Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomedicines ; 10(5)2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35625917

RESUMO

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.

2.
Int J Mol Sci ; 22(21)2021 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-34769278

RESUMO

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.


Assuntos
Interleucina-10/imunologia , Ativação Linfocitária/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nivolumabe/farmacologia , Linfócitos T/imunologia , Humanos , Interferon gama/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Antígeno Ki-67/imunologia , Sistema de Sinalização das MAP Quinases/imunologia
3.
AAPS J ; 23(5): 99, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34401948

RESUMO

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.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/imunologia , Inibidores de Checkpoint Imunológico/efeitos adversos , Fígado/patologia , Neoplasias/tratamento farmacológico , Animais , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/epidemiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Modelos Animais de Doenças , Humanos , Incidência , Fígado/efeitos dos fármacos , Fígado/imunologia , Regeneração Hepática/efeitos dos fármacos , Regeneração Hepática/imunologia , Neoplasias/imunologia , Índice de Gravidade de Doença
4.
Int J Mol Sci ; 21(23)2020 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-33261003

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Imunoensaio , Imunoterapia , Leucócitos Mononucleares/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Antígenos CD/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Granzimas/metabolismo , Humanos , Ativação Linfocitária/efeitos dos fármacos , Nivolumabe/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Doadores de Tecidos
6.
AAPS J ; 22(4): 80, 2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32462555

RESUMO

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.


Assuntos
Autoanticorpos/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Urato Oxidase/administração & dosagem , Administração Intravenosa , Animais , Autoanticorpos/imunologia , Autoanticorpos/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Supressores da Gota/administração & dosagem , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Regulação para Cima/fisiologia
7.
MAbs ; 12(1): 1685814, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31774346

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Bevacizumab/química , Isotipos de Imunoglobulinas/química , Solução Salina/química , Trastuzumab/química , Proteínas Sanguíneas/química , Glucose , Humanos , Concentração de Íons de Hidrogênio , Imunoconjugados/química , Infusões Intravenosas , Ponto Isoelétrico , Espectrometria de Massas , Agregados Proteicos
8.
Oncoimmunology ; 8(1): e1512456, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30546949

RESUMO

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.

9.
Oncotarget ; 7(11): 12447-63, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26799670

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Neoplasias Cerebelares/tratamento farmacológico , Cisplatino/farmacologia , Meduloblastoma/tratamento farmacológico , Piperazinas/farmacologia , Proteína Fosfatase 2/antagonistas & inibidores , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Linhagem Celular Tumoral , Neoplasias Cerebelares/enzimologia , Neoplasias Cerebelares/patologia , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Meduloblastoma/enzimologia , Meduloblastoma/patologia , Camundongos , Camundongos SCID , Piperazinas/administração & dosagem , Proteína Fosfatase 2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncoimmunology ; 4(12): e1052929, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26587324

RESUMO

We have recently discovered that BRAF inhibitors induce potent macrophage responses that confer melanoma resistance to therapy. Our studies lay a foundation for the hypothesis that macrophages switch their role from a passenger to a driver for tumor survival during therapeutic treatment, suggesting that agents that target macrophages can be an important component of "cocktail" anticancer therapy.

11.
Immunol Rev ; 268(1): 160-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26497519

RESUMO

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.


Assuntos
Imunoglobulina G/metabolismo , Receptores de IgG/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Regulação da Expressão Gênica , Humanos , Imunoglobulina G/química , Imunoglobulina G/imunologia , Imunoterapia , Infecções/imunologia , Infecções/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interferon gama/metabolismo , Ligantes , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Especificidade de Órgãos/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/uso terapêutico , Transdução de Sinais , Fator de Necrose Tumoral alfa/antagonistas & inibidores
12.
Virology ; 476: 372-376, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25589240

RESUMO

Transfusion of blood and blood products contaminated with the pathogenic form of prion protein Prp(sc), thought to be the causative agent of variant a Creutzfeldt-Jakob disease (vCJD), may result in serious consequences in recipients with a compromised immune system, for example, as seen in HIV-1 infection. In the present study, we demonstrate that treatment of peripheral blood monocyte-derived macrophages (MDM) with PrP106-126, a synthetic domain of PrP(sc) that has intrinsic functional activities related to the full-length protein, markedly increased their susceptibility to HIV-1 infection, induced cytokine secretion, and enhanced their migratory behavior in response to N-formyl-l-methionyl-l-leucyl-l-phenylalanine (fMLP). Live-cell imaging of MDM cultured in the presence of PrP106-126 showed large cell clusters indicative of cellular activation. Tyrosine kinase inhibitor STI-571, protein kinase C inhibitor K252B, and cyclin-dependent kinase inhibitor olomoucine attenuated PrP106-126-induced altered MDM functions. These findings delineate a previously undefined functional role of PrP106-126-mediated host cell response in promoting HIV-1 pathogenesis.


Assuntos
Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Macrófagos/virologia , Monócitos/virologia , Proteínas PrPSc/farmacologia , Células Cultivadas , Infecções por HIV/metabolismo , Humanos , Proteínas PrPSc/metabolismo
13.
J Immunol ; 194(3): 983-9, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25512601

RESUMO

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.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Quinases da Família src/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Monócitos/efeitos dos fármacos , Fosforilação , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica
14.
MAbs ; 6(6): 1377-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25484046

RESUMO

Antibodies evoke cellular responses through the binding of their Fc region to Fc receptors, most of which contain immunoreceptor tyrosine-based activation motif domains and are thus considered "activating." However, there is a growing appreciation of these receptors for their ability to deliver an inhibitory signal as well. We previously described one such phenomenon whereby interferon (IFN)γ signaling is inhibited by immune complex signaling through FcγRI. To understand the implications of this in the context of therapeutic antibodies, we assessed individual IgG subclasses to determine their ability to deliver this anti-inflammatory signal in monocyte-derived macrophages. Like IgG1, we found that IgG4 is fully capable of inhibiting IFNγ-mediated events. In addition, F(ab')2 fragments that interfere with FcγRI signaling reversed this effect. For mAbs developed with either an IgG1 or an IgG4 constant region for indications where inflammation is undesirable, further examination of a potential Fc-dependent contribution to their mechanism of action is warranted.


Assuntos
Imunoglobulina G/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Receptores de IgG/imunologia , Células Cultivadas , Citometria de Fluxo , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunofenotipagem , Interferon gama/imunologia , Interferon gama/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais/imunologia
15.
J Biol Chem ; 286(42): 36709-23, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21878628

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Ativação de Macrófagos/fisiologia , Monócitos/metabolismo , Proteínas Proto-Oncogênicas c-hck/metabolismo , Araquidonato 15-Lipoxigenase/biossíntese , Antígenos CD36/metabolismo , Ativação Enzimática/fisiologia , Humanos , Interleucina-13/biossíntese , Janus Quinase 2/metabolismo , Lectinas Tipo C/metabolismo , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase 6/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Monoaminoxidase/metabolismo , Monócitos/citologia , Fosforilação/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-13/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/metabolismo
16.
J Immunol ; 185(9): 5211-24, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20861348

RESUMO

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.


Assuntos
Araquidonato 15-Lipoxigenase/biossíntese , Regulação da Expressão Gênica/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monócitos/enzimologia , Araquidonato 15-Lipoxigenase/imunologia , Proteína de Ligação a CREB , DNA de Cadeia Simples , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Humanos , Immunoblotting , Imunoprecipitação , Interleucina-13/imunologia , Interleucina-13/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Monócitos/imunologia , Fosforilação , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Transfecção
17.
Blood ; 115(3): 549-58, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19965653

RESUMO

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.


Assuntos
Anexina A2/fisiologia , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Receptor 4 Toll-Like/fisiologia , Animais , Anexina A2/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células Cultivadas , Feminino , Humanos , Inflamação/genética , Inflamação/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Multimerização Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
18.
J Immunol Methods ; 330(1-2): 86-95, 2008 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-18191414

RESUMO

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.


Assuntos
Bioensaio/métodos , Quimiocina CCL2/metabolismo , Quimiotaxia de Leucócito , Monócitos/imunologia , Peritonite/imunologia , Proteína Quinase C/metabolismo , Transdução de Sinais/imunologia , Transfecção/métodos , Transferência Adotiva , Animais , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Quimiotaxia de Leucócito/genética , Modelos Animais de Doenças , Estudos de Viabilidade , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Monócitos/enzimologia , Monócitos/transplante , Mutação , Peritonite/induzido quimicamente , Peritonite/enzimologia , Proteína Quinase C/genética , Proteína Quinase C beta , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais/genética , Tioglicolatos , Fatores de Tempo
19.
J Leukoc Biol ; 82(5): 1174-84, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17715360

RESUMO

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.


Assuntos
Anexina A2/farmacologia , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/metabolismo , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/citologia , Monócitos/imunologia , NF-kappa B/genética , NF-kappa B/metabolismo , Fagocitose , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de IgG/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
20.
Am J Obstet Gynecol ; 196(4): 394.e1-11; discussion 394.e11-3, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17403432

RESUMO

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.


Assuntos
Neoplasias do Endométrio/genética , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , NF-kappa B/metabolismo , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Neoplasias do Endométrio/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Genes Reporter , Humanos , NF-kappa B/genética , Reação em Cadeia da Polimerase , Gravidez , Probabilidade , Ligação Proteica , Estudos de Amostragem , Sensibilidade e Especificidade , Transdução de Sinais , Ativação Transcricional , Trofoblastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...