Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 204(8): 2242-2256, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32161097

RESUMEN

Type I IFNs play a complex role in determining the fate of microbial pathogens and may also be deleterious to the host during bacterial and viral infections. Upon ligand binding, a receptor proximal complex consisting of IFN-α and -ß receptors 1 and 2 (IFNAR1, IFNAR2, respectively), tyrosine kinase 2 (Tyk2), Jak1, and STAT2 are assembled and promote the phosphorylation of STAT1 and STAT2. However, how the IFNARs proximal complex is assembled upon binding to IFN is poorly understood. In this study, we show that the membrane-associated pore-forming protein Perforin-2 (P2) is critical for LPS-induced endotoxic shock in wild-type mice. Type I IFN-mediated JAK-STAT signaling is severely impaired, and activation of MAPKs and PI3K signaling pathways are delayed in P2-deficient mouse bone marrow-derived macrophages, mouse embryonic fibroblasts (MEFs), and human HeLa cells upon IFN stimulation. The P2 N-glycosylated extracellular membrane attack complex/perforin domain and the P2 domain independently associate with the extracellular regions of IFNAR1 and IFNAR2, respectively, in resting MEFs. In addition, the P2 cytoplasmic tail domain mediated the constitutive interaction between STAT2 and IFNAR2 in resting MEFs, an interaction that is dependent on the association of the extracellular regions of P2 and IFNAR2. Finally, the constitutive association of P2 with both receptors and STAT2 is critical for the receptor proximal complex assembly and reciprocal transphosphorylation of Jak1 and Tyk2 as well as the phosphorylation and activation of STAT1 and STAT2 upon IFN-ß stimulation.


Asunto(s)
Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Transducción de Señal/inmunología , Animales , Células Cultivadas , Células HeLa , Humanos , Lipopolisacáridos , Ratones , Ratones Noqueados , Choque Séptico/inducido químicamente , Choque Séptico/inmunología
2.
Microb Pathog ; 154: 104852, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33762201

RESUMEN

Cellular Perforin-2 (MPEG1) is a pore-forming MACPF family protein that plays a critical role in the defense against bacterial pathogens. Macrophages, neutrophils, and several other cell types that are part of the front line of innate defenses constitutively express high levels of Perforin-2; whereas, most other cell types must be induced to express Perforin-2 by interferons (α, ß and γ) and/or PAMPs such as LPS. In this study, we demonstrate that many bacterial pathogens can limit the expression of Perforin-2 in cells normally inducible for Perforin-2 expression, while ordinarily commensal or non-pathogenic bacteria triggered high levels of Perforin-2 expression in these same cell types. The mechanisms by which pathogens suppress Perforin-2 expression was explored further using Salmonella enterica serovar Typhimurium and cultured MEFs as well as intestinal epithelial cell lines. These studies identified multiple factors required to minimize the expression of Perforin-2 in cell types inducible for Perforin-2 expression. These included the PmrAB and PhoPQ two-component systems, select LPS modification enzymes and the Type III secretion effector protein AvrA.


Asunto(s)
Lipopolisacáridos , Salmonella typhimurium , Proteínas Bacterianas/genética , Células Epiteliales , Fibroblastos , Perforina/genética , Serogrupo
3.
J Immunol ; 199(8): 2721-2728, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28877989

RESUMEN

Stimulation of several TNF receptor family proteins has been shown to dampen inflammatory disease in murine models through augmenting the number and/or activity of regulatory T cells (Tregs). We recently found that one molecule, 4-1BB, used binding to Galectin-9 to exert its immunosuppressive effects and drive expansion of CD8+Foxp3- Tregs. We now show that ligation of another TNFR family molecule, DR3, which has previously been found to strongly expand CD4+Foxp3+ Tregs and suppress inflammation, also requires Galectin-9. We found that the extracellular region of DR3 directly binds to Galectin-9, and that Galectin-9 associates with DR3 in Tregs. From studies in vitro with Galectin-9-/- CD4+ T cells and Tregs, we found that stimulatory activity induced by ligating DR3 was in part dependent on Galectin-9. In vivo, in a model of experimental autoimmune encephalomyelitis, we show that an agonist of DR3 suppressed disease, correlating with expansion of CD4+Foxp3+ Tregs, and this protective effect was lost in Galectin-9-/- mice. Similar results were seen in an allergic lung inflammation model. Thus, we demonstrate a novel function of Galectin-9 in facilitating activity of DR3 related to Treg-mediated suppression.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Galectinas/metabolismo , Inflamación/inmunología , Esclerosis Múltiple/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Galectinas/genética , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal
4.
Immunity ; 30(5): 668-70, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19464990

RESUMEN

Perforation of membranes and pore formation is mediated by polymerization of proteins of the immune system, complement C9 and Perforin, which share the conserved MACPF domain. In this issue of Immunity, Baran et al. (2009) identify the molecular mechanism initiating polymerization as charge interactions in the MACPF domain.


Asunto(s)
Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Perforina/metabolismo , Animales , Humanos , Perforina/genética , Estructura Terciaria de Proteína/fisiología
5.
Biol Blood Marrow Transplant ; 23(5): 757-766, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28219835

RESUMEN

Regulatory T cells (Tregs) are critical for self-tolerance. Although adoptive transfer of expanded Tregs limits graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation (HSCT), ex vivo generation of large numbers of functional Tregs remains difficult. Here, we demonstrate that in vivo targeting of the TNF superfamily receptor TNFRSF25 using the TL1A-Ig fusion protein, along with IL-2, resulted in transient but massive Treg expansion in donor mice, which peaked within days and was nontoxic. Tregs increased in multiple compartments, including blood, lymph nodes, spleen, and colon (GVHD target tissue). Tregs did not expand in bone marrow, a critical site for graft-versus-malignancy responses. Adoptive transfer of in vivo-expanded Tregs in the setting of MHC-mismatched or MHC-matched allogeneic HSCT significantly ameliorated GVHD. Critically, transplantation of Treg-expanded donor cells facilitated transplant tolerance without GVHD, with complete sparing of graft-versus-malignancy. This approach may prove valuable as a therapeutic strategy promoting transplantation tolerance.


Asunto(s)
Traslado Adoptivo/métodos , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia , Trasplante de Células Madre Hematopoyéticas/métodos , Linfocitos T Reguladores/trasplante , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Inmunoglobulinas/farmacología , Interleucina-2/farmacología , Ratones , Ratones Endogámicos BALB C , Autotolerancia , Linfocitos T Reguladores/citología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
6.
Gynecol Oncol ; 145(3): 413-419, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28392126

RESUMEN

OBJECTIVES: Cancer-testis (CT) antigens have been proposed as potential targets for cancer immunotherapy. Our objective was to evaluate the expression of a panel of CT antigens in epithelial ovarian cancer (EOC) tumor specimens, and to determine if antigen sharing occurs between tumors. METHODS: RNA was isolated from EOC tumor specimens, EOC cell lines and benign ovarian tissue specimens. Real time-PCR analysis was performed to determine the expression level of 20 CT antigens. RESULTS: A total of 62 EOC specimens, 8 ovarian cancer cell lines and 3 benign ovarian tissues were evaluated for CT antigen expression. The majority of the specimens were: high grade (62%), serous (68%) and advanced stage (74%). 58 (95%) of the EOC tumors analyzed expressed at least one of the CT antigens evaluated. The mean number of CT antigen expressed was 4.5 (0-17). The most frequently expressed CT antigen was MAGE A4 (65%). Antigen sharing analysis showed the following: 9 tumors shared only one antigen with 62% of the evaluated specimens, while 37 tumors shared 4 or more antigens with 82%. 5 tumors expressed over 10 CT antigens, which were shared with 90% of the tumor panel. CONCLUSION: CT antigens are expressed in 95% of EOC tumor specimens. However, not a single antigen was universally expressed across all samples. The degree of antigen sharing between tumors increased with the total number of antigens expressed. These data suggest a multi-epitope approach for development of immunotherapy for ovarian cancer treatment.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Ováricas/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Neoplásico/genética , ARN Neoplásico/inmunología , Adulto Joven
7.
Infect Immun ; 84(4): 1083-1091, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26831467

RESUMEN

The host-encoded Perforin-2 (encoded by the macrophage-expressed gene 1, Mpeg1), which possesses a pore-forming MACPF domain, reduces the viability of bacterial pathogens that reside within membrane-bound compartments. Here, it is shown that Perforin-2 also restricts the proliferation of the intracytosolic pathogen Listeria monocytogenes Within a few hours of systemic infection, the massive proliferation of L. monocytogenes in Perforin-2(-/-)mice leads to a rapid appearance of acute disease symptoms. We go on to show in cultured Perforin-2(-/-)cells that the vacuole-to-cytosol transitioning of L. monocytogenesis greatly accelerated. Unexpectedly, we found that in Perforin-2(-/-)macrophages,Listeria-containing vacuoles quickly (≤ 15 min) acidify, and that this was coincident with greater virulence gene expression, likely accounting for the more rapid translocation of L. monocytogenes to its replicative niche in the cytosol. This hypothesis was supported by our finding that aL. monocytogenes strain expressing virulence factors at a constitutively high level replicated equally well in Perforin-2(+/+)and Perforin-2(-/-)macrophages. Our findings suggest that the protective role of Perforin-2 against listeriosis is based on it limiting the intracellular replication of the pathogen. This cellular activity of Perforin-2 may derive from it regulating the acidification of Listeria-containing vacuoles, thereby depriving the pathogen of favorable intracellular conditions that promote its virulence gene activity.


Asunto(s)
Citosol/fisiología , Listeria monocytogenes/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Vacuolas/fisiología , Animales , Células Cultivadas , Citosol/microbiología , Regulación de la Expresión Génica/fisiología , Listeria monocytogenes/ultraestructura , Listeriosis/metabolismo , Listeriosis/microbiología , Proteínas de la Membrana/genética , Ratones , Proteínas Citotóxicas Formadoras de Poros/genética , Estructura Terciaria de Proteína
8.
Blood ; 123(19): 3045-55, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24687086

RESUMEN

Tumor relapse is the primary cause of mortality in patients with hematologic cancers following autologous hematopoietic stem cell transplantation (HSCT). Vaccination early after HSCT can exploit both the state of lymphopenia and minimal residual disease for generating antitumor immunity. Here, multiple vaccinations using lymphoma cells engineered to secrete heat shock protein fusion gp96-Ig within 2 weeks of T cell-replete syngeneic HSCT led to cross-presentation and increased survival of lymphoma-bearing mice. To enhance vaccine efficacy, interleukin (IL)-2 was directed to predominantly memory phenotype CD8(+) T lymphocytes and natural killer (NK) cells via administration bound to anti-IL-2 monoclonal antibody clone S4B6 (IL-2S4B6). Combination therapy with gp96-Ig vaccination and coordinated infusions of IL-2S4B6 resulted in marked prolongation of survival, which directly correlated with ~500% increase in effector CD8(+) T-cell numbers. Notably, this dual regimen elicited large increases in both donor CD8(+) T and NK cells, but not CD4(+) T lymphocytes; the former 2 populations are essential for both vaccine efficacy and protection against opportunistic infections after HSCT. Indeed, IL-2S4B6-treated HSCT recipients infected with Listeria monocytogenes exhibited decreased bacterial levels. These preclinical studies validate a new strategy particularly well suited to the post-HSCT environment, which may augment adaptive and innate immune function in patients with malignant disease receiving autologous HSCT.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Trasplante de Médula Ósea , Proteínas de Choque Térmico/inmunología , Interleucina-2/inmunología , Linfoma/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Terapia Combinada , Interacciones Huésped-Patógeno/inmunología , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Recuento de Linfocitos , Linfoma/patología , Linfoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Resultado del Tratamiento , Vacunación/métodos
9.
J Autoimmun ; 57: 14-23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25533628

