Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 46(2): 205-219, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28190711

RESUMEN

Adaptive cellular immunity is initiated by antigen-specific interactions between T lymphocytes and dendritic cells (DCs). Plasmacytoid DCs (pDCs) support antiviral immunity by linking innate and adaptive immune responses. Here we examined pDC spatiotemporal dynamics during viral infection to uncover when, where, and how they exert their functions. We found that pDCs accumulated at sites of CD8+ T cell antigen-driven activation in a CCR5-dependent fashion. Furthermore, activated CD8+ T cells orchestrated the local recruitment of lymph node-resident XCR1 chemokine receptor-expressing DCs via secretion of the XCL1 chemokine. Functionally, this CD8+ T cell-mediated reorganization of the local DC network allowed for the interaction and cooperation of pDCs and XCR1+ DCs, thereby optimizing XCR1+ DC maturation and cross-presentation. These data support a model in which CD8+ T cells upon activation create their own optimal priming microenvironment by recruiting additional DC subsets to the site of initial antigen recognition.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Transgénicos
2.
J Cell Sci ; 133(5)2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31649144

RESUMEN

Dendritic cells (DCs) expressing the chemokine receptor XCR1 are specialized in antigen cross-presentation to control infections with intracellular pathogens. XCR1-positive (XCR1+) DCs are attracted by XCL1, a γ-chemokine secreted by activated CD8+ T cells and natural killer cells. Rat cytomegalovirus (RCMV) is the only virus known to encode a viral XCL1 analog (vXCL1) that competes for XCR1 binding with the endogenous chemokine. Here we show that vXCL1 from two different RCMV strains, as well as endogenous rat XCL1 (rXCL1) bind to and induce chemotaxis exclusively in rat XCR1+ DCs. Whereas rXCL1 activates the XCR1 Gi signaling pathway in rats and humans, both of the vXCL1s function as species-specific agonists for rat XCR1. In addition, we demonstrate constitutive internalization of XCR1 in XCR1-transfected HEK293A cells and in splenic XCR1+ DCs. This internalization was independent of ß-arrestin 1 and 2 and was enhanced after binding of vXCL1 and rXCL1; however, vXCL1 appeared to be a stronger agonist. These findings suggest a decreased surface expression of XCR1 during DC cultivation at 37°C, and subsequent impairment of chemotactic activity and XCR1+ DC function.


Asunto(s)
Quimiocinas C/metabolismo , Reactividad Cruzada , Células Dendríticas/inmunología , Muromegalovirus/inmunología , Receptores de Quimiocina/metabolismo , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiotaxis , Células Asesinas Naturales/inmunología , Ratas , Receptores Acoplados a Proteínas G/metabolismo
3.
Am J Respir Cell Mol Biol ; 59(4): 437-447, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29676593

RESUMEN

B-cell interaction with follicular helper T cells and subsequent differentiation of B cells into high-affinity APCs normally takes place in secondary lymphoid organs. The costimulator ICOS plays a key role in this process and is therefore considered as an attractive target to modulate exaggerated B-cell responses in autoimmune or allergic diseases. Inflamed tissues were recently recognized as additional sites of active T-cell/B-cell interaction. To analyze whether ICOS costimulation is also important there, we employed a mouse airway inflammation model that allows direct comparison of immune reactions in the lung-draining lymph node and the lung tissue as well as assessment of the relative importance of dendritic cells versus B cells as APCs. In both organs, ICOS regulated the pool size of antigen-specific T and B cells and B-cell differentiation into germinal center(-like) cells but not into antibody-secreting cells. In the lymph node, lack of ICOS costimulation drastically reduced the frequency of T follicular helper cells but did not affect production of T-helper cell type 2 (Th2) cytokines. Vice versa in the lung tissue, ICOS did not change PD-1 expression on infiltrating T cells but regulated Th2 cytokine production, a process for which ICOS ligand expression on B cells was of particular importance. Taken together, the results of this study show that ICOS differentially regulates effector T cells in secondary lymphoid organs and inflamed tissues but that blockade of the ICOS pathway is suitable to target T cell-dependent B cell responses at both sites.


Asunto(s)
Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Inflamación/patología , Tejido Linfoide/metabolismo , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Inmunoglobulina A/metabolismo , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/metabolismo , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/patología , Receptor de Muerte Celular Programada 1/metabolismo
4.
Immunity ; 31(5): 823-33, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19913446

RESUMEN

The expression of the chemokine receptor XCR1 and the function of its ligand XCL1 (otherwise referred to as ATAC, lymphotactin, or SCM-1) remained elusive to date. In the present report we demonstrated that XCR1 is exclusively expressed on murine CD8(+) dendritic cells (DCs) and showed that XCL1 is a potent and highly specific chemoattractant for this DC subset. CD8(+) T cells abundantly secreted XCL1 8-36 hr after antigen recognition on CD8(+) DCs in vivo, in a period in which stable T cell-DC interactions are known to occur. Functionally, XCL1 increased the pool of antigen-specific CD8(+) T cells and their capacity to secrete IFN-gamma. Absence of XCL1 impaired the development of cytotoxicity to antigens cross-presented by CD8(+) DCs. The XCL1-XCR1 axis thus emerges as an integral component in the development of efficient cytotoxic immunity in vivo.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Quimiocinas C/farmacología , Células Dendríticas/efectos de los fármacos , Humanos , Inmunohistoquímica , Células Asesinas Naturales/inmunología , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Bazo/citología
5.
J Immunol ; 194(3): 1069-79, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25520399

RESUMEN

Current subunit vaccines are incapable of inducing Ag-specific CD8(+) T cell cytotoxicity needed for the defense of certain infections and for therapy of neoplastic diseases. In experimental vaccines, cytotoxic responses can be elicited by targeting of Ag into cross-presenting dendritic cells (DC), but almost all available systems use target molecules also expressed on other cells and thus lack the desired specificity. In the present work, we induced CD8(+) T cell cytotoxicity by targeting of Ag to XCR1, a chemokine receptor exclusively expressed on murine and human cross-presenting DC. Targeting of Ag with a mAb or the chemokine ligand XCL1 was highly specific, as determined with XCR1-deficient mice. When applied together with an adjuvant, both vector systems induced a potent cytotoxic response preventing the outgrowth of an inoculated aggressive tumor. By generating a transgenic mouse only expressing the human XCR1 on its cross-presenting DC, we could demonstrate that targeting of Ag using human XCL1 as vector is fully effective in vivo. The specificity and efficiency of XCR1-mediated Ag targeting to cross-presenting DC, combined with its lack of adverse effects, make this system a prime candidate for the development of therapeutic cytotoxic vaccines in humans.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Cambio de Clase de Inmunoglobulina , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Receptores de Quimiocina/genética , Receptores Acoplados a Proteínas G/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Carga Tumoral
6.
J Virol ; 88(1): 292-302, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24155383

RESUMEN

Cytomegaloviruses (CMV) have developed various strategies to escape the immune system of the host. One strategy involves the expression of virus-encoded chemokines to modulate the host chemokine network. We have identified in the English isolate of rat CMV (murid herpesvirus 8 [MuHV8]) an open reading frame encoding a protein homologous to the chemokine XCL1, the only known C chemokine. Viral XCL1 (vXCL1), a glycosylated protein of 96 amino acids, can be detected 13 h postinfection in the supernatant of MuHV8-infected rat embryo fibroblasts. vXCL1 exclusively binds to CD4(-) rat dendritic cells (DC), a subset of DC that express the corresponding chemokine receptor XCR1. Like endogenous rat XCL1, vXCL1 selectively chemoattracts XCR1(+) CD4(-) DC. Since XCR1(+) DC in mice and humans have been shown to excel in antigen cross-presentation and thus in the induction of cytotoxic CD8(+) T lymphocytes, the virus has apparently hijacked this gene to subvert cytotoxic immune responses. The biology of vXCL1 offers an interesting opportunity to study the role of XCL1 and XCR1(+) DC in the cross-presentation of viral antigens.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiocinas C/metabolismo , Reactividad Cruzada , Citomegalovirus/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos Virales/inmunología , Secuencia de Bases , Linfocitos T CD4-Positivos/metabolismo , Quimiotaxis de Leucocito , Citomegalovirus/inmunología , Cartilla de ADN , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Procesamiento Proteico-Postraduccional , Ratas , Ratas Endogámicas Lew , Homología de Secuencia de Aminoácido
7.
Proc Natl Acad Sci U S A ; 109(22): 8664-9, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22552227

RESUMEN

Follicular T-helper (T(FH)) cells cooperate with GL7(+)CD95(+) germinal center (GC) B cells to induce antibody maturation. Herein, we identify the transcription factor IRF4 as a T-cell intrinsic precondition for T(FH) cell differentiation and GC formation. After immunization with protein or infection with the protozoon Leishmania major, draining lymph nodes (LNs) of IFN-regulatory factor-4 (Irf4(-/-)) mice lacked GCs and GC B cells despite developing normal initial hyperplasia. GCs were also absent in Peyer's patches of naive Irf4(-/-) mice. Accordingly, CD4(+) T cells within the LNs and Peyer's patches failed to express the T(FH) key transcription factor B-cell lymphoma-6 and other T(FH)-related molecules. During chronic leishmaniasis, the draining Irf4(-/-) LNs disappeared because of massive cell death. Adoptive transfer of WT CD4(+) T cells or few L. major primed WT T(FH) cells reconstituted GC formation, GC B-cell differentiation, and LN cell survival. In support of a T-cell intrinsic IRF4 activity, Irf4(-/-) T(FH) cell differentiation was not rescued by close neighborhood to transferred WT T(FH) cells. Together with its known B lineage-specific roles during plasma cell maturation and class switch, our study places IRF4 in the center of antibody production toward T-cell-dependent antigens.


Asunto(s)
Diferenciación Celular/inmunología , Centro Germinal/inmunología , Factores Reguladores del Interferón/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Supervivencia Celular/inmunología , Femenino , Citometría de Flujo , Expresión Génica , Centro Germinal/citología , Centro Germinal/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interleucinas/genética , Interleucinas/inmunología , Interleucinas/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/trasplante
8.
Eur J Immunol ; 43(12): 3233-43, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23963995

RESUMEN

Attenuation of T cell-mediated damage of blood endothelial cells (BECs) in transplanted organs is important to prevent transplant vasculopathy (TV) and chronic rejection. Here, we assessed the importance of minor histocompatibility antigen (mHA) distribution and different coinhibitory molecules for T cell-BEC interaction. A transgenic mHA was directed specifically to BECs using the Tie2 promoter and cellular interactions were assessed in graft-versus-host disease-like and heterotopic heart transplantation settings. We found that cognate CD4(+) T-cell help was critical for the activation of BEC-specific CD8(+) T cells. However, systemic mHA expression on BECs efficiently attenuated adoptively transferred, BEC-specific CD4(+) and CD8(+) T cells and hence prevented tissue damage, whereas restriction of mHA expression to heart BECs precipitated the development of TV. Importantly, the lack of the coinhibitory molecules programmed death-1 (PD-1) and B and T lymphocyte attenuator fostered the initial activation of BEC-specific CD4(+) T cells, but did not affect development of TV. In contrast, TV was significantly augmented in the absence of PD-1 on BEC-specific CD8(+) T cells. Taken together, these results indicate that antigen distribution in the vascular bed determines the impact of coinhibition and, as a consequence, critically impinges on T cell-mediated vascular immunopathology.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Endoteliales/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón , Antígenos de Histocompatibilidad Menor/inmunología , Enfermedades Vasculares/inmunología , Aloinjertos , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/biosíntesis , Antígenos de Histocompatibilidad Menor/genética , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
9.
J Immunol ; 189(1): 234-44, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661090

RESUMEN

We have established a comprehensive in vivo mouse model for the CD4(+) T cell response to an "innocuous" versus "dangerous" exogenous Ag and developed an in vivo test for tolerance. In this model, specific gene-expression signatures, distinctive upregulation of early T cell-communication molecules, and differential expansion of effector T cells (Teff) and regulatory T cells (Treg) were identified as central correlates of T cell tolerance and T cell immunity. Different from essentially all other T cell-activation molecules, ICOS was found to be induced in the immunity response and not by T cells activated under tolerogenic conditions. If expressed, ICOS did not act as a general T cell costimulator but selectively caused a massive expansion of effector CD4(+) T cells, leaving the regulatory CD4(+) T cell compartment largely undisturbed. Thus, ICOS strongly contributed to the dramatic change in the balance between Ag-specific Teff and Treg from ∼1:1 at steady state to 21:1 at the height of the immune response. This newly defined role for the balance of Teff to Treg, together with its known key function in T cell help for B cells, establishes ICOS as a central mediator of immunity. Given its exceptionally selective induction on CD4(+) T cells under inflammatory, but not tolerogenic, conditions, ICOS emerges as a pivotal effector molecule in the early decision between tolerance and immunity to exogenous Ag.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/fisiología , Inmunidad Adaptativa/genética , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Tolerancia Inmunológica/genética , Proteína Coestimuladora de Linfocitos T Inducibles/deficiencia , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
10.
J Am Soc Nephrol ; 24(4): 543-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411785

RESUMEN

Although the spleen is a major site where immune tolerance to circulating innocuous antigens occurs, the kidney also contributes. Circulating antigens smaller than albumin are constitutively filtered and concentrated in the kidney and reach the renal lymph node by lymphatic drainage, where resident dendritic cells (DCs) capture them and induce tolerance of specific cytotoxic T cells through unknown mechanisms. Here, we found that the coinhibitory cell surface receptor programmed death 1 (PD-1) on cytotoxic T cells mediates to their tolerance. Renal lymph node DCs of the CD8(+) XCR1(+) subset, which depend on the transcription factor Batf3, expressed the PD-1 cognate ligand PD-L1. Batf3-dependent DCs in the renal lymph node presented antigen that had been concentrated in the kidney and used PD-L1 to induce apoptosis of cytotoxic T cells. In contrast, T cell tolerance in the spleen was independent of PD-1, PD-L1, and Batf3. In summary, these results clarify how the kidney/renal lymph node system tolerizes the immune system against circulating innocuous antigens.


Asunto(s)
Antígenos/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Riñón/inmunología , Ganglios Linfáticos/inmunología , Proteínas Represoras/metabolismo , Animales , Antígenos/sangre , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Células Dendríticas/metabolismo , Riñón/metabolismo , Ganglios Linfáticos/metabolismo , Ratones , Ratones Mutantes , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Represoras/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
11.
J Immunol ; 184(2): 550-4, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20008296

RESUMEN

Proinflammatory Th1 cells can produce large amounts of the immunosuppressive cytokine IL-10, thereby facilitating the self-limitation of inflammatory responses. Recently, we identified the Notch pathway as a main regulator of IL-10 production by Th1 cells. In this study, we show that plasmacytoid dendritic cells (pDCs), by means of their unique high-level expression of the Notch ligand Delta-like (Dll)-4, activate the Notch receptor on T cells to induce robust IL-10 production in vitro and in vivo. pDCs display a distinct pattern of Notch ligands compared with conventional dendritic cells, marked by the constitutive expression of Dll-4, the only Notch ligand to induce IL-10 expression in vivo, and Dll-1, while at the same time lacking the expression of Jagged. We provide a new mechanism for IL-10 induction by pDCs underlining the importance of the Dll-4/Notch axis in the regulation of inflammatory T cell responses.


Asunto(s)
Células Dendríticas/inmunología , Interleucina-10/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Inflamación , Mediadores de Inflamación/metabolismo , Ligandos , Ratones , Ratones Endogámicos BALB C
12.
J Immunol ; 184(4): 1810-20, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20083650

RESUMEN

Foxp3(+)CD4(+) regulatory T cells (Tregs) are crucial in maintaining self-tolerance and limiting immune responses to pathogens. Shifting the sensitive balance between Tregs and effector T cells requires extensive knowledge of the homeostatic properties of the different T cell populations. For the investigation of Treg homeostatic expansion, we introduce in this study novel BAC transgenic mice, designated Foxp3.LuciDTR, coexpressing enhanced GFP, luciferase for bioluminescence imaging of Tregs, and the diphtheria toxin receptor (DTR) for specific ablation of Tregs. Of several founder lines, Foxp3.LuciDTR-4 mice displayed approximately 95% Treg depletion following injection of DT, resulting in activation of conventional CD4(+) T cells, probably due to lack of control by Tregs. In contrast, Foxp3.LuciDTR-3 mice displayed only approximately 70% Treg depletion without concomitant activation of CD4(+) T cells and represented, therefore, a suitable model to study Treg homeostasis in an environment where other T cell populations were not altered. After depletion, the Treg compartment recovered to its original size in approximately 2 wk. This recovery was mediated in a thymus-independent fashion by homeostatic proliferation of the surviving, nondepleted Tregs. The proliferating Tregs acquired an activated phenotype and maintained their suppressive capacity. Studies involving DT-mediated depletion of dendritic cells in CD11c.DOG mice showed that dendritic cells were required for optimal Treg homeostasis. In addition, IL-2 was identified as an essential factor for homeostatic recovery of the Treg compartment. These results show that Treg homeostasis is specifically regulated by the size of the Treg compartment and is independent of proliferation of conventional T cells.


Asunto(s)
Proliferación Celular , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Homeostasis/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Toxina Diftérica/genética , Factores de Transcripción Forkhead/genética , Homeostasis/genética , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/metabolismo
13.
J Immunol ; 182(6): 3349-56, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265111

RESUMEN

Tolerogenic dendritic cells (DC) play an important role in maintaining peripheral T cell tolerance in steady-state conditions through induction of anergic, IL-10-producing T cells with suppressive properties. ICOS, an activation-induced member of the CD28 family on T cells, is involved in the induction of IL-10, which itself could contribute to induction of anergy and development of suppressive T cells. Therefore, we analyzed the functional role of ICOS in the differentiation process of human CD4(+) T cells upon their interaction with tolerogenic DC. We compared the functional properties of CD4(+) T cells from healthy volunteers and ICOS-deficient patients after stimulation with tolerogenic DC. We report that induction of T cell anergy and suppressive capacity is completely blocked after knockdown of ICOS expression in T cells as well as after blocking of ICOS-ICOS ligand interaction in DC/T cell cocultures. Moreover, CD4(+) T cells from ICOS-deficient patients were completely resistant to anergy induction and differentiation into suppressive T cells even after supplementation of IL-10. Furthermore, ICOS/ICOS ligand interaction stabilizes IL-10R expression on T cells and thus renders them sensitive to IL-10 effects. Taken together, these results indicate a crucial role for ICOS in the induction of peripheral tolerance maintained by tolerogenic DC mediated mostly via an IL-10-independent mechanism.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/fisiología , Anergia Clonal/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Anergia Clonal/genética , Técnicas de Cocultivo , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/patología , Células Dendríticas/patología , Técnicas de Silenciamiento del Gen , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles , Interleucina-10/fisiología , Activación de Linfocitos/genética , Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
14.
J Exp Med ; 197(2): 181-93, 2003 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-12538658

RESUMEN

The studies performed to date analyzed the overall participation of the inducible costimulator (ICOS) in model diseases, but did not yield information on the nature and function of ICOS-expressing T cells in vivo. We examined ICOS(+) T cells in the secondary lymphoid organs of nonmanipulated mice, in the context of an "unbiased" immune system shaped by environmental antigens. Using single cell analysis, ICOS(low) cells were found to be loosely associated with the early cytokines interleukin (IL)-2, IL-3, IL-6, and interferon (IFN)-gamma. ICOS(medium) cells, the large majority of ICOS(+) T cells in vivo, were very tightly associated with the synthesis of the T helper type 2 (Th2) cytokines IL-4, IL-5, and IL-13, and these cells exhibited potent inflammatory effects in vivo. In contrast, ICOS(high) T cells were highly and selectively linked to the anti-inflammatory cytokine IL-10. Overall, these data seem to indicate that ICOS cell surface density serves as a regulatory mechanism for the release of cytokines with different immunological properties. Further in vivo functional experiments with in vitro-activated T cells strongly suggested that the ICOS(+) population, although representing in vivo only around 10% of T cells bearing early or late activation markers, nevertheless encompasses virtually all effector T cells, a finding with major diagnostic and therapeutic implications.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD4-Positivos/inmunología , Inflamación/etiología , Interleucina-10/biosíntesis , Traslado Adoptivo , Animales , Antiinflamatorios/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Citocinas/biosíntesis , Técnicas In Vitro , Proteína Coestimuladora de Linfocitos T Inducibles , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/inmunología , Fenotipo , Células Th2/inmunología
15.
Ann Rheum Dis ; 69(8): 1495-501, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20498202

RESUMEN

OBJECTIVES: Inducible costimulator (ICOS) and its ligand (ICOSL) regulate T and B cell responses. Glucose-6-phosphate isomerase (G6PI)-induced arthritis requires T and B lymphocytes. It was hypothesised that blocking ICOS/ICOSL interactions ameliorates G6PI-induced arthritis and reduces G6PI-specific B and T lymphocyte responses. METHODS: DBA/1 mice were injected with a blocking, non-depleting anti-ICOSL monoclonal antibodies (mAbs) during the induction or effector phase of G6PI-induced arthritis. G6PI-specific antibody responses were measured by ELISA. G6PI-specific T helper (Th) cell responses were assayed by polychromatic flow cytometry. RESULTS: Transient blockade of ICOS/ICOSL interactions profoundly reduced the severity of G6PI-induced arthritis. ELISA and proliferation assays showed no clear ex vivo correlates of protection. Polychromatic flow cytometry revealed two major findings: the absolute number of G6PI-specific Th cells was markedly diminished in secondary lymphatic organs from mice with blocked ICOS/ICOSL interactions. Within the pool of G6PI-specific Th cells the frequency of interleukin 17 (IL17), interferon gamma or tumour necrosis factor alpha producers or polyfunctional Th cells (expressing two or more of these cytokines) was higher in treated than in control mice. CONCLUSIONS: ICOS costimulation is not mandatory for the differentiation of Th1 or Th17 cells. Instead, the lack of ICOS costimulation results in reduced survival of G6PI-specific Th cells irrespective of their functional differentiation. This study demonstrates that a thorough examination of the quantity and the quality of antigen-specific immune responses is useful to determine ex vivo correlates of efficacy for immunomodulating treatments.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Antígenos de Diferenciación de Linfocitos T/inmunología , Artritis Experimental/prevención & control , Artritis Reumatoide/prevención & control , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Diferenciación Celular/inmunología , Citocinas/biosíntesis , Glucosa-6-Fosfato/inmunología , Inmunoglobulinas/biosíntesis , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Interleucina-17/análisis , Ligandos , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos DBA , Proteínas/inmunología , Células TH1/inmunología
16.
MAbs ; 12(1): 1834818, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33151105

RESUMEN

Simulating a viral infection in tumor cells is an attractive concept to eliminate tumor cells. We previously reported the molecular design and the in vitro potency of recombinant monoclonal antibodies fused to a virus-derived peptide MHC class I complex that bypass the peptide processing and MHC loading pathway and directly displays a viral peptide in an MHC class I complex on the tumor cell surface. Here, we show that a vaccination-induced single peptide-specific CD8 T cell response was sufficient to eliminate B16 melanoma tumor cells in vivo in a fully immunocompetent, syngeneic mouse tumor model when mice were treated with mouse pMHCI-IgGs fusion proteins targeting the mouse fibroblast activation protein. Tumor growth of small, established B16 lung metastases could be controlled. The pMHCI-IgG had similar potency as an analogous pan-CD3 T-cell bispecific antibody. In contrast to growth control of small tumors, none of the compounds controlled larger solid tumors of MC38 cancer cells, despite penetration of pMHCI-IgGs into the tumor tissue and clear attraction and activation of antigen-specific CD8 T cells inside the tumor. pMHCI-IgG can have a similar potency as classical pan-T-cell recruiting molecules. The results also highlight the need to better understand immune suppression in advanced solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/inmunología , Melanoma Experimental/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/inmunología
17.
Front Immunol ; 9: 2806, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619244

RESUMEN

XCL1 is the ligand for XCR1, a chemokine receptor uniquely expressed on cross-presenting dendritic cells (DC) in mouse and man. We are interested in establishing therapeutic vaccines based on XCL1-mediated targeting of peptides or proteins into these DC. Therefore, we have functionally analyzed various XCL1 domains in highly relevant settings in vitro and in vivo. Murine XCL1 fused to ovalbumin (XCL1-OVA) was compared to an N-terminal deletion variant lacking the first seven N-terminal amino acids and to several C-terminal (deletion) variants. Binding studies with primary XCR1+ DC revealed that the N-terminal region stabilizes the binding of XCL1 to its receptor, as is known for other chemokines. Deviating from the established paradigm for chemokines, the N-terminus does not contain critical elements for inducing chemotaxis. On the contrary, this region appears to limit the chemotactic action of XCL1 at higher concentrations. A participation of the XCL1 C-terminus in receptor binding or chemotaxis could be excluded in a series of experiments. Binding studies with apoptotic and necrotic XCR1-negative cells suggested a second function for XCL1: marking of stressed cells for uptake into cross-presenting DC. In vivo studies using CD8+ T cell proliferation and cytotoxicity as readouts confirmed the critical role of the N-terminus for antigen targeting, and excluded any involvement of the C-terminus in the uptake, processing, and presentation of the fused OVA antigen. Together, these studies provide basic data on the function of the various XCL1 domains as well as relevant information on XCL1 as an antigen carrier in therapeutic vaccines.


Asunto(s)
Quimiocinas C , Células Dendríticas/inmunología , Portadores de Fármacos , Ovalbúmina , Receptores de Quimiocina/inmunología , Proteínas Recombinantes de Fusión , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Quimiocinas C/química , Quimiocinas C/genética , Quimiocinas C/farmacología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Células Dendríticas/citología , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Ratones , Ratones Transgénicos , Ovalbúmina/química , Ovalbúmina/genética , Ovalbúmina/farmacología , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Relación Estructura-Actividad , Vacunas/química , Vacunas/genética , Vacunas/farmacología
18.
Curr Opin Immunol ; 16(3): 321-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15134781

RESUMEN

The analysis of recent data reveals that T-cell co-stimulation is a hierarchical process with elements of mutual interdependence between individual co-stimulators. The expression and function of co-stimulatory molecules is biased on various T-cell subsets and is dependent on the T-cell differentiation state. The classical paradigm of T-cell co-stimulation by professional antigen-presenting cells has to incorporate the newly recognized concept of T-cell co-stimulation in the interaction with peripheral tissues, such as endothelial or epithelial cells. The two signal paradigm of T-cell co-stimulation is being replaced by a multisignal integration concept of central and peripheral co-stimulation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Superficie/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Ratones , Transducción de Señal/inmunología
19.
Nat Commun ; 7: 10875, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26915335

RESUMEN

Autoimmune diseases and other inflammatory conditions are characterized by large lymphocytic tissue infiltrates in which T and B cells can be found in close contact. Here, using a murine airway inflammation model, we compare antigen-specific T and B cells in lung tissue versus lung-draining lymph node. In the lung we identify a B-cell population exhibiting a classical germinal centre phenotype without being organized into ectopic lymphoid tissue. By contrast, classical CXCR5(+) Bcl-6(+) T follicular helper cells are not present. Nevertheless, lung-infiltrating T cells exhibit follicular helper-like properties including the potential to provide help to naive B cells. The lung tissue is also a survival niche for memory T and B cells remaining in residual peribronchial infiltrates after resolution of inflammation. Collectively, this study shows the importance of T/B cooperation not only in lymph nodes but also in inflamed peripheral tissues for local antibody responses to infection and autoimmunity.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Pulmón/inmunología , Cooperación Linfocítica/inmunología , Neumonía/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Formación de Anticuerpos/inmunología , Autoinmunidad/inmunología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamación , Ganglios Linfáticos/inmunología , Tejido Linfoide/inmunología , Ratones , Ratones Transgénicos , Ovalbúmina/toxicidad , Neumonía/inducido químicamente , Proteínas Proto-Oncogénicas c-bcl-6 , Receptores de Antígenos de Linfocitos T/genética , Receptores CXCR5
20.
Clin Transl Immunology ; 4(10): e47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26682055

RESUMEN

Dendritic cells (DC) in the thymus have an important role in the establishment of central tolerance by promoting negative selection of autoreactive T cells and regulatory T-cell differentiation. Whereas human DC have recently been studied in various tissues in more detail, thymic DC subsets are still ill-defined. In the present work, we studied the binding of 71 monoclonal antibodies (mAb) submitted to the HLDA10 workshop to human CD123(+) plasmacytoid DC and the two subsets of conventional DC (cDC, CD141(+) and CD11b(+)) isolated from thymus tissue of infants undergoing corrective heart surgery. Within the panel, we found mAb binding to thymic pDC and both cDC subsets (for example, anti-Clec12A, TIM-3, Clec4A, CCR5, Axl, FLT3), but most of them additionally reacted with other thymic cell types. MAb directed to CD85h (ILT1) and the C-type lectin Clec7A (now CD369) reacted selectively with both cDC subsets, but not with other cells. Only one mAb directed to CD85g (ILT7) stained thymic pDC in a highly specific manner. Clec9A (DNGR1, now CD370) was the only tested HLDA10 antigen exclusively expressed on thymic CD141(+) cDC. The present report summarizes all data obtained.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA