Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 862757, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35967294

RESUMEN

VISTA (PD-1H) is an immune regulatory molecule considered part of the next wave of immuno-oncology targets. VISTA is an immunoglobulin (Ig) superfamily cell surface molecule mainly expressed on myeloid cells, and to some extent on NK cells and T cells. In previous preclinical studies, some VISTA-targeting antibodies provided immune inhibitory signals, while other antibodies triggered immune stimulatory signals. Importantly, for therapeutic antibodies, the isotype backbone can have a strong impact on antibody function. To elucidate the mode of action of immune stimulatory anti-VISTA antibodies, we studied three different anti-human VISTA antibody clones, each on three different IgG isotypes currently used for therapeutic antibodies: unaltered IgG1 (IgG1-WT), IgG1-KO (IgG1-LL234,235AA-variant with reduced Fc-effector function), and IgG4-Pro (IgG4- S228P-variant with stabilized hinge region). Antibody functionality was analysed in mixed leukocyte reaction (MLR) of human peripheral blood mononuclear cells (PBMCs), as a model system for ongoing immune reactions, on unstimulated human PBMCs, as a model system for a resting immune system, and also on acute myeloid leukemia (AML) patient samples to evaluate anti-VISTA antibody effects on primary tumor material. The functions of three anti-human VISTA antibodies were determined by their IgG isotype backbones. An MLR of healthy donor PBMCs was effectively augmented by anti-VISTA-IgG4-Pro and anti-VISTA-IgG1-WT antibodies, as indicated by increased levels of cytokines, T cell activation markers and T cell proliferation. However, in a culture of unstimulated PBMCs of single healthy donors, only anti-VISTA-IgG1-WT antibodies increased the activation marker HLA-DR on resting myeloid cells, and chemokine levels. Interestingly, interactions with different Fc-receptors were required for these effects, namely CD64 for augmentation of MLR, and CD16 for activation of resting myeloid cells. Furthermore, anti-VISTA-IgG1-KO antibodies had nearly no impact in any model system. Similarly, in AML patient samples, anti-VISTA-antibody on IgG4-Pro backbone, but not on IgG1-KO backbone, increased interactions, as a novel readout of activity, between immune cells and CD34+ AML cancer cells. In conclusion, the immune stimulatory effects of antagonistic anti-VISTA antibodies are defined by the antibody isotype and interaction with different Fc-gamma-receptors, highlighting the importance of understanding these interactions when designing immune stimulatory antibody therapeutics for immuno-oncology applications.


Asunto(s)
Antígenos B7/inmunología , Neoplasias , Receptores Fc , Humanos , Inmunoglobulina G , Leucocitos Mononucleares , Receptores de IgG
2.
Oncoimmunology ; 11(1): 2080328, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35756842

RESUMEN

Upregulation of inhibitory receptors, such as lymphocyte activation gene-3 (LAG-3), may limit the antitumor activity of therapeutic antibodies targeting the programmed cell death protein-1 (PD-1) pathway. We describe the binding properties of ezabenlimab, an anti-human PD-1 antibody, and BI 754111, an anti-human LAG-3 antibody, and assess their activity alone and in combination. Ezabenlimab bound with high affinity to human PD-1 (KD = 6 nM) and blocked the interaction of PD-1 with PD-L1 and PD-L2. Ezabenlimab dose-dependently increased interferon-γ secretion in human T cells expressing PD-1 in co-culture with PD-L1-expressing dendritic cells. Administration of ezabenlimab to human PD-1 knock-in mice dose-dependently inhibited growth of MC38 tumors. To reduce immunogenicity, ezabenlimab was reformatted from a human IgG4 to a chimeric variant with a mouse IgG1 backbone (BI 905725) for further in vivo studies. Combining BI 905725 with anti-mouse LAG-3 antibodies improved antitumor activity versus BI 905725 monotherapy in the MC38 tumor model. We generated BI 754111, which bound with high affinity to human LAG-3 and prevented LAG-3 interaction with its ligand, major histocompatibility complex class II. In an in vitro model of antigen-experienced memory T cells expressing PD-1 and LAG-3, interferon-γ secretion increased by an average 1.8-fold versus isotype control (p = 0.027) with BI 754111 monotherapy, 6.9-fold (p < 0.0001) with ezabenlimab monotherapy and 13.2-fold (p < 0.0001) with BI 754111 plus ezabenlimab. Overall, ezabenlimab and BI 754111 bound to their respective targets with high affinity and prevented ligand binding. Combining ezabenlimab with BI 754111 enhanced in vitro activity versus monotherapy, supporting clinical investigation of this combination (NCT03156114; NCT03433898).


Asunto(s)
Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Animales , Anticuerpos Bloqueadores , Anticuerpos Monoclonales/farmacología , Estudios Clínicos como Asunto , Inhibidores de Puntos de Control Inmunológico , Interferón gamma , Ligandos , Ratones
3.
Mol Cancer Ther ; 20(11): 2250-2261, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34482286

RESUMEN

Despite some impressive clinical results with immune checkpoint inhibitors, the majority of patients with cancer do not respond to these agents, in part due to immunosuppressive mechanisms in the tumor microenvironment. High levels of adenosine in tumors can suppress immune cell function, and strategies to target the pathway involved in its production have emerged. CD73 is a key enzyme involved in adenosine production. This led us to identify a novel humanized antagonistic CD73 antibody, mAb19, with distinct binding properties. mAb19 potently inhibits the enzymatic activity of CD73 in vitro, resulting in an inhibition of adenosine formation and enhanced T-cell activation. We then investigated the therapeutic potential of combining CD73 antagonism with other immune modulatory and chemotherapeutic agents. Combination of mAb19 with a PD-1 inhibitor increased T-cell activation in vitro Interestingly, this effect could be further enhanced with an agonist of the adenosine receptor ADORA3. Adenosine levels were found to be elevated upon doxorubicin treatment in vivo, which could be blocked by CD73 inhibition. Combining CD73 antagonism with doxorubicin resulted in superior responses in vivo Furthermore, a retrospective analysis of rectal cancer patient samples demonstrated an upregulation of the adenosine pathway upon chemoradiation, providing further rationale for combining CD73 inhibition with chemotherapeutic agents.This study demonstrates the ability of a novel CD73 antibody to enhance T-cell function through the potent suppression of adenosine levels. In addition, the data highlight combination opportunities with standard of care therapies as well as with an ADORA3 receptor agonist to treat patients with solid tumors.


Asunto(s)
5'-Nucleotidasa/antagonistas & inhibidores , Adenosina/uso terapéutico , Terapia de Inmunosupresión/métodos , Adenosina/farmacología , Animales , Femenino , Humanos , Ratones , Microambiente Tumoral
4.
Cancer Immunol Res ; 7(11): 1891-1899, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31484656

RESUMEN

Drugs targeting the programmed cell death protein 1 (PD-1) pathway are approved as therapies for an increasing number of cancer entities, including renal cell carcinoma. Despite a significant increase in overall survival, most treated patients do not show durable clinical responses. A combination of checkpoint inhibitors could provide a promising improvement. The aim of the study was to determine the most promising checkpoint blockade combination for renal cell carcinoma patients. Tumor-infiltrating lymphocytes (TIL) and autologous peripheral blood mononuclear cells (PBMC) were isolated from patients undergoing surgery for primary tumors. Cells were stained for multicolor flow cytometry to determine the (co)expression of five inhibitory receptors (iR), PD-1, LAG-3, Tim-3, BTLA, and CTLA-4, on T-cell populations. The function of these TILs was assessed by intracellular cytokine staining after in vitro stimulation in the presence or absence of PD-1 ± LAG-3 or Tim-3-specific antibodies. Although the percentage of iR+ T cells was low in PBMCs, both CD4+ and CD8+ T cells showed increased frequencies of PD-1+, LAG-3+, and Tim-3+ cells on TILs. The most frequent iR combination was PD-1 and LAG-3 on both CD4+ and CD8+ TILs. Blockade of PD-1 resulted in significant LAG-3, but not Tim-3, upregulation. The dual blockade of PD-1 and LAG-3, but not PD-1 and Tim-3, led to increased IFNγ release upon in vitro stimulation. Together, these data suggest that dual blockade of PD-1 and LAG-3 is a promising checkpoint blockade combination for renal cell carcinoma.


Asunto(s)
Antígenos CD/inmunología , Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Subgrupos de Linfocitos T/inmunología , Anciano , Anticuerpos Bloqueadores/inmunología , Antígenos CD/metabolismo , Carcinoma de Células Renales/patología , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Femenino , Humanos , Interferón gamma/metabolismo , Neoplasias Renales/patología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/metabolismo , Subgrupos de Linfocitos T/metabolismo , Proteína del Gen 3 de Activación de Linfocitos
5.
Front Immunol ; 8: 1471, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163535

RESUMEN

TNF receptor type 2 (TNFR2) has gained attention as a costimulatory receptor for T cells and as critical factor for the development of regulatory T cells (Treg) and myeloid suppressor cells. Using the TNFR2-specific agonist TNCscTNF80, direct effects of TNFR2 activation on myeloid cells and T cells were investigated in mice. In vitro, TNCscTNF80 induced T cell proliferation in a costimulatory fashion, and also supported in vitro expansion of Treg cells. In addition, activation of TNFR2 retarded differentiation of bone marrow-derived immature myeloid cells in culture and reduced their suppressor function. In vivo application of TNCscTNF80-induced mild myelopoiesis in naïve mice without affecting the immune cell composition. Already a single application expanded Treg cells and improved suppression of CD4 T cells in mice with chronic inflammation. By contrast, multiple applications of the TNFR2 agonist were required to expand Treg cells in naïve mice. Improved suppression of T cell proliferation depended on expression of TNFR2 by T cells in mice repeatedly treated with TNCscTNF80, without a major contribution of TNFR2 on myeloid cells. Thus, TNFR2 activation on T cells in naïve mice can lead to immune suppression in vivo. These findings support the important role of TNFR2 for Treg cells in immune regulation.

6.
Immun Inflamm Dis ; 3(4): 374-85, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26734459

RESUMEN

The immune system in sepsis is impaired as seen by reduced numbers and function of immune cells and impaired antigen-specific antibody responses. We studied T cell function in septic mice using cecal ligation and puncture (CLP) as a clinically relevant mouse model for sepsis. The proliferative response of CD4(+) and CD8(+) T cells was suppressed in septic mice. Adoptive transfer experiments demonstrated that the T cells were not intrinsically altered by CLP. Instead, the septic host environment was responsible for this T cell suppression. While CLP-induced suppression was dependent on TNF activity, neither the activation of TNF receptors type 1 nor TNF receptor type 2 alone was sufficient to generate sepsis-induced suppression showing that the two TNF receptors can substitute each other. Specific depletion of regulatory T (Treg) cells improved the impaired T cell proliferation in septic recipients demonstrating participation of Treg in sepsis-induced suppression. In summary, sepsis leads to TNF-dependent suppression of T cell proliferation in vivo involving induction of Treg cells.

7.
Immun Inflamm Dis ; 2(2): 121-30, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25400932

RESUMEN

TNF and TNF receptor type 2 (TNFR2) have been shown to be important for generation of myeloid-derived suppressor cells (MDSC). In order to analyze whether and how TNFR2 passes the effect of TNF on, myeloid cells from TNFR2-deficient mice were compared to respective cells from wild-type mice. Primary TNFR2-deficient myeloid cells showed reduced production of NO and IL-6 which was attributable to CD11b(+) CD11c(-) Ly6C(+) Ly6G(-) immature monocytic MDSC. TNFR2-deficient MDSC isolated from bone marrow were less suppressive for T cell proliferation compared to WT-derived MDSC. These differences on myeloid cells between the two mouse lines were still observed after co-culture of bone marrow cells from the two mouse lines together during myeloid cell differentiation, which demonstrated that the impaired functional capacity of TNFR2-deficient cells was independent of soluble factors but required membrane expression of TNFR2. Similarly, adoptive transfer of TNFR2-deficient bone marrow cells into wild-type hosts did not rescue the TNFR2-specific phenotype of bone marrow-derived myeloid cells. Therefore, membrane TNFR2 expression determines generation and function of monocytic MDSC.

8.
Innate Immun ; 20(7): 712-20, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24122297

RESUMEN

Sepsis-induced immune reactions are reduced in TNF receptor 2 (TNFR2)-deficient mice as previously shown. In order to elucidate the underlying mechanisms, the functional integrity of myeloid cells of TNFR2-deficient mice was analyzed and compared to wild type (WT) mice. The capacity of dendritic cells to produce IL-12 was strongly impaired in TNF-deficient mice, mirroring impaired production of IL-12 by WT dendritic cells in sepsis or after LPS or TNF pre-treatment. In addition, TNFR2-deficient mice were refractory to LPS pre-treatment and also to hyper-sensitization by inactivated Propionibacterium acnes, indicating habituation to inflammatory stimuli by the immune response when TNFR2 is lacking. Constitutive expression of TNF mRNA in kidney, liver, spleen, colon and lung tissue, and the presence of soluble TNFR2 in urine of healthy WT mice supported the conclusion that TNF is continuously present in naïve mice and controlled by soluble TNFR2. In TNFR2-deficient mice endogenous TNF levels cannot be balanced and the continuous exposure to enhanced TNF levels impairs dendritic cell function. In conclusion, TNF pre-exposure suppresses secondary inflammatory reactions of myeloid cells; therefore, continuous control of endogenous TNF by soluble TNFR2 seems to be essential for the maintenance of adequate sensitivity to inflammatory stimuli.


Asunto(s)
Células Dendríticas/metabolismo , Inflamación/metabolismo , Interleucina-12/biosíntesis , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Presentadoras de Antígenos , Citocinas/biosíntesis , Células Dendríticas/efectos de los fármacos , Citometría de Flujo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Propionibacterium acnes/metabolismo
9.
Eur Cytokine Netw ; 23(1): 15-20, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22449555

RESUMEN

In an experimental model of immune-complex-mediated glomerulonephritis, mice excreted increased levels of urinary protein starting three days after the induction. Mice lacking the TNF receptor type 2 (TNFR2) were protected from early proteinuria and enhanced mortality. Analysis of the molecular basis of the mechanisms of glomerulonephritis revealed that naïve mice continuously excrete soluble TNF-neutralizing TNFR2 in urine. Mice kept in a specific pathogen-free environment did not go on to develop early proteinuria or enhanced mortality, following induction of glomerulonephritis. TNFR2-deficient mice were protected from early proteinuria and enhanced mortality only when housed conventionally. Mice producing human TNFR2 that can be activated by mouse TNF, in addition to mouse TNFR2, did not demonstrate enhanced susceptibility to the lethal effects of glomerulonephritis, indicating that pro-inflammatory signalling via TNFR2 does not account for a sensitizing effect. Finally, we suggest that the protective effect seen in mice lacking TNFR2 results rather from environment-induced attenuation by low dose bacterial endotoxins than from missing pro-inflammatory signalling via the TNFR2.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Glomerulonefritis/inmunología , Riñón/patología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Análisis de Varianza , Animales , Anticuerpos/efectos adversos , Complejo Antígeno-Anticuerpo/metabolismo , Creatinina/sangre , Creatinina/orina , Membrana Basal Glomerular/inmunología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteinuria/orina , Conejos , Receptores Tipo II del Factor de Necrosis Tumoral/sangre , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/orina , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/orina
10.
Eur Cytokine Netw ; 23(1): 12-4, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22449588

RESUMEN

Proteinuria represents a parameter for a damaged filtration capacity of the kidney. We investigated how inflammation influences the development of experimental, immune complex-mediated glomerulonephritis by monitoring proteinuria. Mice pre-treated with LPS or TNF, one day before induction of glomerulonephritis, excreted high levels of protein in the urine immediately after the induction of glomerulonephritis, in contrast to non-treated mice where proteinuria increased steadily after day 3. Protein levels in the urine of pre-treated mice remained elevated over the 15-day observation time. The severity of proteinuria at later times correlated with the degree of tissue pathology and mortality in individual mice. Pre-treatment with inflammatory agents accelerated the development of proteinuria and induced more severe kidney damage.


Asunto(s)
Glomerulonefritis/inmunología , Riñón/patología , Lipopolisacáridos/inmunología , Proteinuria/orina , Factor de Necrosis Tumoral alfa/inmunología , Análisis de Varianza , Animales , Anticuerpos/efectos adversos , Creatinina/sangre , Creatinina/orina , Membrana Basal Glomerular/inmunología , Glomerulonefritis/inducido químicamente , Glomerulonefritis/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Conejos , Proteínas Recombinantes , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Eur J Immunol ; 42(2): 341-52, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22105154

RESUMEN

Immunosuppression, impaired cytokine production and high susceptibility to secondary infections are characteristic for septic patients, and for mice after induction of polymicrobial septic peritonitis by sublethal cecal ligation and puncture (CLP). Here, we demonstrate that CLP markedly altered subsequent B-cell responses. Total IgG and IgM levels, as well as the memory B-cell response, were increased in septic mice, but antigen-specific primary antibody production was strongly impaired. We found that two days after CLP, CD11b(+) splenocytes were activated as demonstrated by the increased expression of activation markers, expression of arginase and production of NO by immature myeloid cells. The in vivo clearance of a bacterial infection was not impaired. DCs demonstrated reduced IL-12 production and altered antigen presentation, resulting in decreased proliferation but enhanced IFN-γ production by CD4(+) cells. CD4(+) T cells from mice immunized on day 2 after CLP showed reduced Th1 and Th2 cytokine production. In addition, there was an increase in Treg cells. Interestingly, levels of immature B cells decreased but levels of mature B cells increased two days after CLP. However, adoptive transfer of naïve CD4(+) T cells, naïve B cells, or naïve DCs did not rescue the antigen-specific antibody response.


Asunto(s)
Linfocitos B/metabolismo , Ciego/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Sepsis/inmunología , Traslado Adoptivo , Animales , Formación de Anticuerpos , Presentación de Antígeno , Linfocitos B/inmunología , Linfocitos B/patología , Antígeno CD11b/metabolismo , Antígenos CD4/metabolismo , Ciego/microbiología , Ciego/patología , Ciego/cirugía , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Epítopos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/patología , Infecciones por Escherichia coli/fisiopatología , Humanos , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Peritonitis , Sepsis/complicaciones , Sepsis/patología , Sepsis/fisiopatología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
12.
J Allergy Clin Immunol ; 124(5): 1070-7.e1-11, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19665781

RESUMEN

BACKGROUND: The spread of type I allergic diseases has reached epidemic dimensions. The success of therapeutic intervention is limited, and hence prophylactic vaccination is now seriously considered. However, immunization of healthy individuals requires safety standards far beyond those applicable for therapeutic approaches. mRNAs encoding allergen molecules represent an attractive tool for preventive vaccination because of the inherent safety features of this vaccine type. OBJECTIVE: In the current study we investigated whether mRNA constructs would be capable of protecting against type I allergic reactions in a murine model using the grass pollen allergen Phl p 5 and 28 other major pollen, food, animal, mold, and latex allergens. METHODS: BALB/c mice were immunized intradermally either with conventional or replicase-based mRNA constructs. Subsequently, animals were sensitized by means of subcutaneous injection of allergen/alum, followed by airway provocation. IgG1/IgG2a/IgE titers were determined by using ELISAs. Allergen-specific functional IgE levels were assessed by using the basophil release assay. Measurement of cytokines in splenocyte cultures and bronchoalveolar lavage fluids were performed by using enzyme-linked immunosorbent spot assays/sandwich ELISAs. Eosinophil and CD8(+) counts in bronchoalveolar lavage specimens were determined by means of flow cytometry. Airway hyperreactivity was assessed with whole-body plethysmography and invasive resistance/dynamic compliance measurement. RESULTS: mRNA vaccination proved its antiallergic efficacy in terms of IgG subclass distribution, functional IgE suppression, reduction of IL-4 and IL-5 levels, induction of IFN-gamma-producing cells, and reduction of airway hyperreactivity and eosinophil counts in the lung. CONCLUSION: Immunization with mRNA induces T(H)1-biased immune responses similar to those elicited through DNA-based vaccination but additionally offers the advantage of a superior safety profile.


Asunto(s)
Alérgenos/genética , Hipersensibilidad/prevención & control , ARN Mensajero/uso terapéutico , Vacunación/métodos , Vacunas/uso terapéutico , Alérgenos/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/inmunología , Vacunas/genética , Vacunas/inmunología
13.
Cancer Immunol Immunother ; 58(11): 1809-18, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19319531

RESUMEN

This study demonstrates that CD8+ T cells in the tumor microenvironment display reduced functionality and hyporesponsiveness. TGF-beta contributed markedly to the tumor-infiltrating CD8+ T cells' (TILs) reduced functionality, which could be reversed using a small molecule TGF-beta inhibitor. Upon T-cell receptor (TCR) activation, the activation of ITK and ERK kinases were reduced in CD8+ TILs, as compared to splenic CD8+ T cells: TGF-beta inhibitor could reverse this phenomenon. This study demonstrates for the first time the association of the Spred-1 gene, an inhibitor of the Ras/MAPK pathway, with CD8+ TILs and TGF-beta activity. Spred-1 was upregulated in CD8+ TILs and TGF-beta enhanced the expression of Spred-1 in effector/memory CD8+ T cells and not in rested/memory CD8+ T cells. Based on these findings, this study supports the hypothesis that TGF-beta mediates an inhibitory mechanism on CD8+ TILs involving TCR-signaling blockade and the upregulation of Spred-1, thus implicating Spred-1 as a potential new target for future anti-tumor immune studies.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Linfocitos Infiltrantes de Tumor/fisiología , Neoplasias Experimentales/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Proteínas Represoras/fisiología , Factor de Crecimiento Transformador beta/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Tolerancia Inmunológica , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
14.
Curr Pharm Des ; 15(7): 809-25, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19275644

RESUMEN

In recent years, complexes formed from a cytokine and antibodies against that respective cytokine (cytokine/Ab complex) have been shown to induce remarkable powerful changes in the immune system. Strong interest exists especially for complexes formed with Interleukin (IL)-2 and anti-IL-2-antibody (IL-2/Ab complex). IL-2/Ab complex activates maturation and proliferation in CD8(+) T cells and natural killer (NK) cells to a much higher degree than conventional IL-2 therapy. In addition, IL-2/Ab complex does not stimulate regulatory T cells as much as IL-2 alone. This suggests the possibility to replace the conventional IL-2 therapy with a therapy using low-dose IL-2/Ab complex. Further synthetic cytokine/Ab complexes are studied currently, including IL-3/Ab complex for its effects on the mast cell population, and IL-4/Ab complex and IL-7/Ab complex for inducing B and T cell expansion and maturation. Cytokine complexes can also be made from a cytokine and its soluble receptor. Pre-association of IL-15 with soluble IL-15 receptor alpha produces a complex with strong agonistic functions that lead to an expansion of CD8(+) T cells and NK cells. However, cytokine/Ab complexes also occur naturally in humans. A multitude of auto-antibodies to cytokines are found in human sera, and many of these auto-antibodies build cytokine/Ab complexes. This review presents naturally occurring auto-antibodies to cytokines and cytokine/Ab complexes in health and disease. It further summarizes recent research on synthetic cytokine/Ab complexes with a focus on the basic mechanisms behind the function of cytokine/Ab complexes.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Anticuerpos Monoclonales/sangre , Complejo Antígeno-Anticuerpo/sangre , Autoanticuerpos/sangre , Citocinas/sangre , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/inmunología , Citocinas/inmunología , Enfermedad/etiología , Humanos , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología
15.
J Immunol ; 182(2): 774-83, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124720

RESUMEN

The biological role of Langerin+ dendritic cells (DCs) such as Langerhans cells and a subset of dermal DCs (dDCs) in adaptive immunity against cutaneous pathogens remains enigmatic. Thus, we analyzed the impact of Langerin+ DCs in adaptive T cell-mediated immunity toward Leishmania major parasites in a Lang-DTR mouse model that allows conditional diphtheria toxin (DT)-induced ablation of Langerin+ DCs in vivo. For the first time, infection experiments with DT-treated Lang-DTR mice revealed that proliferation of L. major-specific CD8+ T cells is significantly reduced during the early phase of the immune response following depletion of Langerin+ DCs. Consequently, the total number of activated CD8+ T cells within the draining lymph node and at the site of infection is diminished. Furthermore, we show that the impaired CD8+ T cell response is due to the absence of Langerin+ dDCs and not Langerhans cells. Nevertheless, the CD4+ T cell response is not altered and the infection is cleared as effectively in DT-treated Lang-DTR mice as in control mice. This clearly demonstrates that Langerin+ DCs are, in general, dispensable for an efficient adaptive immune response against L. major parasites. Thus, we propose a novel concept that, in the experimental model of leishmaniasis, priming of CD4+ T cells is mediated by Langerin- dDCs, whereas Langerin+ dDCs are involved in early priming of CD8+ T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Células Dendríticas/clasificación , Células Dendríticas/parasitología , Leishmaniasis Cutánea/inmunología , Activación de Linfocitos/inmunología , Animales , Antígenos de Superficie/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/parasitología , Femenino , Técnicas de Sustitución del Gen/métodos , Inmunidad Innata , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Células de Langerhans/parasitología , Lectinas Tipo C/genética , Leishmania major/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/parasitología , Ganglios Linfáticos/patología , Activación de Linfocitos/genética , Lectinas de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
Vaccine ; 27(7): 987-94, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19110021

RESUMEN

Tumor-associated antigens are weakly immunogenic. Human carcinoembryonic antigen (CEA) is overexpressed on a wide range of human carcinomas and represents an attractive target for cancer immunotherapy. This study analyzes the ability of a Saccharomyces cerevisiae vector containing the transgene encoding CEA (yeast-CEA) to activate human dendritic cells (DCs) and stimulate CEA-specific T-cell responses. We demonstrate for the first time that treatment with yeast-CEA can activate human DCs, resulting in increases in surface expression of CD80, CD83, CD54, CD58, and MHC class II, and increased production by DCs of IL-12p70, TNF-alpha, IFN-gamma, IL-8, IL-2, IL-13, IL-10, and IL-1beta. We also show that human DCs treated with yeast-CEA can activate CEA-specific T-cell lines and can act as antigen-presenting cells (APCs) to generate CEA-specific T-cell lines capable of lysing CEA(+) human tumor cells. Gene expression profiles of human DCs treated with yeast-CEA show increased expression of numerous genes involved in the production of chemokines and cytokines and their receptors, and genes related to antigen uptake, antigen presentation, and signal transduction.


Asunto(s)
Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/inmunología , Células Dendríticas/inmunología , Saccharomyces cerevisiae/genética , Antígenos CD/análisis , Antígeno Carcinoembrionario/genética , Línea Celular Tumoral , Citocinas/biosíntesis , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/química , Perfilación de la Expresión Génica , Humanos , Activación de Linfocitos , Saccharomyces cerevisiae/inmunología , Linfocitos T Citotóxicos/inmunología
17.
Blood ; 112(4): 1290-8, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18541719

RESUMEN

Patent lymphatic filariasis is characterized by antigen-specific T-cell unresponsiveness with diminished IFN-gamma and IL-2 production and defects in dendritic cell (DC) function. Because Toll-like receptors (TLRs) play an important role in pathogen recognition and TLR expression is diminished on B and T cells of filaria-infected individuals, we examined the effect of live microfilariae (mf) on expression and function of TLRs in human DCs. We show that mf-exposed monocyte-derived human DCs (mhDCs) demonstrate marked diminution of TLR3 and TLR4 mRNA expression compared with mf-unexposed mhDCs that translated into loss of function in response to appropriate TLR ligands. Exposure to mf significantly down-regulated production of IFN-alpha, MIP-1alpha, IL-12p70, and IL-1alpha following activation with poly I:C, and of IL-12p40 following activation with poly I:C or LPS. mRNA expression of MyD88, the adaptor molecule involved in TLR4 signaling, was significantly diminished in mhDCs after exposure to mf. Moreover, mf interfered with NF-kappaB activation (particularly p65 and p50) following stimulation with poly I:C or LPS. These data suggest that mf interfere with mhDC function by altering TLR expression and interfering with both MyD88-dependent signaling and a pathway that ultimately diminishes NF-kappaB activity. This down-regulated NF-kappaB activity impairs mhDC-produced cytokines needed for full T-cell activation.


Asunto(s)
Células Dendríticas/parasitología , Helmintos/inmunología , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/inmunología , Regulación hacia Abajo , Humanos , Activación de Linfocitos , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/metabolismo , ARN Mensajero/análisis , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética
18.
J Immunol ; 180(7): 5118-29, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354238

RESUMEN

IL-2 is well described as a cytokine with two markedly distinct functionalities: as a necessary signal during CD4(+) and CD8(+) T cell activation/expansion and as an essential cytokine for the maintenance of CD4(+)CD25(+)FoxP3(+) T cells (regulatory T (T(REG)) cells) during homeostasis. In this study we demonstrate for the first time that, compared with the use of IL-2 alone, a complex of IL-2 and anti-IL-2 Ab (IL-2 complex) enhances the effectiveness of a viral vaccine in a mouse model with known Ag specificity. IL-2 complex led to an increase in the number of Ag-specific effector/memory CD8(+) T cells, cytokine production, and CTL lysis following Ag-specific restimulation in a vaccination setting. Our results further demonstrate that this effect is temporary and declines over the course of a few days after the IL-2 complex treatment cycle. Moreover, in contrast to the use of IL-2 alone, IL-2 complex greatly increased the ratio of effector/memory CD8(+) T cells to T(REG) cells. This phenomenon can thus potentially be used in the enhancement of immune responses to vaccination.


Asunto(s)
Anticuerpos/inmunología , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Interleucina-2/inmunología , Linfocitos T Reguladores/inmunología , Vacunas/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Ratones , Factores de Tiempo
19.
Vaccine ; 26(4): 509-21, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18155327

RESUMEN

Recombinant Saccharomyces cerevisiae (yeast) represents a unique and attractive vehicle to deliver antigens in vaccine immunotherapy protocols for cancer or infectious disease, in that it has been shown to be extremely safe and can be administered multiple times to hosts. In the studies reported here, we describe the effects of treatment with recombinant yeast on murine immature dendritic cells (DCs). Yeast expressing human carcinoembryonic antigen (CEA) as a model antigen was studied. Injection of mice subcutaneously with yeast-CEA resulted in rapid increases in MHC class II(+) cells and total antigen-presenting cells in draining lymph nodes. Post-treatment with yeast-CEA, DCs rapidly elevated both MHC class I and class II, numerous costimulatory molecules and other DC maturation markers, and secreted a range of Type I inflammatory cytokines. Gene expression arrays also revealed the rapid up-regulation of numerous cytokine and chemokine mRNAs, as well as genes involved in signal transduction and antigen uptake. Functional studies demonstrated enhanced allospecific reactivity of DCs following treatment with yeast-CEA or control yeast. Additionally, treatment of DCs with yeast-CEA resulted in specific activation of CEA-specific CD8(+) T cells in an MHC-restricted manner in vitro. Lastly, vaccination of CEA-transgenic mice with yeast-CEA elicited antigen-specific CD4(+) and CD8(+) immune responses in vivo. Thus, these studies taken together form a scientific rationale for the use of recombinant yeast in vaccination protocols for cancer or infectious diseases.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Vectores Genéticos/inmunología , Saccharomyces cerevisiae/inmunología , Vacunación , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Citocinas/biosíntesis , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inyecciones Subcutáneas , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Saccharomyces cerevisiae/genética
20.
J Immunol ; 179(6): 3524-34, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785786

RESUMEN

The role of different levels of costimulation in conjunction with signal 1 in the activation of memory CD8+ T cells remains elusive. In this study, we demonstrate, in a mouse model with the influenza nucleoprotein epitope NP68, that mouse early memory (effector/memory) CD8+ T cells that were generated with high levels of costimulation have reduced CTL functionality compared with those that were generated with low levels of costimulation. This reduction is associated with increased phosphorylation of the negative regulatory site 292 on Zap70 and a decrease in granzyme B levels. Furthermore, we show that enhanced costimulation reduces proliferation and cytokine production of effector/memory CD8+ T cells in response to intermediate and weak TCR stimulation, in contrast to previously described positive effects of costimulation on naive CD8+ T cells. This effect is associated with the expression of ICAM-1 on APCs. Together, our results indicate that enhanced costimulation can lead to reduced functionality in effector/memory CD8+ T cells. This compromised effector function of effector/memory CD8+ T cells in response to high levels of costimulation can have important implications for designing immunotherapeutic strategies to enhance immune responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Regulación hacia Abajo/inmunología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Regulación hacia Arriba/inmunología , Sustitución de Aminoácidos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Relación Dosis-Respuesta Inmunológica , Granzimas/antagonistas & inhibidores , Granzimas/biosíntesis , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Nucleocápside , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Nucleoproteínas/metabolismo , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...