RESUMEN

The CD30 ligand (CD30L)/CD30 axis plays a critical role in Th1 and Th17 cell differentiation. However, the role in the pathogenesis of central nervous system autoimmunity remains unknown. Here we show the resistance for experimental autoimmune encephalomyelitis (EAE) with markedly reduced induction of antigen-specific Th1 and Th17 cells in CD30L knockout mice. Bone marrow chimera experiments indicated that CD30L on bone marrow-derived cells were critical for the development of EAE and that CD30L reverse signaling in CD4 T cells was dispensable for the pathogenic Th17 cell differentiation at the induction phase. Adoptive transfer experiment revealed an additional role for CD30L in the environment at the effector phase. In vivo neutralization of CD30L by soluble murine CD30-Immunoglobulin fusion protein before disease onset or even after disease onset significantly ameliorated the clinical symptoms. These results indicate that CD30L/CD30 signaling is critically involved in antigen-specific CD4 T cell responses at both the induction and effector phase, thus could be a new target molecule for the treatment of central nervous system autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Ligando CD30/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Antígeno Ki-1/inmunología , Traslado Adoptivo , Animales , Autoinmunidad/genética , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Ligando CD30/genética , Ligando CD30/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Antígeno Ki-1/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
10.
J Immunol ; 190(4): 1540-50, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23319737

RESUMEN

TNF superfamily member 15 (TL1A) is the ligand for TNFR superfamily (TNFRSF)25. We previously reported that TNFRSF25 stimulation with an agonist Ab, 4C12, expands pre-existing CD4(+)Foxp3(+) regulatory T cells (Tregs) in vivo. To determine how the physiological ligand differs from the Ab, we generated a soluble mouse TL1A-Ig fusion protein that forms a dimer of TL1A trimers in solution with an apparent molecular mass of 516 kDa. In vitro, TL1A-Ig mediated rapid proliferation of Foxp3(+) Tregs and a population of CD4(+)Foxp3(-) conventional T cells. TL1A-Ig also blocked de novo biogenesis of inducible Tregs and it attenuated the suppressive function of Tregs. TNFRSF25 stimulation by TL1A-Ig in vivo induced expansion of Tregs such that they increased to 30-35% of all CD4(+) T cells in the peripheral blood within 5 d of treatment. Treg proliferation in vivo was dependent on TCR engagement with MHC class II. Elevated Treg levels can be maintained for at least 20 d with daily injections of TL1A-Ig. TL1A-Ig-expanded Tregs expressed high levels of activation/memory markers KLRG1 and CD103 and were highly suppressive ex vivo. TL1A-Ig-mediated Treg expansion in vivo was protective against allergic lung inflammation, a mouse model for asthma, by reversing the ratio of conventional T cells to Tregs in the lung and blocking eosinophil exudation into the bronchoalveolar fluid. Thus, TL1A-Ig fusion proteins are highly active and tightly controllable agents to stimulate Treg proliferation in vivo, and they are uniquely able to maintain high levels of expanded Tregs by repeated administration.


Asunto(s)
Proteínas Recombinantes de Fusión/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/aislamiento & purificación , Animales , Células CHO , Línea Celular Tumoral , Clonación Molecular , Cricetinae , Citometría de Flujo , Genes Reporteros , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Células 3T3 NIH , Plásmidos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Transfección , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
11.
J Immunol ; 190(6): 2495-9, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23401588

RESUMEN

Vaccine-induced protection against infection by HIV or highly pathogenic and virulent SIV strains has been limited. In a proof-of-concept study, we show that a novel vaccine approach significantly protects rhesus macaques from mucosal infection by the highly pathogenic strain SIVmac251. We vaccinated three cohorts of 12 macaques each with live, irradiated vaccine cells secreting the modified endoplasmic reticulum chaperone gp96-Ig. Cohort 1 was vaccinated with cells secreting gp96(SIV)Ig carrying SIV peptides. In addition, Cohort 2 received recombinant envelope protein SIV-gp120. Cohort 3 was injected with cells secreting gp96-Ig (no SIV Ags) vaccines. Cohort 2 was protected from infection. After seven rectal challenges with highly pathogenic SIVmac251, the hazard ratio was 0.27, corresponding to a highly significant, 73% reduced risk for viral acquisition. The apparent success of the novel vaccine modality recommends further study.


Asunto(s)
Mucosa Intestinal/inmunología , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/uso terapéutico , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Estudios de Cohortes , Femenino , Células HEK293 , Humanos , Inyecciones Intraperitoneales , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Macaca mulatta , Masculino , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Enfermedades del Recto/inmunología , Enfermedades del Recto/prevención & control , Enfermedades del Recto/virología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Linfocitos T Citotóxicos/virología
12.
J Immunol ; 189(7): 3311-8, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22956587

RESUMEN

TNFR superfamily (TNFRSF)4 (OX40, CD134) and TNFRSF25 are costimulatory receptors that influence CD4(+) and CD8(+) T cell responses to cognate Ag. Independently, these receptors have been described to stimulate overlapping functions, including enhanced proliferation and activation for both regulatory T cells (CD4(+)Foxp3(+); Tregs) and conventional T cells (CD4(+)Foxp3(-) or CD8(+)Foxp3(-); Tconvs). To determine the relative functionality of TNFRSF4 and TNFRSF25 in T cell immunity, the activity of TNFRSF4 and TNFRS25 agonistic Abs was compared in the context of both traditional protein/adjuvant (OVA/aluminum hydroxide) and CD8(+)-specific heat shock protein-based (gp96-Ig) vaccine approaches. These studies demonstrate that both TNFRSF4 and TNFRSF25 independently and additively costimulate vaccine-induced CD8(+) T cell proliferation following both primary and secondary Ag challenge. In contrast, the activities of TNFRSF4 and TNFRSF25 were observed to be divergent in the costimulation of CD4(+) T cell immunity. TNFRSF4 agonists were potent costimulators of OVA/aluminum hydroxide-induced CD4(+) Tconv proliferation, but they only weakly costimulated Treg proliferation and IgG2a production, whereas TNFRSF25 agonists were strong costimulators of Treg proliferation, producers of IgG1, IgG2a, and IgG2b, and weak costimulators of CD4(+) Tconv proliferation. Interestingly, Ag-specific cellular and humoral responses were uncoupled upon secondary immunization, which was dramatically affected by the presence of TNFRSF4 or TNFRSF25 costimulation. These studies highlight the overlapping but nonredundant activities of TNFRSF4 and TNFRSF25 in T cell immunity, which may guide the application of receptor agonistic agents as vaccine adjuvants for infectious disease and tumor immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunización Secundaria , Activación de Linfocitos/inmunología , Receptores OX40/administración & dosificación , Miembro 25 de Receptores de Factores de Necrosis Tumoral/administración & dosificación , Vacunas/administración & dosificación , Vacunas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/fisiología , Compuestos de Alumbre/administración & dosificación , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Proliferación Celular , Células Cultivadas , Reactividad Cruzada/inmunología , Inmunización Secundaria/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Receptores OX40/fisiología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología
13.
J Immunol ; 189(4): 1780-91, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22786769

RESUMEN

Thymic-derived natural T regulatory cells (Tregs) are characterized by functional and phenotypic heterogeneity. Recently, a small fraction of peripheral Tregs has been shown to express Klrg1, but it remains unclear as to what extent Klrg1 defines a unique Treg subset. In this study, we show that Klrg1(+) Tregs represent a terminally differentiated Treg subset derived from Klrg1(-) Tregs. This subset is a recent Ag-responsive and highly activated short-lived Treg population that expresses enhanced levels of Treg suppressive molecules and that preferentially resides within mucosal tissues. The development of Klrg1(+) Tregs also requires extensive IL-2R signaling. This activity represents a distinct function for IL-2, independent from its contribution to Treg homeostasis and competitive fitness. These and other properties are analogous to terminally differentiated short-lived CD8(+) T effector cells. Our findings suggest that an important pathway driving Ag-activated conventional T lymphocytes also operates for Tregs.


Asunto(s)
Receptores Inmunológicos/inmunología , Receptores de Interleucina-2/inmunología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Lectinas Tipo C , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Receptores Inmunológicos/metabolismo , Receptores de Interleucina-2/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
14.
Semin Immunol ; 22(3): 105-12, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20226686

RESUMEN

Despite enormous effort, promising pre-clinical data in animal studies and over 900 clinical trials in the United States, no cancer vaccine has ever been approved for clinical use. Over the past decade a great deal of progress has been in both laboratory and clinical studies defining the interactions between developing tumors and the immune system. The results of these studies provide a rationale that may help explain the failure of recent therapeutic cancer vaccines in terms of vaccine principles, in selecting which tumors are the most appropriate to target and instruct the design and implementation of state-of-the-art cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia Activa , Neoplasias/inmunología , Neoplasias/terapia , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Humanos , Resultado del Tratamiento
15.
Infect Immun ; 81(10): 3923-34, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918785

RESUMEN

Interleukin-17A (IL-17A)-producing γδ T cells are known to be activated following Mycobacterium bovis bacillus Calmette-Guérin (BCG) infection. Here, we show that CD30, a member of the tumor necrosis factor (TNF) receptor superfamily, is important for activation of IL-17A-producing γδ T cells after BCG infection. Vγ1(-) Vγ4(-) γδ T cells preferentially expressing Vγ6/Vδ1 genes were identified as the major source of IL-17A in the peritoneal cavity during the early stage of BCG infection. The number of IL-17A-producing Vγ1(-) Vγ4(-) γδ T cells bearing Vγ6 increased in peritoneal exudate cells (PEC) of wild-type (WT) mice but not in those of CD30 knockout (KO) mice in response to BCG infection. Consistently, CD30 ligand (CD30L) or CD30 expression, predominantly by Vγ1(-) Vγ4(-) γδ T cells, was rapidly upregulated after BCG infection. Inhibition of CD30L/CD30 signaling by in vivo administration of a soluble CD30 and immunoglobulin fusion protein (CD30-Ig) severely impaired activation of IL-17A-producing Vγ1(-) Vγ4(-) γδ T cells in WT mice, while stimulating CD30L/CD30 signaling by in vivo administration of agonistic anti-CD30 monoclonal antibody (MAb) restored IL-17A production by Vγ1(-) Vγ4(-) γδ T cells in CD30L KO mice after BCG infection. These results suggest that CD30 signaling plays an important role in the activation of IL-17A-producing Vγ1(-) Vγ4(-) γδ T cells bearing Vγ6 at an early stage of BCG infection.


Asunto(s)
Inmunidad Innata/fisiología , Interleucina-17/metabolismo , Antígeno Ki-1/metabolismo , Mycobacterium bovis , Subgrupos de Linfocitos T/metabolismo , Tuberculosis/inmunología , Animales , Anticuerpos Monoclonales , Líquido Ascítico/citología , Ligando CD30/genética , Ligando CD30/metabolismo , Citocinas , Regulación de la Expresión Génica/inmunología , Interleucina-17/genética , Antígeno Ki-1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tuberculosis/microbiología
16.
Cancer Immunol Immunother ; 62(1): 87-99, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22772949

RESUMEN

The mechanisms by which B lymphocytes inhibit anti-tumor immunity remain poorly understood. Murine EMT-6 mammary tumors grow readily in immune competent mice (BALB/c), but poorly in B-cell-deficient µ(-/-) BALB/c mice (BCDM). T regulatory cell (Treg) expansion and function were impaired in BCDM compared with BALB/c. In this study, we compared tumor growth, Treg cell proliferation, tumor lymphocyte infiltration and cytolytic T cell activity in BALB/c, BCDM and BCDM partially reconstituted with B cells by adoptive transfer (BCDM+B). Partial reconstitution of BCDM with adoptively transferred B cells restored EMT-6 tumor growth, which was independent of IL-10 secretion by B cells. Instead, high frequencies of intratumoral B cells were associated with increased recruitment and proliferation of Treg cells within the tumor microenvironment. The B-cell-dependent accumulation of Treg within the tumor microenvironment was associated with reduced tumor infiltration by CD49+ NK and CD8+ T cells and reduced cytotoxic T cell activity against EMT-6 targets. Our studies indicate that tumor-dependent immunosuppression of T-cell-mediated anti-tumor immunity is coordinated within the tumor microenvironment by B-cell-dependent cross talk with Treg cells, which does not require production of IL-10 by B cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Interleucina-10/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Traslado Adoptivo , Animales , Proliferación Celular , Citometría de Flujo , Interleucina-10/inmunología , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor Cross-Talk/inmunología
17.
J Virol ; 86(19): 10606-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22811539

RESUMEN

Ocular infection with herpes simplex virus 1 (HSV-1) results in a chronic immunoinflamammtory reaction in the cornea, which is primarily orchestrated by CD4(+) T cells. Hence, targeting proinflammatory CD4(+) T cells or increasing the representation of cells that regulate their function is a relevant therapeutic strategy. In this report, we demonstrate that effective therapeutic control can be achieved using a combination of approaches under circumstances where monotherapy is ineffective. We use a convenient and highly effective monoclonal antibody (MAb) approach with MAbT25 to expand cells that express the tumor necrosis factor receptor superfamily member 25 (TNFRSF25). In naïve animals, these are predominantly cells that are Foxp3-positive regulatory T cells. MAbT25 treatment before or at the time of initial HSV infection was an effective means of reducing the severity of subsequent stromal keratitis lesions. However, MAbT25 treatment was not effective if given 6 days after infection since it expanded proinflammatory effector T cells, which also express TNFRSF25. Therefore, the MAbT25 procedure was combined with galectin-9 (Gal-9), an approach that compromises the activity of T cells involved in tissue damage. The combination therapy provided highly effective lesion control over that achieved by treatment with one of them. The beneficial outcome of the combination therapy was attributed to the expansion of the regulatory T cell population that additionally expressed activation markers such as CD103 needed to access inflammatory sites. Additionally, there was a marked reduction of CD4(+) gamma interferon-producing effector T cells responsible for orchestrating the tissue damage. The approach that we describe has potential application to control a wide range of inflammatory diseases, in addition to stromal keratitis, an important cause of human blindness.


Asunto(s)
Anticuerpos/química , Galectinas/metabolismo , Herpes Simple/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Simplexvirus/metabolismo , Animales , Anticuerpos Monoclonales/química , Antígenos CD/biosíntesis , Apoptosis , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Córnea/virología , Cricetinae , Femenino , Factores de Transcripción Forkhead/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inflamación , Cadenas alfa de Integrinas/biosíntesis , Queratitis Herpética/inmunología , Queratitis Herpética/virología , Cinética , Ratones , Ratones Endogámicos C57BL
18.
Eur J Immunol ; 41(1): 164-71, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21182087

RESUMEN

The T-cell functions of a proliferation-inducing ligand (APRIL, also known as TNFSF13) remain largely undefined. We previously showed that APRIL suppressed Th2 cytokine production in cultured CD4(+) T cells and Th2 antibody responses. Here we show that APRIL suppresses allergic lung inflammation, which is associated with diminished expression of the transcription factor c-maf. Mice deficient in the April gene (April(-/-) mice) had significantly aggravated lung inflammation compared with WT mice in the ovalbumin-induced allergic lung inflammation model. Likewise, blockade of APRIL in WT mice by the APRIL-receptor fusion protein, transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI)-Ig, enhanced lung inflammation. Transfer of APRIL-sufficient, ovalbumin-specific, TCR-transgenic CD4(+) T (OT-II) cells to April(-/-) mice restored the suppressive effect of APRIL on lung inflammation. Mechanistically, the expression of the Th2 cytokine transcription factor c-maf, but not GATA-3, was markedly enhanced in April(-/-) CD4(+) T cells at the RNA and protein level and under non-polarizing (Th neutral, ThN) and Th2-polarizing conditions. Since c-maf transactivates the IL-4 gene, the increased c-maf expression in April(-/-) mice readily explains increased Th2 cytokine production. Independent of its effect on IL-4, APRIL suppressed IL-13 expression. APRIL thus may regulate lung inflammation in a dual way, by acting on c-maf expression and by directly controlling IL-13 production.


Asunto(s)
Asma/inmunología , Pulmón/inmunología , Neumonía/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Asma/patología , Linfocitos T CD4-Positivos/inmunología , Factor de Transcripción GATA3/inmunología , Interleucina-13/biosíntesis , Interleucina-13/inmunología , Interleucina-4/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Neumonía/patología , Proteínas Proto-Oncogénicas c-maf/inmunología , Proteínas Recombinantes de Fusión/inmunología , Células Th2/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
19.
Eur J Immunol ; 41(10): 2947-54, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21739429

RESUMEN

CD30 ligand (CD30L) plays an important role in the amplification and/or activation of effector CD4(+) T cells, irrespective of Th cell subset. To examine the role of CD30L in allergic rhinitis, we evaluated an OVA model of allergic rhinitis in CD30L knock out (KO) mice on a BALB/c background sensitized with OVA. Symptoms of allergic rhinitis such as eosinophil infiltration into the nasal mucosa were drastically diminished in OVA-sensitized CD30L KO mice following intranasal challenge with OVA. The levels of OVA-specific IgE in the sera and the Th2 response in nasopharynx-associated lymphoid tissues and cervical LNs of CD30L KO mice were significantly lower than those of WT mice following intranasal challenge with OVA. Intranasal administration of CD30-Ig during the effector phase with OVA significantly prevented the development of allergic rhinitis in WT mice. These results suggest that CD30L plays an important role in allergic rhinitis and that the inhibition of CD30L/CD30 signaling might be useful as a novel biological therapy for allergic rhinitis.


Asunto(s)
Ligando CD30/metabolismo , Antígeno Ki-1/metabolismo , Rinitis Alérgica Perenne/inmunología , Células Th2/inmunología , Administración Intranasal , Animales , Ligando CD30/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Movimiento Celular , Eosinófilos/inmunología , Eosinófilos/metabolismo , Inmunoglobulina E/sangre , Inmunoglobulina G/farmacología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mucosa Nasal/inmunología , Nasofaringe/inmunología , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/farmacología
20.
J Immunol ; 185(4): 2222-30, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20639486

RESUMEN

A CD30 ligand (CD30L; CD153) and its receptor, CD30, is a membrane-associated glycoprotein belonging to the TNF superfamily and TNFR superfamily. These were expressed preferentially by activated CD4(+)T cells. In this paper, we show that CD44(low)CD62(hi)CD4(+) T cells from CD30L(-/-) or CD30(-/-) mice exhibited impaired differentiation into Th17 cells but an increased ability to produce IL-2 after in vitro culture under Th17-polarizing conditions. Neutralization with IL-2 by anti-IL-2 mAb partly restored the ability of Th17 differentiation in CD30L(-/-) or CD30(-/-) T cells. Stimulation via CD30L by immobilized anti-CD30L mAb suppressed IL-2 production by CD30(-/-)CD4(+) T cells, indicating that the reverse signal to CD30L is responsible for downregulation of IL-2 production. In vivo Th17 differentiation of CD30L(-/-)CD4(+)CD45RB(high) T cells was also impaired after transfer into SCID mice, whereas CD30L(+/+)CD4(+)CD45RB(high) T cells normally differentiated into Th17 cells in CD30L(-/-) SCID mice. The results of these studies demonstrate that CD30L/CD30 signaling executed by the T-T cell interaction plays a critical role in Th17 cell differentiation, at least partly via downregulation of IL-2 production.


Asunto(s)
Ligando CD30/inmunología , Antígeno Ki-1/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T/inmunología , Animales , Ligando CD30/genética , Ligando CD30/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Citometría de Flujo , Expresión Génica , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-2/metabolismo , Antígeno Ki-1/genética , Antígeno Ki-1/metabolismo , Cinética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA