): 29-30, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702631

RESUMEN

Infection with malaria parasites frequently induces total immune suppression, which makes it difficult for the host to maintain long-lasting immunity. Here we show that depletion of CD4(+)CD25(+) regulatory T cells (T(reg)) protects mice from death when infected with a lethal strain of Plasmodium yoelii, and that this protection is associated with an increased T-cell responsiveness against parasite-derived antigens. These results suggest that activation of T(reg) cells contributes to immune suppression during malaria infection, and helps malaria parasites to escape from host immune responses.


Asunto(s)
Antígenos CD4/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Receptores de Interleucina-2/inmunología , Linfocitos T/inmunología , Animales , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C
5.
Nat Med ; 9(8): 1047-54, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12847520

RESUMEN

Members of the suppressor of cytokine signaling (SOCS) family are involved in the pathogenesis of many inflammatory diseases. SOCS-3 is predominantly expressed in T-helper type 2 (T(H)2) cells, but its role in T(H)2-related allergic diseases remains to be investigated. In this study we provide a strong correlation between SOCS-3 expression and the pathology of asthma and atopic dermatitis, as well as serum IgE levels in allergic human patients. SOCS-3 transgenic mice showed increased T(H)2 responses and multiple pathological features characteristic of asthma in an airway hypersensitivity model system. In contrast, dominant-negative mutant SOCS-3 transgenic mice, as well as mice with a heterozygous deletion of Socs3, had decreased T(H)2 development. These data indicate that SOCS-3 has an important role in regulating the onset and maintenance of T(H)2-mediated allergic immune disease, and suggest that SOCS-3 may be a new therapeutic target for the development of antiallergic drugs.


Asunto(s)
Asma/inmunología , Dermatitis Atópica/inmunología , Hipersensibilidad/inmunología , Proteínas/metabolismo , Proteínas Represoras , Células Th2/inmunología , Factores de Transcripción , Animales , Hiperreactividad Bronquial/inmunología , Pruebas de Provocación Bronquial , Proteínas de Unión al ADN/metabolismo , Humanos , Hipersensibilidad/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas/genética , Factor de Transcripción STAT4 , Transducción de Señal/fisiología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Células Th2/fisiología , Transactivadores/metabolismo
6.
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
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.
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
9.
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
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.
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
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.
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
14.
Int J Biochem Cell Biol ; 37(4): 700-6, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15694829

RESUMEN

Malaria is one of the most life-threatening infectious diseases worldwide. Specific immunity to natural infection is acquired slowly despite a high degree of repeated exposure and rarely continues for a long time even in endemic areas. Malaria parasites have evolved to acquire diverse immune evasion mechanisms that evoke poor immune responses and allow infection of individuals previously exposed. The shrewd schema of malaria parasites also hampers the development of effective vaccines. Furthermore, some of those mechanisms are essential for malaria pathogenesis. In this article, an outline of protective immunity to malaria is given, then strategies used by malaria parasites to evade host immunity, including antigen diversity/polymorphism, antigen variation and total immune suppression, are reviewed. Finally, trials to control malaria based on accumulating insights into the host-parasite relationship are discussed.


Asunto(s)
Malaria/inmunología , Plasmodium/patogenicidad , Animales , Humanos , Estadios del Ciclo de Vida , Malaria/terapia , Vacunas contra la Malaria/uso terapéutico , Plasmodium/inmunología , Plasmodium/fisiología
15.
Microbes Infect ; 6(5): 468-74, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15109961

RESUMEN

Prior to the activation of CD4+ T cells, exogenous proteins are digested by endo/lysosomal enzymes in antigen-presenting cells (APCs) to produce antigenic peptides that are presented on MHC class II molecules. In the studies described here, the functional significance of cathepsin L for antigen processing and Th1/Th2 differentiation in experimental leishmaniasis was investigated. We first demonstrated that cathepsin L is one of the candidates for endo/lysosomal enzymes in the processing of soluble Leishmania antigen (SLA) by using CLIK148, a specific inhibitor of cathepsin L. Treatment of BALB/c or DBA/2 mice with CLIK148 exacerbated the disease by enhancing an SLA-specific Th2-type response such as IL-4 production. CLIK148 did not exert any direct influence on Leishmania major promastigotes themselves or on the course of L. major infection in SCID mice. Taken together, these findings suggest that treatment of host mice with CLIK148 affects the processing of SLA in APCs, resulting in the potentiation of Th2-type immune responses and thus leading to exacerbation of the disease. Furthermore, endo/lysosomal cathepsin L was found to be functionally distinct from previously described cathepsins B and D.


Asunto(s)
Presentación de Antígeno , Catepsinas/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Células TH1/inmunología , Animales , Antígenos de Protozoos/metabolismo , Catepsina B/antagonistas & inhibidores , Catepsina B/metabolismo , Catepsina L , Catepsinas/antagonistas & inhibidores , Células Cultivadas , Cisteína Endopeptidasas , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Compuestos Epoxi/farmacología , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Interferón gamma/análisis , Interleucina-4/análisis , Leishmaniasis Cutánea/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Inhibidores de Proteasas/farmacología , Piridinas/farmacología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
16.
Microbes Infect ; 4(1): 1-11, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11825769

RESUMEN

We investigated the roles of gamma delta T, NK, and NK1.1(+) T-like (NKT) cells in protective immunity against infection with Toxoplasma gondii. gamma delta T cells, NKT and NK cells, and NK cells in BALB/c mice were depleted by treatment with anti-TCR-gamma delta monoclonal antibody (mAb), anti-interleukin-2 receptor beta chain (IL-2R beta) mAb, and anti-asialoGM1 Ab, respectively, and these mice were infected with T. gondii. Treatment of mice with anti-TCR-gamma delta mAb aggravated toxoplasmosis, while treatment with anti-asialoGM1 Ab had no effects. Treatment with anti-IL-2R beta mAb enhanced the expression of heat shock protein 65 (HSP65) and gamma interferon (IFN-gamma) mRNA, while it inhibited interleukin-4 (IL-4) mRNA expression, ameliorating toxoplasmosis. In addition to NK cells, anti-IL-2R beta mAb eliminated cells expressing IL-2R beta and intermediate levels of CD3 (IL-2R beta(+) CD3(int)). Mice treated with anti-IL-2R beta mAb decreased the number of DX5(+) CD3(int) cells, which are considered to be equivalent to NK1.1(+)T cells in NK1.1 allele-negative strains. IL-2R beta(+) CD3(int) cells isolated from splenic and hepatic lymphoid cells were confirmed to express the TCR-V alpha 14 transcript. The magnitude of HSP65 induction in macrophages correlated with the protective potential against T. gondii infection after treatment with the antibodies, supporting our previous finding that gamma delta T cells play an essential role in the induction of HSP65 in host macrophages. Interestingly, NKT cells suppressed the expression of gamma delta T cell-induced HSP65 and IFN-gamma. Furthermore, depletion of IL-2R beta(+) CD3(int) cells suppressed the IL-4 mRNA expression. These results suggest that NKT cells may be the cells responsible for suppression of protective immunity against T. gondii infection by interfering with the gamma delta T cell-induced HSP65 expression, possibly through the generation of IL-4.


Asunto(s)
Proteínas Bacterianas , Chaperoninas/metabolismo , Células Asesinas Naturales/inmunología , Macrófagos/metabolismo , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Chaperonina 60 , Femenino , Gangliósido G(M1)/inmunología , Inmunidad Innata , Interferón gamma/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Interleucina-2/inmunología , Linfocitos T/inmunología
17.
Cancer Gene Ther ; 9(2): 156-63, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11857033

RESUMEN

We studied interleukin (IL)-12 gene therapy using a gene gun as a new autologous vaccination strategy for cancer. In the first experiment, BALB/c mice were inoculated with syngeneic murine renal cancer cells (Renca) intradermally in the abdomen. This was followed by an injection of IL-12 expression plasmid using the gene gun. About 40% of the mice exhibited rejection of the tumor after the treatment and these mice also acquired immunological resistance against a secondary challenge with Renca cells. Based on these results, we examined whether antitumor activity can be potentiated when mice undergo combination treatment with intradermal inoculation of irradiated Renca cells and transfection with IL-12 gene. Inoculation of irradiated Renca cells alone was partially effective in inducing antitumor immunity, whereas the combined treatment remarkably intensified this effect. Moreover, this combined treatment inhibited tumor establishment and enhanced survival of the mice with tumor infiltration by CD4(+) and CD8(+) T cells, even when the treatment was started after tumor-implantation at a distant site. This antitumor effect was antigen specific and we confirmed the induction of antitumor cytotoxic T cells by this treatment. These results show that local cutaneous transfer of IL-12 expression plasmid using gene gun technology enhances systemic and specific antitumor immunity primed by irradiated tumor cells.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/terapia , Interleucina-12/genética , Neoplasias Renales/terapia , Plásmidos/genética , Células Tumorales Cultivadas/efectos de la radiación , Animales , Biolística , Carcinoma de Células Renales/inmunología , División Celular/fisiología , Terapia Combinada , Cartilla de ADN/química , Expresión Génica , Terapia Genética , Vectores Genéticos , Técnicas para Inmunoenzimas , Inyecciones Intralesiones , Interleucina-12/metabolismo , Neoplasias Renales/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología
18.
J Dermatol Sci ; 34(3): 209-19, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15113591

RESUMEN

BACKGROUND: Autologous whole tumor cell-based vaccinations would seem to be ideal since such vaccinations, in contrast to vaccination with a single defined antigen, have the potential to elicit a broad type of T-cell immune response to tumor-associated antigens. OBJECTIVE: We modified formaldehyde (formalin)-fixed mouse melanoma cells and investigated the utility of those cells as sources of tumor antigens for immunotherapy. METHODS: C57BL/6 or the proteasome activator PA28alpha-knockout mice were intradermally inoculated with 1% formalin-fixed B16 cells three times at weekly intervals either before or after tumor challenge. Simultaneously, interleukin-12 gene was transferred into the skin around immunization sites using gene gun technology. The effects were evaluated by tumor growth, antigen-specific interferon-gamma production in splenic lymphocytes, and activation of dendritic cells. RESULTS: Fixed cells directly induced production of tumor necrosis factor-alpha in dendritic cells more effectively than did frozen and thawed cells. More than 60% of the mice immunized with fixed cells and interleukin-12 rejected the challenged B16 tumor. CD4+ T cells from those mice produced a significant amount of interferon-gamma in response to melanoma cells. Furthermore, this combined treatment showed antitumor immunity initiated by CD8+ and CD4+ T cells in the therapeutic experiments. PA28alpha/beta appeared not to be required for the development of CD8+ T cells, although it is known to be essential for the development of CD8+ T cells specific for tyrosinase-related protein-2, one of melanocyte-lineage differentiated antigens. CONCLUSION: These results suggest that formalin-fixed autologous melanoma cells have a potential to function as effective antigen sources for immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Melanoma/inmunología , Melanoma/prevención & control , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/prevención & control , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Muerte Celular/inmunología , Femenino , Fijadores , Formaldehído , Interferón gamma/metabolismo , Interleucina-12/inmunología , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal , Proteínas/genética , Neoplasias Cutáneas/terapia , Bazo/citología , Bazo/inmunología , Células Tumorales Cultivadas
19.
Parasitol Int ; 53(4): 337-44, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15464443

RESUMEN

Isozyme analysis with 18 enzyme loci was conducted on 146 isolates of Trypanosoma cruzi from Mexico, Guatemala, Colombia, Ecuador, Peru, Brazil, Bolivia, Paraguay and Chile. Forty-four different MLGs (groups of isolates with identical multilocus genotypes) were identified and a phylogeny was constructed. The phylogenetic tree consisted of two main groups (T. cruzi I, T. cruzi II), and the latter was further divided into two subgroups (T. cruzi IIa, T. cruzi IIb-e). Evidence of hybridization between different MLGs of T. cruzi II was found, which means that genetic exchanges seem to have occurred in South American T. cruzi. On the other hand, the persistence of characteristic T. cruzi I and T. cruzi II isozyme patterns in single small villages in Bolivia and Guatemala suggested that genetic exchange is very rare between major lineages. A significant difference in genetic diversity was shown between T. cruzi I and T. cruzi II from several indices of population genetics. Two possibilities could explain this genetic variation in the population: differences in evolutionary history and/or different tendencies to exchange genetic material. Broad-scale geographic distributions of T. cruzi I and T. cruzi IIb-e were different; T. cruzi I occurred in Central America and south to Bolivia and Brazil, while T. cruzi IIb-e occurred in the central and southern areas of South America, overlapping with T. cruzi I in Brazil and Bolivia.


Asunto(s)
Enfermedad de Chagas/epidemiología , Variación Genética , Filogenia , Trypanosoma cruzi/clasificación , Animales , Enfermedad de Chagas/parasitología , ADN Protozoario/análisis , Genotipo , Guatemala/epidemiología , Humanos , Isoenzimas/genética , México/epidemiología , América del Sur/epidemiología , Triatoma/parasitología , Trypanosoma cruzi/genética , Trypanosoma cruzi/patogenicidad
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA