Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Immunol ; 206(1): 181-192, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33277387

RESUMO

MCP-1-induced monocyte chemotaxis is a crucial event in inflammation and atherogenesis. Identifying the important signal transduction pathways that control monocyte chemotaxis can unravel potential targets for preventive therapies in inflammatory disease conditions. Previous studies have shown that the focal adhesion kinase Pyk2 plays a critical role in monocyte motility. In this study, we investigated the MCP-1-mediated activation of Pyk2 (particularly by the phosphorylation of Tyr402) in primary human peripheral blood monocytes. We showed that MCP-1 induces Src phosphorylation in a similar time frame and that the MCP-1-induced Pyk2 tyrosine phosphorylation is controlled by the Src family kinase. We also report, in this study, that PKCß, an isoform of PKC, is required for both Src and Pyk2 activation/phosphorylation in response to MCP-1 stimulation. We identified Lyn as the specific Src kinase isoform that is activated by MCP-1 and acts upstream of Pyk2 in primary monocytes. Furthermore, Lyn is found to be indispensable for monocyte migration in response to MCP-1 stimulation. Moreover, our coimmunoprecipitation studies in monocytes revealed that PKCß, Pyk2, and Lyn exist constitutively in a molecular complex. To our knowledge, our study has uncovered a novel PKCß-Lyn-Pyk2 signaling cascade in primary monocytes that regulates MCP-1-induced monocyte adhesion and migration.


Assuntos
Quimiocina CCL2/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Monócitos/fisiologia , Complexos Multiproteicos/metabolismo , Proteína Quinase C beta/metabolismo , Quinases da Família src/metabolismo , Adesão Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiotaxia , Humanos , Fosforilação , Cultura Primária de Células , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transdução de Sinais
2.
J Biol Chem ; 293(36): 14040-14064, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30021838

RESUMO

Monoamine oxidase A (MAO-A) is a mitochondrial flavoenzyme implicated in the pathogenesis of atherosclerosis and inflammation and also in many neurological disorders. MAO-A also has been reported as a potential therapeutic target in prostate cancer. However, the regulatory mechanisms controlling cytokine-induced MAO-A expression in immune or cancer cells remain to be identified. Here, we show that MAO-A expression is co-induced with 15-lipoxygenase (15-LO) in interleukin 13 (IL-13)-activated primary human monocytes and A549 non-small cell lung carcinoma cells. We present evidence that MAO-A gene expression and activity are regulated by signal transducer and activator of transcription 1, 3, and 6 (STAT1, STAT3, and STAT6), early growth response 1 (EGR1), and cAMP-responsive element-binding protein (CREB), the same transcription factors that control IL-13-dependent 15-LO expression. We further established that in both primary monocytes and in A549 cells, IL-13-stimulated MAO-A expression, activity, and function are directly governed by 15-LO. In contrast, IL-13-driven expression and activity of MAO-A was 15-LO-independent in U937 promonocytic cells. Furthermore, we demonstrate that the 15-LO-dependent transcriptional regulation of MAO-A in response to IL-13 stimulation in monocytes and in A549 cells is mediated by peroxisome proliferator-activated receptor γ (PPARγ) and that signal transducer and activator of transcription 6 (STAT6) plays a crucial role in facilitating the transcriptional activity of PPARγ. We further report that the IL-13-STAT6-15-LO-PPARγ axis is critical for MAO-A expression, activity, and function, including migration and reactive oxygen species generation. Altogether, these results have major implications for the resolution of inflammation and indicate that MAO-A may promote metastatic potential in lung cancer cells.


Assuntos
Interleucina-13/fisiologia , Monoaminoxidase/metabolismo , Monócitos/metabolismo , Células A549 , Araquidonato 15-Lipoxigenase/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Inflamação , Neoplasias Pulmonares/patologia , Monoaminoxidase/fisiologia , Metástase Neoplásica , PPAR gama/metabolismo , Fator de Transcrição STAT6/metabolismo , Células U937
4.
J Immunol ; 198(12): 4855-4867, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28500072

RESUMO

Macrophage accumulation is a critical step during development of chronic inflammation, initiating progression of many devastating diseases. Leukocyte-specific integrin αDß2 (CD11d/CD18) is dramatically upregulated on macrophages at inflammatory sites. Previously we found that CD11d overexpression on cell surfaces inhibits in vitro cell migration due to excessive adhesion. In this study, we have investigated how inflammation-mediated CD11d upregulation contributes to macrophage retention at inflammatory sites during atherogenesis. Atherosclerosis was evaluated in CD11d-/-/ApoE-/- mice after 16 wk on a Western diet. CD11d deficiency led to a marked reduction in lipid deposition in aortas and isolated macrophages. Macrophage numbers in aortic sinuses of CD11d-/- mice were reduced without affecting their apoptosis and proliferation. Adoptive transfer of fluorescently labeled wild-type and CD11d-/- monocytes into ApoE-/- mice demonstrated similar recruitment from circulation, but reduced accumulation of CD11d-/- macrophages within the aortas. Furthermore, CD11d expression was significantly upregulated on macrophages in atherosclerotic lesions and M1 macrophages in vitro. Interestingly, expression of the related ligand-sharing integrin CD11b was not altered. This difference defines their distinct roles in the regulation of macrophage migration. CD11d-deficient M1 macrophages demonstrated improved migration in a three-dimensional fibrin matrix and during resolution of peritoneal inflammation, whereas migration of CD11b-/- M1 macrophages was not affected. These results prove the contribution of high densities of CD11d to macrophage arrest during atherogenesis. Because high expression of CD11d was detected in several inflammation-dependent diseases, we suggest that CD11d/CD18 upregulation on proinflammatory macrophages may represent a common mechanism for macrophage retention at inflammatory sites, thereby promoting chronic inflammation and disease development.


Assuntos
Aterosclerose/imunologia , Vasos Sanguíneos/patologia , Antígenos CD11/genética , Antígenos CD18/genética , Cadeias alfa de Integrinas/genética , Macrófagos/imunologia , Animais , Aorta/imunologia , Aorta/patologia , Apolipoproteínas E/deficiência , Aterosclerose/etiologia , Aterosclerose/patologia , Vasos Sanguíneos/imunologia , Antígenos CD11/imunologia , Antígenos CD18/imunologia , Dieta Ocidental , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Cadeias alfa de Integrinas/deficiência , Cadeias alfa de Integrinas/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Peritonite/imunologia , Peritonite/patologia , Ativação Transcricional , Regulação para Cima
5.
J Biol Chem ; 288(4): 2778-88, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23184931

RESUMO

IL-13 is a potent stimulator of alternative monocyte/macrophage activation. During alternative activation, the expression of several proteins is induced including 15-lipoxygenase (15-LO), a lipid-peroxidating enzyme and the scavenger receptor CD36. We previously reported that α(M)ß(2) integrin activation or clustering suppresses the expression of both 15-LO and CD36. In this study we focused on exploring the molecular mechanisms that down-regulate CD36 expression and CD36-mediated foam cell formation in IL-13-stimulated monocytes/macrophages after α(M)ß(2) activation. Our studies reveal that α(M)ß(2) integrin activation inhibits the IL-13 activation of several critical pathways that are required for macrophage alternative activation; namely, blocking Jak2 and Tyk2 phosphorylation, which bind to the cytoplasmic tails of the IL-4Rα/IL-13Rα1 complex. This leads to the inhibition of tyrosine phosphorylation of Stats (Stat1, Stat3, and Stat6) and prevents the formation of a signaling complex (containing p38MAPK, PKCδ, and Stat3) that are critical for the expression of both 15-LO and CD36. Jak2-mediated Hck activation is also inhibited, thereby preventing Stats serine phosphorylation, which is essential for downstream Stat-dependent gene transcription. Moreover, inhibition of Jak2, Tyk2, or their downstream target 15-LO with antisense oligonucleotides profoundly inhibits IL-13-induced CD36 expression and CD36-dependent foam cell formation, whereas13(S) Hydroperoxyoctadecadienoic acid (HPODE), a 15-LO product and peroxisome proliferator-activated receptor-γ ligand, completely restores CD36 expression in monocytes treated with 15-LO antisense. α(M)ß(2) integrin activation controls CD36 expression and foam cell formation in alternatively activated monocyte/macrophages by blocking Tyk2/Jak2 phosphorylation via a 15-LO-dependent pathway. The discovery of this mechanism helps our understanding of the potential role of alternatively activated macrophages in atherogenesis and highlights the impact of integrin α(M)ß(2) on this process.


Assuntos
Células Espumosas/citologia , Antígeno de Macrófago 1/metabolismo , Macrófagos/metabolismo , Receptores de Interleucina-13/metabolismo , Animais , Aterosclerose , Antígenos CD36/biossíntese , Separação Celular , Feminino , Citometria de Fluxo , Humanos , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Janus Quinase 2/metabolismo , Lipídeos/química , Macrófagos/citologia , Camundongos , Transdução de Sinais , TYK2 Quinase/metabolismo
6.
J Lipid Res ; 54(2): 436-47, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23160182

RESUMO

Monocyte chemoattractant protein-1 (MCP-1)-induced monocyte chemotaxis is a major event in inflammatory disease. Our prior studies have demonstrated that MCP-1-dependent chemotaxis requires release of arachidonic acid (AA) by activated cytosolic phospholipase A(2) (cPLA(2)). Here we investigated the involvement of AA metabolites in chemotaxis. Neither cyclooxygenase nor lipoxygenase pathways were required, whereas pharmacologic inhibitors of both the cytochrome-P450 (CYP) and the soluble epoxide hydrolase (sEH) pathways blocked monocyte chemotaxis to MCP-1. To verify specificity, we demonstrated that the CYP and sEH products epoxyeiscosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids (DHETs), respectively, restored chemotaxis in the presence of the inhibitors, indicating that sEH-derived products are essential for MCP-1-driven chemotaxis. Importantly, DHETs also rescued chemotaxis in cPLA(2)-deficient monocytes and monocytes with blocked Erk1/2 activity, because Erk controls cPLA(2) activation. The in vitro findings regarding the involvement of CYP/sEH pathways were further validated in vivo using two complementary approaches measuring MCP-1-dependent chemotaxis in mice. These observations reveal the importance of sEH in MCP-1-regulated monocyte chemotaxis and may explain the observed therapeutic value of sEH inhibitors in treatment of inflammatory diseases, cardiovascular diseases, pain, and even carcinogenesis. Their effectiveness, often attributed to increasing EET levels, is probably influenced by the impairment of DHET formation and inhibition of chemotaxis.


Assuntos
Quimiocina CCL2/metabolismo , Quimiotaxia , Epóxido Hidrolases/química , Epóxido Hidrolases/metabolismo , Monócitos/citologia , Animais , Ácido Araquidônico/biossíntese , Quimiotaxia/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Ácidos Graxos Monoinsaturados/química , Ácidos Graxos Monoinsaturados/metabolismo , Feminino , Humanos , Lipoxigenase/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/metabolismo , Fosfolipases A2 Citosólicas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Solubilidade
7.
Circ Res ; 108(5): 544-54, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21252155

RESUMO

RATIONALE: The alternative activation of monocytes by interleukin (IL)-13 and IL-4 is a significant component of the inflammatory response. The consequences of alternative activation in inflammatory diseases remain to be determined. OBJECTIVE: In this report, we explored how integrins, receptors important for monocyte migration to inflammatory sites, regulate IL-13-mediated monocyte activation. We focused on the analysis of 2 proteins, which are upregulated during the alternative activation and are important for the development of atherosclerosis, an oxidative enzyme 15-lipoxygenase (15-LO) and a scavenger receptor CD36. METHODS AND RESULTS: We found that adhesion of resting monocytes through ß(2) integrins and inside-out activation of ß(2) integrins by monocyte chemoattractant protein-1 did not change IL-13-stimulated 15-LO upregulation; however, preincubation of monocytes with the antibody MEM48, which generates full activation of ß(2) integrins, significantly inhibited 15-LO mRNA and protein expression. In contrast, activation of ß(1) integrins had no effect on 15-LO expression. Analysis of integrin clustering through α(M), α(L), α(X), and α(D) subunits demonstrated the pivotal role for integrin α(M)ß(2) in inhibiting 15-LO expression. IL-13 treatment upregulates 15-LO-dependent CD36 expression on human monocytes; our studies showed that ß(2) integrin activation and α(M) integrin clustering significantly inhibited IL-13-dependent CD36 mRNA and protein expression, as well as CD36-related foam cell formation. Moreover, IL-13 stimulation of α(M)-deficient peritoneal macrophages demonstrated an upregulated level of 15-LO induction, CD36 expression, and lipid accumulation as compared with wild-type controls. CONCLUSIONS: The adhesion of monocytes/macrophages through activated integrin α(M)ß(2) has a regulatory and potential atheroprotective function during the alternative activation of macrophages.


Assuntos
Células Espumosas/citologia , Células Espumosas/metabolismo , Ativação de Macrófagos/fisiologia , Antígeno de Macrófago 1/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Animais , Araquidonato 15-Lipoxigenase/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígenos CD36/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Interleucina-13/farmacologia , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , Fragmentos de Peptídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de IgG/metabolismo
8.
Inflamm Res ; 62(11): 991-1001, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23974215

RESUMO

OBJECTIVE AND DESIGN: We designed a study to detect downstream phosphorylation targets of PKCß in MCP-1-induced human monocytes. METHODS: Two-dimensional gel electrophoresis was performed for monocytes treated with MCP-1 in the presence or absence of PKCß antisense oligodeoxyribonucleotides (AS-ODN) or a PKCß inhibitor peptide, followed by phospho- and total protein staining. Proteins that stained less intensely with the phospho-stain, when normalized to the total protein stain, in the presence of PKCß AS-ODN or the PKCß inhibitor peptide, were sequenced. RESULTS: Of the proteins identified, vimentin was consistently identified using both experimental approaches. Upon (32)P-labeling and vimentin immunoprecipitation, increased phosphorylation of vimentin was observed in MCP-1 treated monocytes as compared to the untreated monocytes. Both PKCß AS-ODN and the PKCß inhibitor reduced MCP-1-induced vimentin phosphorylation. The IP of monocytes with anti-vimentin antibody and immunoblotting with a PKCß antibody revealed that increased PKCß becomes associated with vimentin upon MCP-1 activation. Upon MCP-1 treatment, monocytes were shown to secrete vimentin and secretion depended on PKCß expression and activity. CONCLUSIONS: We conclude that vimentin, a major intermediate filament protein, is a phosphorylation target of PKCß in MCP-1-treated monocytes and that PKCß phosphorylation is essential for vimentin secretion. Our recently published studies have implicated vimentin as a potent stimulator of the innate immune receptor Dectin-1 as reported by Thiagarajan et al. (Cardiovasc Res 99:494-504, 2013). Taken together our findings suggest that inhibition of PKCß regulates vimentin secretion and, thereby, its interaction with Dectin-1 and downstream stimulation of superoxide anion production. Thus, PKCß phosphorylation of vimentin likely plays an important role in propagating inflammatory responses.


Assuntos
Quimiocina CCL2/imunologia , Monócitos/imunologia , Proteína Quinase C beta/imunologia , Vimentina/imunologia , Células Cultivadas , Humanos , Fosforilação
9.
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
10.
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
11.
Circulation ; 115(23): 2939-47, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17533179

RESUMO

BACKGROUND: Signal transducer and activator of transcription 1 (Stat1) potently regulates gene expression after stimulation by certain cytokines involved in tumorigenesis and host defenses. The present study investigated a novel role for Stat1 in foam cell formation and atherosclerosis. METHODS AND RESULTS: Inhibition of Stat1 activity by a Stat1-specific DNA "decoy" oligomer transfected into differentiated human THP-1 cells, and deficiency of stat1 in mouse macrophages significantly inhibited foam cell formation assessed by lipid staining and cholesteryl ester accumulation compared with control cells. The mechanism of Stat1 regulation of foam cell formation was uniquely dependent on the scavenger receptor CD36. Blunted Stat1 activity and stat1 deficiency significantly decreased expression of CD36 but not of scavenger receptor-A compared with controls, as assessed by immunoblotting and flow cytometry. Deficiency of CD36 but not scavenger receptor-A in mouse macrophages removed any dependency of foam cell formation on Stat1. In an intraperitoneal model of foam cell formation in which foam cells form in vivo independently of the model ligands used in vitro, stat1 deficiency significantly inhibited foam cell formation and CD36 expression. Transplantation of bone marrow from apolipoprotein e-/- x stat1-/- mice into lethally irradiated, atherosclerosis-susceptible apolipoprotein e-/- recipients significantly reduced both en face aortic lesion coverage and aortic root lesions compared with recipients of bone marrow from genetically matched apolipoprotein e-/- mice. CONCLUSIONS: Stat1 regulates CD36 expression and foam cell formation in macrophages in vitro; the Stat1 regulation of foam cell formation requires CD36. The regulation of CD36 expression by Stat1 may be important in other pathophysiological CD36-dependent events. Stat1 deficiency reduces atherosclerosis in an apolipoprotein e-/- atherosclerosis-susceptible bone marrow transplantation model.


Assuntos
Aterosclerose/etiologia , Antígenos CD36/genética , Células Espumosas/citologia , Macrófagos Peritoneais/citologia , Fator de Transcrição STAT1/fisiologia , Animais , Aterosclerose/patologia , Transplante de Medula Óssea , Antígenos CD36/análise , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Macrófagos Peritoneais/metabolismo , Camundongos , Fator de Transcrição STAT1/deficiência
12.
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
13.
Cell Signal ; 19(2): 359-66, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16963226

RESUMO

Phospholipases A(2) (PLA(2)) are potent regulators of the inflammatory response. We have observed that Group IV cPLA(2) activity is required for the production of superoxide anion (O(2)(-)) in human monocytes [Li Q., Cathcart M.K. J. Biol. Chem. 272 (4) (1997) 2404-2411.]. We have previously identified PKCalpha as a kinase pathway required for monocyte O(2)(-) production [Li Q., Cathcart M.K. J. Biol. Chem. 269 (26) (1994) 17508-17515.]. We therefore investigated the potential interaction between PKCalpha and cPLA(2) by evaluating the requirement for specific PKC isoenzymes in the process of activating cPLA(2) enzymatic activity and protein phosphorylation upon monocyte activation. We first showed that general PKC inhibitors and antisense oligodeoxyribonucleotides (ODN) to the cPKC group of PKC enzymes inhibited cPLA(2) activity. To distinguish between PKCalpha and PKCbeta isoenzymes in regulating cPLA(2) protein phosphorylation and enzymatic activity, we employed our previously characterized PKCalpha or PKCbeta isoenzyme-specific antisense ODN [Li Q., Subbulakshmi V., Fields A.P., Murray, N.R., Cathcart M.K., J. Biol. Chem. 274 (6) (1999) 3764-3771]. Suppression of PKCalpha expression, but not PKCbeta expression, inhibited cPLA(2) protein phosphorylation and enzymatic activity. Additional studies ruled out a contribution by Erk1/2 to cPLA(2) phosphorylation and activation. We also found that cPLA(2) co-immunoprecipitated with PKCalpha and vice versa. In vitro studies demonstrated that PKCalpha could directly phosphorylate cPLA(2).and enhance enzymatic activity. Finally, we showed that addition of arachidonic acid restored the production of O(2)(-) in monocytes defective in either PKCalpha or cPLA(2) expression. Taken together, our data suggest that PKCalpha, but not PKCbeta, is the predominant cPKC isoenzyme required for cPLA(2) protein phosphorylation and maximal induction of cPLA(2) enzymatic activity upon activation of human monocytes. Our data also support the concept that the requirements for PKCalpha and cPLA(2) in O(2)(-) generation are solely due to their seminal role in generating arachidonic acid.


Assuntos
Regulação Enzimológica da Expressão Gênica , Monócitos/enzimologia , Fosfolipases A/metabolismo , Proteína Quinase C-alfa/metabolismo , Ácido Araquidônico/farmacologia , Células Cultivadas , Ativação Enzimática , Humanos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fosfolipases A/genética , Fosforilação , Proteína Quinase C-alfa/fisiologia
14.
Mol Cell Biol ; 23(11): 3918-28, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12748293

RESUMO

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.


Assuntos
Araquidonato 15-Lipoxigenase/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Interleucina-13/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Serina/metabolismo , Transativadores/metabolismo , Animais , Araquidonato 15-Lipoxigenase/genética , Células Cultivadas , DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Monócitos/citologia , Monócitos/metabolismo , Fosforilação , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno
15.
J Leukoc Biol ; 77(3): 414-20, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15591124

RESUMO

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.


Assuntos
Monócitos/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Separação Celular , Humanos , Oligonucleotídeos Antissenso/farmacologia , Fosfoproteínas/efeitos dos fármacos , Fosforilação , Proteína Quinase C-delta , Superóxidos/metabolismo , Zimosan/antagonistas & inibidores , Zimosan/farmacologia
16.
Cancer Res ; 63(7): 1676-83, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12670922

RESUMO

It is now understood that the genetic plasticity of cancer cells can lead to alterations that confer selective growth advantages to the tumor, some of which play a role in immune escape. A number of mutations veiling tumor cells from host immune defenses have been well characterized but more recent studies suggest that a variety of tumors can also express products that are actually toxic for the immune effectors. A component of this tumor-induced T-cell death has been attributed to receptor-mediated apoptosis. Some tumors, however, synthesize soluble factors that mediate similar effects. In this regard, we previously showed that supernatants from explanted renal cell carcinoma (RCC) tumors sensitized normal T cells to activation induced cell death, and the responsible products had the features of gangliosides. We have also shown that renal tumor lines, including SK-RC-45, induce apoptosis of both Jurkat cells and normal T lymphocytes. Here, we used the ganglioside synthesis inhibitor PPPP to define the role of gangliosides in RCC cell line (SK-RC-45)-mediated T cell and Jurkat cell apoptosis and to elucidate the proapoptotic molecular events by which the glycosphingolipids produce their effects. The ganglioside-synthesizing SK-RC-45 line stimulated the TUNEL (terminal deoxynucleotidyl transferase-mediated nick end labeling) positivity of cocultured T cells by a mechanism that involved decreasing lymphocyte expression levels of Bcl-2 and Bcl-(XL), inducing cytochrome c release from their mitochondria and activating caspases 9 and 3. These proapoptotic events were partially or completely abrogated when tumor cells were pretreated with PPPP for 5 days before the SK-RC-45/T lymphocyte coincubation, a regimen that reduced tumor-associated ganglioside levels by 70-80%. Our results suggest that gangliosides may be key mediators of RCC-induced T-cell apoptosis and imply that they contribute to the T-cell dysfunction in the tumor microenvironment.


Assuntos
Apoptose/fisiologia , Carcinoma de Células Renais/patologia , Comunicação Celular/fisiologia , Gangliosídeos/fisiologia , Neoplasias Renais/patologia , Linfócitos T/patologia , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/metabolismo , Caspases/metabolismo , Técnicas de Cocultura , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Gangliosídeos/antagonistas & inibidores , Gangliosídeos/biossíntese , Humanos , Células Jurkat , Neoplasias Renais/imunologia , Neoplasias Renais/metabolismo , Mitocôndrias/metabolismo , Propanolaminas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Pirrolidinas/farmacologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Tumorais Cultivadas , Proteína bcl-X
17.
Arterioscler Thromb Vasc Biol ; 24(1): 23-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14525794

RESUMO

Monocyte extravasation into the vessel wall has been shown to be a critical step in the development of atherosclerosis. Upon activation, monocytes produce a burst of superoxide anion due to activation of the NADPH oxidase enzyme complex. Monocyte-derived superoxide anion contributes to oxidant stress in inflammatory sites, is required for monocyte-mediated LDL oxidation, and alters basic cell functions such as adhesion and proliferation. We hypothesize that monocyte-derived superoxide anion production contributes to atherosclerotic lesion formation. In this brief review, we summarize our current understanding of the signal transduction pathways regulating NADPH oxidase activation and related superoxide anion production in activated human monocytes. Novel pathways are identified that may serve as future targets for therapeutic intervention in this pathogenic process. The contributions of superoxide anion and NADPH oxidase to atherogenesis are discussed. Future experiments are needed to clarify the exact role of NADPH oxidase-derived superoxide anion in atherogenesis, particularly that derived from monocytes.


Assuntos
Arteriosclerose/etiologia , Macrófagos/fisiologia , Monócitos/fisiologia , NADPH Oxidases/metabolismo , Superóxidos/metabolismo , Angiotensina II/fisiologia , Animais , Arteriosclerose/enzimologia , Arteriosclerose/patologia , Arteriosclerose/prevenção & controle , Sinalização do Cálcio , Humanos , Macrófagos/enzimologia , Camundongos , Monócitos/enzimologia , Estresse Oxidativo , Fosfolipases A/fisiologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/fisiologia , Processamento de Proteína Pós-Traducional , Vasculite/complicações , Vasculite/enzimologia , Vasculite/patologia
18.
J Leukoc Biol ; 72(3): 580-9, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12223527

RESUMO

Interleukin (IL)-13 regulates monocyte function and is a potent stimulator of 15-lipoxygenase expression. In different cell types, the functional IL-13 receptor complex can be comprised of variable protein components and has not been thoroughly examined in human monocytes. Here, we identify the receptor components and upstream signaling events initiated by IL-13 in primary human blood monocytes. The expression, phosphorylation and associated Jak kinases of the known, variable receptor components, IL-4R(alpha), IL-2Rgammac, IL-13R(alpha)1 and IL-13R(alpha)2, were examined. We determined that IL-4R(alpha) and IL13R(alpha)1 are phosphorylated upon exposure to IL-13. Although IL-2Rgammac is also expressed, it is not phosphorylated upon exposure to IL-13. Evaluation of the presence of IL-13R(alpha)2 failed to reveal significant mRNA or protein expression. Earlier, our laboratory showed that IL-13 induced the phosphorylation of Jak2 and Tyk2 in monocytes and that expression of both Jaks was essential for downstream signaling by IL-13. Here, we report that Jak2 is associated with IL-4R(alpha), and Tyk2 is associated with the IL-13R(alpha)1 component of the IL-13 receptor complex. Additionally, Stat proteins 1alpha, 3, 5A, 5B, and 6 are phosphorylated in response to IL-13. Further, the nuclear translocation and DNA binding of each of these Stats were induced by IL-13. These data represent the first complete report of the functional IL-13 receptor complex and early signaling events in human monocytes. This information is critical for understanding the IL-13 response of monocytes in inflammation.


Assuntos
Interleucina-13/fisiologia , Proteínas do Leite , Monócitos/metabolismo , Proteínas Proto-Oncogênicas , Receptores de Interleucina/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transporte Ativo do Núcleo Celular , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Inflamação/metabolismo , Fator Gênico 3 Estimulado por Interferon , Interleucina-13/farmacologia , Subunidade alfa1 de Receptor de Interleucina-13 , Janus Quinase 2 , Substâncias Macromoleculares , Monócitos/efeitos dos fármacos , Fosforilação , Processamento de Proteína Pós-Traducional , Subunidades Proteicas , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , RNA Mensageiro/biossíntese , Receptores de Interleucina/química , Receptores de Interleucina/genética , Receptores de Interleucina/fisiologia , Receptores de Interleucina-13 , Receptores de Interleucina-2/química , Receptores de Interleucina-2/metabolismo , Receptores de Interleucina-4/química , Receptores de Interleucina-4/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Fator de Transcrição STAT6 , Transdução de Sinais/efeitos dos fármacos , TYK2 Quinase , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
19.
J Invest Dermatol ; 135(2): 389-399, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25184961

RESUMO

Injury initiates recruitment of macrophages to support tissue repair; however, excessive macrophage activity may exacerbate tissue damage causing further destruction and subsequent delay in wound repair. Here we show that the peroxisome proliferation-activated receptor-γ agonist, rosiglitazone (Rosi), a medication recently reintroduced as a drug to treat diabetes and with known anti-inflammatory properties, paradoxically generates pro-inflammatory macrophages. This is observed in both IL-6-deficient mice and control wild-type mice experimentally induced to produce high titers of auto-antibodies against IL-6, mimicking IL-6 deficiency in human diseases. IL-6 deficiency when combined with Rosi-mediated upregulation of suppressor of cytokine signaling 3 leads to an altered ratio of nuclear signal transducer and activator of transcription 3/NF-κB that allows hyper-induction of inducible nitric oxide synthase (iNOS). Macrophages activated in this manner cause de novo tissue destruction, recapitulating human chronic wounds, and can be reversed in vivo by recombinant IL-6, blocking macrophage infiltration, or neutralizing iNOS. This study provides insight into an unanticipated paradoxical role of Rosi in mediating hyper-inflammatory macrophage activation significant for diseases associated with IL-6 deficiency.


Assuntos
Inflamação/etiologia , Interleucina-6/deficiência , Ativação de Macrófagos/efeitos dos fármacos , Pele/patologia , Tiazolidinedionas/farmacologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Óxido Nítrico Sintase Tipo II/fisiologia , Rosiglitazona , Fator de Transcrição STAT3/fisiologia , Cicatrização
20.
Artigo em Inglês | MEDLINE | ID: mdl-26052543

RESUMO

Monocytes/macrophages are versatile cells centrally involved in host defense and immunity. Th1 cytokines induce a classical activation program in monocytes/macrophages leading to a proinflammatory M1 macrophage phenotype while Th2 cytokines IL-4 and IL-13 promote monocyte differentiation into an alternatively activated, anti-inflammatory M2 macrophage phenotype. Although monoamine oxidase A (MAO-A) is primarily known for its action in the nervous system, several recent studies have identified MAO-A as a signature marker of alternative activation of monocytes/macrophages. In this brief review we explore the signaling pathways/molecules that regulate MAO-A expression in alternatively activated monocytes/macrophages. We further discuss the contribution of MAO-A to the resolution of inflammation and identify potential therapeutic targets for controlling inflammation. Altogether this review provides deeper insight into the role of MAO-A in alternative activation of monocytes/macrophages and their participation in the inflammatory response.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA