Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34769278

RESUMEN

Immune checkpoint inhibitor (ICI) therapy has revolutionized anti-cancer treatment for many late-stage cancer patients. However, ICI therapy has thus far demonstrated limited efficacy for most patients, and it remains unclear why this is so. Interleukin 10 (IL-10) is a cytokine that has been recognized as a central player in cancer biology with its ability to inhibit anti-tumor T cell responses. Recent studies suggest that IL-10 might also exert some intrinsic anti-tumor T cell responses, and clinical studies using recombinant IL-10 alone or in combination with ICI are underway. This paradoxical effect of IL-10 and its underlying mechanisms impacting ICI-modulated T cell responses remain poorly understood. In this study, using an in vitro mixed lymphocyte reaction assay, we found that treatment with ICIs such as the anti-programmed cell death receptor-1 (PD-1) mAb nivolumab elicits a strong expression of IL-10. While neutralization of IL-10 signaling with an anti-IL-10 specific mAb significantly decreases the production of IFN-γ by T cells in a cohort of donor cells, the opposite effect was observed in other donor cells. Similarly, neutralization of IL-10 signaling significantly decreases the expression of T cell activation markers Ki67 and CD25, as well as the production of Granzyme B in a cohort of donor cells, whereas the opposite effect was observed in others. Furthermore, we found that nivolumab and IL-10 differentially modulate the signal transducer and activator of transcription 3 (STAT3) and AKT serine-threonine kinase pathways. Finally, we found that nivolumab activates the mitogen-activated protein kinase (MAPK) pathway, which in turn is responsible for the observed induction of IL-10 production by nivolumab. These findings provide new insights into the mechanisms underlying anti-PD-1-modulated T cell responses by IL-10, which could lead to the discovery of novel combination treatments that target IL-10 and immune checkpoint molecules.


Asunto(s)
Interleucina-10/inmunología , Activación de Linfocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nivolumab/farmacología , Linfocitos T/inmunología , Humanos , Interferón gamma/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Antígeno Ki-67/inmunología , Sistema de Señalización de MAP Quinasas/inmunología
2.
Int J Mol Sci ; 21(23)2020 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-33261003

RESUMEN

The absence of reliable, robust, and non-invasive biomarkers for anti- Programmed cell death protein 1 (PD-1) immunotherapy is an urgent unmet medical need for the treatment of cancer patients. No predictive biomarkers have been established based on the direct assessment of T cell functions, the primary mechanism of action of anti-PD-1 therapy. In this study, we established a model system to test T cell functions modulated by Nivolumab using anti-CD3 monoclonal antibody (mAb)-stimulated peripheral blood mononuclear cells (PBMCs), and characterized T cell functions primarily based on the knowledge gained from retrospective observations of patients treated with anti-PD-1 immunotherapy. During a comprehensive cytokine profile assessment to identify potential biomarkers, we found that Nivolumab increases expression of T helper type 1 (Th1) associated cytokines such as interferon-γ (IFN-γ) and interleukin-2 (IL-2) in a subset of donors. Furthermore, Nivolumab increases production of Th2, Th9, and Th17 associated cytokines, as well as many proinflammatory cytokines such as IL-6 in a subset of donors. Conversely, Nivolumab treatment has no impact on T cell proliferation, expression of CD25, CD69, or Granzyme B, and only modestly increases in the expansion of regulatory T cells. Our results suggest that assessment of cytokine production using a simple PBMC-based T cell functional assay could be used as a potential predictive marker for anti-PD-1 immunotherapy.


Asunto(s)
Biomarcadores/metabolismo , Inmunoensayo , Inmunoterapia , Leucocitos Mononucleares/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Antígenos CD/metabolismo , Proliferación Celular/efectos de los fármacos , Citocinas/biosíntesis , Granzimas/metabolismo , Humanos , Activación de Linfocitos/efectos de los fármacos , Nivolumab/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Donantes de Tejidos
3.
Immunol Rev ; 268(1): 160-74, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497519

RESUMEN

Fcγ receptor I (FcγRI or CD64) is the sole human Fc receptor with high affinity for monovalent IgG. While it contains an immunoreceptor tyrosine-based activation motif in its cytoplasmic domain, binding of FcγRI can result in a complex array of activating and inhibitory outcomes. For instance, binding of monomeric IgG provides a low-intensity tonic signal through FcγRI that is necessary for full interferon γ receptor signaling in the same cell. Interaction of FcγRI with larger high-avidity complexes can result in phagocytosis, the generation of reactive oxygen species, as well as the synthesis and release of inflammatory cytokines. However, numerous reports also document potent anti-inflammatory effects brought about by FcγRI engagement with immune complexes such as the inhibition of IFNγ and TLR4 signaling, and secretion of interleukin-10. This has led to conflicting hypotheses regarding the function of FcγRI, especially with regard to its role in the efficacy of several therapeutic monoclonal antibodies. While many of these issues are still unclear, continued characterization of the regulation and context dependence of FcγRI function, as well as the molecular mechanisms responsible for these various outcomes, will improve our understanding of FcγRI biology as well as the therapeutic strategies designed to harness or constrain its actions.


Asunto(s)
Inmunoglobulina G/metabolismo , Receptores de IgG/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Regulación de la Expresión Génica , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Inmunoterapia , Infecciones/inmunología , Infecciones/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/metabolismo , Ligandos , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Especificidad de Órganos/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/uso terapéutico , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
4.
J Immunol ; 194(3): 983-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25512601

RESUMEN

The regulation of the innate and the adaptive immune responses are extensively intertwined and tightly regulated. Ag-driven immune responses that are modulated by immune complexes (ICs) are known to inhibit IFN-γ-dependent MHC class II expression. We have previously demonstrated that ICs dramatically inhibit IFN-γ-induced activation of human monocytes through the activation of the FcγRI signaling pathway. In the present study we further explore the mechanisms by which ICs regulate IFN-γ activation of human monocytes. We demonstrate that members of the SRC kinase family (SKF) are key mediators of IFN-γ pathway suppression: inhibitors of the SKF reverse the ability of ICs to suppress IFN-γ signaling. Small interfering RNA was used to target specific members of the SKF. The data indicate that SRC and LYN are both required for ICs to elicit their immunosuppressive activity, whereas FYN does not appear to contribute to this function. Similarly, the kinase SYK, though not a member of the SKF, is also demonstrated to be involved in this IC-mediated immunosuppression. Our data suggest a mechanism whereby ICs directly inhibit inflammatory signals by crosslinking FcγRI, resulting in the activation of the specific phosphotyrosine kinases SRC, LYN, and SYK and the concomitant suppression of the IFN-γ signaling pathway.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Familia-src Quinasas/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Activación Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Monocitos/efectos de los fármacos , Fosforilación , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética
5.
J Biol Chem ; 286(42): 36709-23, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21878628

RESUMEN

IL-13 is a Th2 cytokine that promotes alternative activation (M2 polarization) in primary human monocytes. Our studies have characterized the functional IL-13 receptor complex and the downstream signaling events in response to IL-13 stimulation in alternatively activated monocytes/macrophages. In this report, we present evidence that IL-13 induces the activation of a Src family tyrosine kinase, which is required for IL-13 induction of M2 gene expression, including 15-lipoxygenase (15-LO). Our data show that Src kinase activity regulates IL-13-induced p38 MAPK tyrosine phosphorylation via the upstream kinases MKK3 or MKK6. Our findings also reveal that the IL-13 receptor-associated tyrosine kinase Jak2 is required for the activation of both Src kinase as well as p38 MAPK. Further, we found that Src tyrosine kinase-mediated activation of p38 MAPK is required for Stat1 and Stat3 serine 727 phosphorylation in alternatively activated monocytes/macrophages. Additional studies identify Hck as the specific Src family member, stimulated by IL-13 and involved in regulating both p38 MAPK activation and p38 MAPK-mediated 15-LO expression. Finally we show that the Hck regulates the expression of other alternative state (M2)-specific genes (Mannose receptor, MAO-A, and CD36) and therefore conclude that Hck acts as a key regulator controlling gene expression in alternatively activated monocytes/macrophages.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Activación de Macrófagos/fisiología , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-hck/metabolismo , Araquidonato 15-Lipooxigenasa/biosíntesis , Antígenos CD36/metabolismo , Activación Enzimática/fisiología , Humanos , Interleucina-13/biosíntesis , Janus Quinasa 2/metabolismo , Lectinas Tipo C/metabolismo , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa 6/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Monoaminooxidasa/metabolismo , Monocitos/citología , Fosforilación/fisiología , Receptores de Superficie Celular/metabolismo , Receptores de Interleucina-13/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/metabolismo
6.
Blood ; 115(3): 549-58, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965653

RESUMEN

Annexins are a large family of intracellular phospholipid-binding proteins, yet several extracellular roles have been identified. Specifically, annexin A2, found in a heterotetrameric complex with S100A10, not only serves as a key extracellular binding partner for pathogens and host proteins alike, but also can be shed or secreted. We reported previously that soluble annexin A2 tetramer (A2t) activates human monocyte-derived macrophages (MDM), resulting in secretion of inflammatory mediators and enhanced phagocytosis. Although a receptor for A2t has been cloned from bone marrow stromal cells, data contained in this study demonstrate that it is dispensable for A2t-dependent activation of MDM. Furthermore, A2t activates wild-type murine bone marrow-derived macrophages, whereas macrophages from myeloid differentiation factor 88-deficient mice display a blunted response, suggesting a role for Toll-like receptor (TLR) signaling. Small interfering RNA knockdown of TLR4 in human MDM reduced the response to A2t, blocking antibodies against TLR4 (but not TLR2) blocked activation altogether, and bone marrow-derived macrophages from TLR4(-/-) mice were refractory to A2t. These data demonstrate that the modulation of macrophage function by A2t is mediated through TLR4, suggesting a previously unknown, but important role for this stress-sensitive protein in the detection of danger to the host, whether from injury or invasion.


Asunto(s)
Anexina A2/fisiología , Activación de Macrófagos/genética , Macrófagos/metabolismo , Receptor Toll-Like 4/fisiología , Animales , Anexina A2/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Multimerización de Proteína/fisiología , ARN Interferente Pequeño/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
J Immunol ; 185(9): 5211-24, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20861348

RESUMEN

IL-13 induces profound expression of 15-lipoxygenase (15-LO) in primary human monocytes. Our studies have defined the functional IL-13R complex, association of Jaks with the receptor components, and the tyrosine phosphorylation of several Stat molecules in response to IL-13. Furthermore, we identified both p38MAPK and protein kinase Cδ as critical regulators of 15-LO expression. In this study, we report an ERK1/2-dependent signaling cascade that regulates IL-13-mediated 15-LO gene expression. We show the rapid phosphorylation/activation of ERK1/2 upon IL-13 exposure. Our results indicate that Tyk2 kinase is required for the activation of ERK1/2, which is independent of the Jak2, p38MAPK, and protein kinase Cδ pathways, suggesting bifurcating parallel regulatory pathways downstream of the receptor. To investigate the signaling mechanisms associated with the ERK1/2-dependent expression of 15-LO, we explored the involvement of transcription factors, with predicted binding sites in the 15-LO promoter, in this process including Elk1, early growth response-1 (Egr-1), and CREB. Our findings indicate that IL-13 induces Egr-1 nuclear accumulation and CREB serine phosphorylation and that both are markedly attenuated by inhibition of ERK1/2 activity. We further show that ERK1/2 activity is required for both Egr-1 and CREB DNA binding to their cognate sequences identified within the 15-LO promoter. Furthermore, by transfecting monocytes with the decoy oligodeoxyribonucleotides specific for Egr-1 and CREB, we discovered that Egr-1 and CREB are directly involved in regulating 15-LO gene expression. These studies characterize an important regulatory role for ERK1/2 in mediating IL-13-induced monocyte 15-LO expression via the transcription factors Egr-1 and CREB.


Asunto(s)
Araquidonato 15-Lipooxigenasa/biosíntesis , Regulación de la Expresión Génica/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/enzimología , Araquidonato 15-Lipooxigenasa/inmunología , Proteína de Unión a CREB , ADN de Cadena Simple , Activación Enzimática/inmunología , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Humanos , Immunoblotting , Inmunoprecipitación , Interleucina-13/inmunología , Interleucina-13/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Monocitos/inmunología , Fosforilación , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Transfección
8.
Biomedicines ; 10(5)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35625917

RESUMEN

While promising, PD-L1 expression on tumor tissues as assessed by immunohistochemistry has been shown to be an imperfect biomarker that only applies to a limited number of cancers, whereas many patients with PD-L1-negative tumors still respond to anti-PD-(L)1 immunotherapy. Recent studies using patient blood samples to assess immunotherapeutic responsiveness suggests a promising approach to the identification of novel and/or improved biomarkers for anti-PD-(L)1 immunotherapy. In this review, we discuss the advances in our evolving understanding of the regulation and function of PD-L1 expression, which is the foundation for developing blood-based PD-L1 as a biomarker for anti-PD-(L)1 immunotherapy. We further discuss current knowledge and clinical study results for biomarker identification using PD-L1 expression on tumor and immune cells, exosomes, and soluble forms of PD-L1 in the peripheral blood. Finally, we discuss key challenges for the successful development of the potential use of blood-based PD-L1 as a biomarker for anti-PD-(L)1 immunotherapy.

9.
AAPS J ; 23(5): 99, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34401948

RESUMEN

Immune checkpoint inhibitors (ICIs) represent a promising therapy for many types of cancer. However, only a portion of patients respond to this therapy and some patients develop clinically significant immune-mediated liver injury caused by immune checkpoint inhibitors (ILICI), an immune-related adverse event (irAE) that may require the interruption or termination of treatment and administration of systemic corticosteroids or other immunosuppressive agents. Although the incidence of ILICI is lower with monotherapy, the surge in combining ICIs with chemotherapy, targeted therapy, and combination of different ICIs has led to an increase in the incidence and severity of ILICI - a major challenge for development of effective and safe ICI therapy. In this review, we highlight the importance and contribution of the liver microenvironment to ILICI by focusing on the emerging roles of resident liver cells in modulating immune homeostasis and hepatocyte regeneration, two important decisive factors that dictate the initiation, progression, and recovery from ILICI. Based on the proposed contribution of the liver microenvironment on ICILI, we discuss the clinical characteristics of ILICI in patients with preexisting liver diseases, as well as the challenges of identifying prognostic biomarkers to guide the clinical management of severe ILICI. A better understanding of the liver microenvironment may lead to novel strategies and identification of novel biomarkers for effective management of ILICI.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Hígado/patología , Neoplasias/tratamiento farmacológico , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/epidemiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Animales de Enfermedad , Humanos , Incidencia , Hígado/efectos de los fármacos , Hígado/inmunología , Regeneración Hepática/efectos de los fármacos , Regeneración Hepática/inmunología , Neoplasias/inmunología , Índice de Severidad de la Enfermedad
10.
AAPS J ; 22(4): 80, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32462555

RESUMEN

Therapeutic proteins can be potent agents for treating serious diseases, but in many patients these proteins provoke antibody responses that blunt therapeutic efficacy. Intravenous administration of high doses of some proteins induces immune tolerance, but the mechanisms underlying this effect are poorly understood. As a model to study tolerance induction in mice, we used rasburicase, a commercial recombinant uricase used for the treatment of hyperuricemia. Intraperitoneal (i.p.) injection of rasburicase without or with alum adjuvants induced a clear anti-rasburicase antibody response, but intravenous (i.v.) injection did not. The lack of response to i.v. rasburicase was apparently due to active immune suppression since i.v.-treated mice showed blunted antibody and reduced T cell responses to subsequent i.p. injections of rasburicase. This blunted response was associated with a decrease in rasburicase-specific B cell and T cell responses and an increase in proportion of CD4+ FoxP3+ regulatory T cells (Treg) in the spleen. We examined the number of lymphocytes in peripheral blood after rasburicase i.v. injection. Rasburicase caused a transient reduction in B and T cells, but a robust and sustained depletion of rasburicase-specific B cells. Further experiments showed that rasburicase i.v. injection decreased the number of lymphocytes and was associated with apoptosis of both B cells and activated T cells and that the enhanced percentage of Treg cells was likely mediated by a macrophage-dependent pathway. Thus, our data suggest that apoptosis and depletion of antigen-specific B lymphocytes and upregulation of Treg cells may play important roles in the immune suppression induced by intravenous administration of a therapeutic protein.


Asunto(s)
Autoanticuerpos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Urato Oxidasa/administración & dosificación , Administración Intravenosa , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Supresores de la Gota/administración & dosificación , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Regulación hacia Arriba/fisiología
11.
MAbs ; 12(1): 1685814, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31774346

RESUMEN

Therapeutic monoclonal antibodies (mAbs) are commonly administered to patients through intravenous (IV) infusion, which involves diluting the medication into an infusion solution (e.g., saline and 5% dextrose). Using the wrong diluent can cause product aggregation, which may compromise patient safety. We and others have shown that Herceptin® (trastuzumab) and Avastin® (bevacizumab) undergo rapid aggregation upon mixing with dextrose and human plasma in vitro. In this study, we evaluated the compatibility of a panel of 11 therapeutic mAbs with dextrose or saline and human serum. These mAbs were randomly selected for their distinct formulations and IgG isotypes (IgG1, IgG2, IgG4, and Fc-fusion protein). All the mAbs appeared to be compatible with saline and human serum. However, mAbs that were formulated at acidic pH (≤ 6.5) exclusively formed insoluble aggregates upon mixing with dextrose and serum. Such aggregation was not detected for the mAbs that are at neutral pH (7.2-7.5) or in buffers containing sodium chloride. Mass spectrometric analysis revealed that the insoluble aggregates were composed of mAb molecules and several serum proteins (e.g., complement proteins, apolipoprotein, fibronectin) that are characterized by an isoelectric point of pH 5.4-6.7. At proximate pH to the isoelectric point values, those abundant serum proteins appeared to undergo isoelectric precipitation with mAb molecules. Our observations highlight a potential risk of protein aggregation at the blood-IV interface if a diluent is incompatible with a specific mAb formulation. This information has implications in guiding the design of product formulations and the selection of the right diluent for intravenous infusion of therapeutic mAbs.Abbreviations: ADC: antibody-drug conjugate; D5W: 5% dextrose in water; IM: intramuscular; IV: intravenous; LC-MS/MS: liquid chromatography-tandem mass spectrometry; mAb: monoclonal antibody; SC: subcutaneous; pI: isoelectric point.


Asunto(s)
Anticuerpos Monoclonales/química , Bevacizumab/química , Isotipos de Inmunoglobulinas/química , Solución Salina/química , Trastuzumab/química , Proteínas Sanguíneas/química , Glucosa , Humanos , Concentración de Iones de Hidrógeno , Inmunoconjugados/química , Infusiones Intravenosas , Punto Isoeléctrico , Espectrometría de Masas , Agregado de Proteínas
12.
Oncoimmunology ; 8(1): e1512456, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30546949

RESUMEN

Immune checkpoint inhibitors (ICIs) such as the anti-PD-1 antibody Nivolumab, achieve remarkable clinical efficacy in patients with late stage cancers. However, only a small subset of patients benefit from this therapy. Numerous clinical trials are underway testing whether combining ICIs with other anti-cancer therapies can increase this response rate. For example, anti-PD-1/PD-L1 therapy combined with MAP kinase inhibition using BRAF inhibitors (BRAFi) and/or MEK inhibitors (MEKi) are in development for treatment of late stage melanomas. However, the benefits and underlying mechanisms of these combinatorial therapies remain unclear. In the current study, we assess the effects of MAPK inhibition on Nivolumab-induced T cell responses. Using an in vitro mixed lymphocyte reaction assay, we demonstrate that Nivolumab-induced T cell activation is highly heterogeneous. While BRAFi inhibits Nivolumab-induced cytokine production, T cell proliferation, activation markers (CD69, CD25), and Granzyme B in a substantial proportion of donor pairs, a small subset of donor pairs shows an additive effect. MEKi alone significantly inhibits Nivolumab-induced T cell activation; the addition of BRAFi significantly enhances this inhibitory effect. Mechanistically, the effects of BRAFi and/or MEKi on Nivolumab-induced T cell activation may be due to alteration of the activation of the AKT and T cell receptor (TCR) signaling pathways. Our results suggest that MAPK inhibition may not provide a clinical benefit for most melanoma patients being treated with anti-PD-1 therapy.

13.
J Immunol Methods ; 330(1-2): 86-95, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-18191414

RESUMEN

Identification of novel signal transduction pathways regulating monocyte chemotaxis can indicate unique targets for preventive therapies for treatment of chronic inflammatory diseases. To aid in this endeavor we report conditions for optimal transfection of primary human monocytes coupled with a new model system for assessing their chemotactic activity in vivo. This method can be used as a tool to identify the relevant signal transduction pathways regulating human monocyte chemotaxis to MCP-1 in the complex in vivo environment that were previously identified to regulate chemotaxis in vitro. MCP-1-dependent chemotaxis of monocytes is studied in an adoptive transfer model where human monocytes transfected with mutant cDNAs are transferred to mice followed by initiation of peritonitis. Harvesting peritoneal cells at 24 h diminishes the contribution of immunologic responses to the cross-species transfer. Validation of relevant regulatory molecules in vivo is critical for understanding the most relevant therapeutic targets for drug development.


Asunto(s)
Bioensayo/métodos , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito , Monocitos/inmunología , Peritonitis/inmunología , Proteína Quinasa C/metabolismo , Transducción de Señal/inmunología , Transfección/métodos , Traslado Adoptivo , Animales , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Quimiotaxis de Leucocito/genética , Modelos Animales de Enfermedad , Estudios de Factibilidad , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Monocitos/enzimología , Monocitos/trasplante , Mutación , Peritonitis/inducido químicamente , Peritonitis/enzimología , Proteína Quinasa C/genética , Proteína Quinasa C beta , Proteínas Recombinantes de Fusión/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/genética , Tioglicolatos , Factores de Tiempo
14.
J Leukoc Biol ; 82(5): 1174-84, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17715360

RESUMEN

On the surface of the macrophage, annexin A2 tetramer (A2t) serves as a docking protein or recognition element for bacterial and viral pathogens. Plasma levels of free A2t have been reported to increase following infection, although the mechanistic significance of this observation is unclear. Although annexin A2 had generally been thought to play an anti-inflammatory role, soluble A2t stimulates MAP kinase activity in bone marrow stromal cells downstream of a recently cloned receptor. This raises the question of whether A2t activates human macrophages via MAP kinases and whether it might be capable of acting as an inflammatory mediator. To this end, human monocyte-derived macrophages were treated with soluble A2t and MAP kinase phosphorylation, p65 NF-kappaB activation, and inflammatory mRNA and protein levels were measured. It was found that A2t caused rapid phosphorylation of several MAP kinases, as well as translocation of p65 NF-kappaB to the nucleus. A2t stimulated the production of TNF-alpha, IL-1beta, and IL-6, as well as several members of the chemokine family within 24 h, which are capable of recruitment and/or activation of a broad range of leukocyte classes. Furthermore, A2t-activated macrophages demonstrated enhanced phagocytic ability for the ingestion of GFP-expressing Escherichia coli. These data are the first to suggest the participation of an annexin in microbial clearance, as well as the establishment of inflammation and the immune response, including the recruitment and activation of immune cells to the site of infection.


Asunto(s)
Anexina A2/farmacología , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/metabolismo , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Monocitos/citología , Monocitos/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , Fagocitosis , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de IgG/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Leukoc Biol ; 81(4): 1086-92, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17227821

RESUMEN

Antigen-driven immune responses are modulated by immune complexes (ICs), in part through their ability to inhibit IFN-gamma-dependent MHC Class II expression. We have demonstrated previously that ICs dramatically inhibit IFN-gamma-induced activation of human monocytes through the suppression of the JAK/STAT signaling pathway. In the current study, we further explore the mechanisms by which ICs regulate IFN-gamma activation of human monocytes. Consistent with previous studies in monocytes pretreated with ICs, there was a reduction in steady-state levels of RNA by real-time RT-PCR of the IFN-inducible protein 10 gene as well as the FcgammaRI gene. Pull-down assays confirm that IC pretreatment inhibits IFN-gamma-induced STAT1 phosphorylation without affecting the ability of STAT1 to bind to the STAT1-binding domain of the IFN-gamma receptor. In addition, the inhibitory function of ICs was reduced when cells from the FcR common gamma-chain knockout mice were used, supporting the role of the FcgammaRI in this inhibitory pathway. It is unexpected that ICs also require the phosphatase Src homology-2-containing tyrosine phosphatase 1 (SHP-1) to inhibit IFN-gamma induction, as demonstrated by studies with cells from the SHP-1 knockout (motheaten) mice. These data suggest a mechanism of IC-mediated inhibition of IFN-gamma signaling, which requires the ITAM-containing FcgammaRI, as well as the ITIM-dependent phosphatase SHP-1, ultimately resulting in the suppression of STAT1 phosphorylation.


Asunto(s)
Complejo Antígeno-Anticuerpo/farmacología , Interferón gamma/metabolismo , Monocitos/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Cadenas gamma de Inmunoglobulina , Fosforilación , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor de Transcripción STAT1/metabolismo
16.
Am J Obstet Gynecol ; 196(4): 394.e1-11; discussion 394.e11-3, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17403432

RESUMEN

OBJECTIVE: The objective of the study was to determine whether cross-talk occurs between estrogen receptors (ERs) and nuclear factor-kappa-B (NF-kappaB), to assess the functional consequences of such an ER/NF-kappaB interaction, and to identify other unknown regulatory proteins that may participate in the NF-kappaB transcriptional complex. STUDY DESIGN: Electromobility gel shifts, reporter gene assays, and mass spectrometry were used to identify proteins interacting with the NF-kappaB deoxyribonucleic acid (DNA) response element. RESULTS: ER and the p65 subunit of NF-kappaB colocalized on DNA. This interaction was inhibitory for ER transcriptional activity. Sequencing of proteins bound to the NF-kappaB/DNA complex identified DNA-modifying enzymes, scaffolding proteins, chaperones, and elements of the nuclear matrix. CONCLUSION: These studies have identified an inhibitory interaction between estrogen receptors and the p65 subunit of NF-kappaB with implications for estrogen action in pregnancy and cancer. New accessory proteins have also been identified that bind to protein complexes on the NF-kappaB DNA response element.


Asunto(s)
Neoplasias Endometriales/genética , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , FN-kappa B/metabolismo , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Neoplasias Endometriales/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Genes Reporteros , Humanos , FN-kappa B/genética , Reacción en Cadena de la Polimerasa , Embarazo , Probabilidad , Unión Proteica , Muestreo , Sensibilidad y Especificidad , Transducción de Señal , Activación Transcripcional , Trofoblastos/citología
17.
Mol Cell Biol ; 23(11): 3918-28, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12748293

RESUMEN

Interleukin-13 (IL-13) is a cytokine secreted by Th2 lymphocytes that is capable of inducing expression of 15-lipoxygenase (15-LO) in primary human monocytes. We recently demonstrated that induction of 15-LO requires the activation of Jak2 and Tyk2 kinases and Stats 1, 3, 5, and 6. Since IL-13-induced 15-LO expression was inhibited by H7 (a serine-threonine kinase inhibitor), we predicted that Stat serine phosphorylation may also be crucial for 15-LO expression. In this study, we present evidence indicating that IL-13-induced 15-LO mRNA expression was detectable as early as 1 h by real-time reverse transcription-PCR. We found that IL-13 induced a time-dependent serine phosphorylation of both Stat1 and Stat3, detectable at 15 min after IL-13 treatment. In addition, the activation of p38 mitogen-activated protein kinase (MAPK) was detected in a time-dependent fashion, with peak phosphorylation at 15 min after IL-13 treatment. SB202190, a p38 MAPK-specific inhibitor, markedly inhibited IL-13-induced Stat1 and Stat3 serine phosphorylation as well as DNA binding. Furthermore, treatment of cells with Stat1 or Stat3 decoys significantly impaired IL-13-induced 15-LO expression. Taken together, our results provide the first evidence that IL-13 induces p38 MAPK phosphorylation/activation, which regulates Stat1 and Stat3 serine 727 phosphorylation. Both of these events are important steps in IL-13-induced 15-LO expression in human monocytes.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Interleucina-13/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Serina/metabolismo , Transactivadores/metabolismo , Animales , Araquidonato 15-Lipooxigenasa/genética , Células Cultivadas , ADN/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Monocitos/citología , Monocitos/metabolismo , Fosforilación , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos
18.
J Leukoc Biol ; 77(3): 414-20, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15591124

RESUMEN

Phosphorylation of the reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase components p67phox and p47phox accompanies the assembly and activation of this enzyme complex. We have previously reported that activation of human monocytes with opsonized zymosan (ZOP), a potent stimulator of NADPH oxidase activity, results in the phosphorylation of p67phox and p47phox. In this study, we investigated the regulation of p67phox phosphorylation. Although protein kinase C (PKC)alpha has previously been shown to regulate NADPH oxidase activity, we found that inhibition of PKCalpha had no effect on p67phox phosphorylation. Our studies demonstrate that pretreatment of monocytes with antisense oligodeoxyribonucleotides specific for PKCdelta or rottlerin, a selective inhibitor for PKCdelta, inhibited the phosphorylation of p67phox in monocytes, and Go6976, a specific inhibitor for conventional PKCs, PKCalpha and PKCbeta, had no such inhibitory effect. Additional studies indicate that ZOP stimulation of monocytes induces PKCdelta and p67phox to form a complex. We also demonstrate that lysates from activated monocytes as well as PKCdelta immunoprecipitates from activated monocytes can phosphorylate p67phox in vitro and that pretreatment of monocytes with rottlerin blocked the phosphorylation in each case. We further show that recombinant PKCdelta can phosphorylate p67phox in vitro. Finally, we show that PKCdelta-deficient monocytes produce significantly less superoxide anion in response to ZOP stimulation, thus emphasizing the functional significance of the PKCdelta regulation of p67phox phosphorylation. Taken together, this is the first report to describe the requirement of PKCdelta in regulating the phosphorylation of p67phox and the related NADPH oxidase activity in primary human monocytes.


Asunto(s)
Monocitos/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Separación Celular , Humanos , Oligonucleótidos Antisentido/farmacología , Fosfoproteínas/efectos de los fármacos , Fosforilación , Proteína Quinasa C-delta , Superóxidos/metabolismo , Zimosan/antagonistas & inhibidores , Zimosan/farmacología
19.
Oncotarget ; 7(11): 12447-63, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26799670

RESUMEN

The protein phosphatase 2A (PP2A) inhibitor, LB100, has been shown in pre-clinical studies to be an effective chemo- and radio-sensitizer for treatment of various cancers. We investigated effects associated with LB100 treatment alone and in combination with cisplatin for medulloblastoma (MB) in vitro and in vivo in an intracranial xenograft model. We demonstrated that LB100 had a potent effect on MB cells. By itself, LB100 inhibited proliferation and induced significant apoptosis in a range of pediatric MB cell lines. It also attenuated MB cell migration, a pre-requirement for invasion. When used in combination, LB100 enhanced cisplatin-mediated cytotoxic effects. Cell viability in the presence of 1 uM cisplatin alone was 61% (DAOY), 100% (D341), and 58% (D283), but decreased with the addition of 2 µM of LB100 to 26% (DAOY), 67% (D341), and 27% (D283), (p < 0.005). LB100 suppressed phosphorylation of the STAT3 protein and several STAT3 downstream targets. Also, LB100 directly increased cisplatin uptake and overcame cisplatin-resistance in vitro. Finally, LB100 exhibited potent in vivo anti-neoplastic activity in combination with cisplatin in an intracranial xenograft model.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Cisplatino/farmacología , Meduloblastoma/tratamiento farmacológico , Piperazinas/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Línea Celular Tumoral , Neoplasias Cerebelosas/enzimología , Neoplasias Cerebelosas/patología , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Meduloblastoma/enzimología , Meduloblastoma/patología , Ratones , Ratones SCID , Piperazinas/administración & dosificación , Proteína Fosfatasa 2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Leukoc Biol ; 74(1): 118-25, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12832450

RESUMEN

Specific prion peptides have been shown to mimic the pathologic isoform of the prion protein (PrP) and to induce a neurotoxic effect in vitro and in vivo. As monocytic cells are thought to play a role in the transmission and pathogenesis of prion disease, the use of these peptides in regulating monocytic cell function is under intense investigation. In the current study, we characterize the ability of prion peptide PrP(106-126) to activate specific signaling pathways in human monocyte-derived dendritic cells (DCs). Electrophoretic mobility shift assays establish the activation of transcription factor nuclear factor-kappaB within 15 min of exposure, with as little as 25 micro M peptide. This signaling cascade results in the up-regulation of inflammatory cytokines interleukin (IL)-1beta, IL-6, and tumor necrosis factor alpha (TNF-alpha) at the mRNA and protein levels. Phenotypic activation of DCs exposed to PrP(106-126) is partly a result of an autocrine TNF-alpha response and results in an increased ability of these cells to induce lymphocyte proliferation. The effects of PrP(106-126) on DCs were elicited through a receptor complex distinct from that used by human monocytes, demonstrating the ability of this peptide to interact with a multiplicity of receptors on various cell types. Together, these data suggest an involvement of DCs in prion disease pathogenesis.


Asunto(s)
Citocinas/biosíntesis , Células Dendríticas/efectos de los fármacos , FN-kappa B/metabolismo , Fragmentos de Péptidos/farmacología , Priones/farmacología , Células Dendríticas/citología , Células Dendríticas/inmunología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Inflamación/inmunología , Monocitos/citología , N-Formilmetionina Leucil-Fenilalanina/farmacología , FN-kappa B/fisiología , Enfermedades por Prión/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA