Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Immunol ; 208(4): 941-954, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35082159

RESUMO

TPL-2 kinase plays an important role in innate immunity, activating ERK1/2 MAPKs in myeloid cells following TLR stimulation. We investigated how TPL-2 controls transcription in TLR4-stimulated mouse macrophages. TPL-2 activation of ERK1/2 regulated expression of genes encoding transcription factors, cytokines, chemokines, and signaling regulators. Bioinformatics analysis of gene clusters most rapidly induced by TPL-2 suggested that their transcription was mediated by the ternary complex factor (TCF) and FOS transcription factor families. Consistently, TPL-2 induced ERK1/2 phosphorylation of the ELK1 TCF and the expression of TCF target genes. Furthermore, transcriptomic analysis of TCF-deficient macrophages demonstrated that TCFs mediate approximately half of the transcriptional output of TPL-2 signaling, partially via induced expression of secondary transcription factors. TPL-2 signaling and TCFs were required for maximal TLR4-induced FOS expression. Comparative analysis of the transcriptome of TLR4-stimulated Fos -/- macrophages indicated that TPL-2 regulated a significant fraction of genes by controlling FOS expression levels. A key function of this ERK1/2-TCF-FOS pathway was to mediate TPL-2 suppression of type I IFN signaling, which is essential for host resistance against intracellular bacterial infection.


Assuntos
Interferon beta/genética , MAP Quinase Quinase Quinases/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Animais , Regulação da Expressão Gênica , Interferon beta/metabolismo , Lipopolissacarídeos/imunologia , MAP Quinase Quinase Quinases/genética , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição TCF/metabolismo
2.
Biochem J ; 475(1): 329-340, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29229763

RESUMO

The MKK1/2 kinase tumour progression locus 2 (TPL-2) is critical for the production of tumour necrosis factor alpha (TNFα) in innate immune responses and a potential anti-inflammatory drug target. Several earlier pharmaceutical company screens with the isolated TPL-2 kinase domain have identified small-molecule inhibitors that specifically block TPL-2 signalling in cells, but none of these have progressed to clinical development. We have previously shown that TPL-2 catalytic activity regulates TNF production by macrophages while associated with NF-κB1 p105 and ABIN-2, independently of MKK1/2 phosphorylation via an unknown downstream substrate. In the present study, we used a positional scanning peptide library to determine the optimal substrate specificity of a complex of TPL-2, NF-κB1 p105 and ABIN-2. Using an optimal peptide substrate based on this screen and a high-throughput mass spectrometry assay to monitor kinase activity, we found that the TPL-2 complex has significantly altered sensitivities versus existing ATP-competitive TPL-2 inhibitors than the isolated TPL-2 kinase domain. These results imply that screens with the more physiologically relevant TPL-2/NF-κB1 p105/ABIN-2 complex have the potential to deliver novel TPL-2 chemical series; both ATP-competitive and allosteric inhibitors could emerge with significantly improved prospects for development as anti-inflammatory drugs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Anti-Inflamatórios/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Anti-Inflamatórios/síntese química , Expressão Gênica , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Biblioteca de Peptídeos , Peptídeos/síntese química , Ligação Proteica , Inibidores de Proteínas Quinases/síntese química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
3.
Biochem J ; 473(18): 2845-61, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27402796

RESUMO

Previous studies suggested that Toll-like receptor (TLR) stimulation of the p38α MAP kinase (MAPK) is mediated by transforming growth factor-ß-activated kinase 1 (TAK1) activation of MAPK kinases, MKK3, MKK4 and MKK6. We used quantitative mass spectrometry to monitor tumour progression locus 2 (TPL-2)-dependent protein phosphorylation following TLR4 stimulation with lipopolysaccharide, comparing macrophages from wild-type mice and Map3k8(D270A/D270A) mice expressing catalytically inactive TPL-2 (MAP3K8). In addition to the established TPL-2 substrates MKK1/2, TPL-2 kinase activity was required to phosphorylate the activation loops of MKK3/6, but not of MKK4. MKK3/6 activation required IκB kinase (IKK) phosphorylation of the TPL-2 binding partner nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB1) p105, similar to MKK1/2 activation. Tumour necrosis factor (TNF) stimulation of MKK3/6 phosphorylation was similarly dependent on TPL-2 catalytic activity and IKK phosphorylation of NF-κB1 p105. Owing to redundancy of MKK3/6 with MKK4, Map3k8(D270A) mutation only fractionally decreased lipopolysaccharide activation of p38α. TNF activation of p38α, which is mediated predominantly via MKK3/6, was substantially reduced. TPL-2 catalytic activity was also required for MKK3/6 and p38α activation following macrophage stimulation with Mycobacterium tuberculosis and Listeria monocytogenes Our experiments demonstrate that the IKK/NF-κB1 p105/TPL-2 signalling pathway, downstream of TAK1, regulates MKK3/6 and p38α activation in macrophages in inflammation.


Assuntos
Macrófagos/enzimologia , Proteínas Quinases/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Toll-Like/metabolismo , Animais , Ativação Enzimática , Espectrometria de Massas , Camundongos
4.
Crit Rev Immunol ; 34(4): 315-45, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24941159

RESUMO

IL-10 is an important anti-inflammatory cytokine that plays important roles in controlling inflammatory responses and keeping the immune system in check following activation. Loss of IL-10 function in mice or humans results in the development of inflammatory bowel disease in response to an elevated immune response to the gut flora. IL-10 also acts to prevent excessive inflammation during the course of infection and has been implicated in a variety of autoimmune conditions. In response to inflammatory signals, IL-10 can be produced by a number of immune cells including T cells, B cells, macrophages, and dendritic cells. Distinct mechanisms control the production of IL-10 in these different cells types. In this review, we describe recent studies that have looked at the signaling pathways that regulate IL-10 production in these cells. Given the number of cell types that produce IL-10, it is perhaps not surprising that the in vivo source of IL-10 can vary in different immune models. We also describe how work using conditional IL-10 knockout mice or adoptive transfer of IL-10-deficient cells has begun to further our understanding regarding which specific immune cells are required for IL-10 production in vivo under different conditions.


Assuntos
Regulação da Expressão Gênica , Interleucina-10/genética , Transcrição Gênica , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Interleucina-10/metabolismo , Espaço Intracelular/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Especificidade de Órgãos/genética , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
5.
J Immunol ; 190(2): 565-77, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241891

RESUMO

The polarization of macrophages into a regulatory-like phenotype and the production of IL-10 plays an important role in the resolution of inflammation. We show in this study that PGE(2), in combination with LPS, is able to promote an anti-inflammatory phenotype in macrophages characterized by high expression of IL-10 and the regulatory markers SPHK1 and LIGHT via a protein kinase A-dependent pathway. Both TLR agonists and PGE(2) promote the phosphorylation of the transcription factor CREB on Ser(133). However, although CREB regulates IL-10 transcription, the mutation of Ser(133) to Ala in the endogenous CREB gene did not prevent the ability of PGE(2) to promote IL-10 transcription. Instead, we demonstrate that protein kinase A regulates the phosphorylation of salt-inducible kinase 2 on Ser(343), inhibiting its ability to phosphorylate CREB-regulated transcription coactivator 3 in cells. This in turn allows CREB-regulated transcription coactivator 3 to translocate to the nucleus where it serves as a coactivator with the transcription factor CREB to induce IL-10 transcription. In line with this, we find that either genetic or pharmacological inhibition of salt-inducible kinases mimics the effect of PGE(2) on IL-10 production.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Interleucina-10/biossíntese , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Interleucina-10/genética , Camundongos , Fenótipo , Fosforilação/efeitos dos fármacos , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
6.
J Immunol ; 189(6): 2784-92, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22904308

RESUMO

Macrophages are an important source of cytokines following infection. Stimulation of macrophages with TLR agonists results in the secretion of TNF-α, IL-6, and IL-12, and the production of these cytokines is controlled by multiple feedback pathways. Macrophages also produce IL-10, which acts to inhibit proinflammatory cytokine production by macrophages via a JAK/STAT3-dependent pathway. We show in this paper that, Ruxolitinib, a recently described selective inhibitor of JAKs, increases TNF, IL-6, and IL-12 secretion in mouse bone marrow-derived macrophages stimulated with LPS. This effect is largely due to its ability to block IL-10-mediated feedback inhibition on cytokine transcription in macrophages. Similar results were also obtained with a second structurally unrelated Jak inhibitor, Tofacitinib. In addition, LPS induced the production of IFN-ß, which was then able to activate JAKs in macrophages, resulting in the stimulation of STAT1 phosphorylation. The initial induction of IL-10 was independent of JAK signaling; however, inhibition of JAKs did reduce IL-10 secretion at later time points. This reflected a requirement for the IFN-ß feedback loop to sustain IL-10 transcription following LPS stimulation. In addition to IL-10, IFN-ß also helped sustain IL-6 and IL-12 transcription. Overall, these results suggest that inhibition of JAKs may increase the inflammatory potential of macrophages stimulated with TLR4 agonists.


Assuntos
Células da Medula Óssea/imunologia , Citocinas/biossíntese , Retroalimentação , Interleucina-10/antagonistas & inibidores , Janus Quinases/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Regulação para Cima/imunologia , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/patologia , Células Cultivadas , Mediadores da Inflamação/fisiologia , Interferon Tipo I/fisiologia , Interleucina-10/fisiologia , Janus Quinases/fisiologia , Macrófagos/enzimologia , Macrófagos/patologia , Camundongos , Nitrilas , Pirazóis/farmacologia , Pirimidinas , Transdução de Sinais/imunologia , Receptores Toll-Like/fisiologia
7.
Biosci Rep ; 40(8)2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32725155

RESUMO

Mef2 transcription factors comprise a family of four different isoforms that regulate a number of processes including neuronal and muscle development. While roles for Mef2C and Mef2D have been described in B-cell development their role in immunity has not been extensively studied. In innate immune cells such as macrophages, TLRs drive the production of both pro- and anti-inflammatory cytokines. IL-10 is an important anti-inflammatory cytokine produced by macrophages and it establishes an autocrine feedback loop to inhibit pro-inflammatory cytokine production. We show here that macrophages from Mef2D knockout mice have elevated levels of IL-10 mRNA induction compared with wild-type cells following LPS stimulation. The secretion of IL-10 was also higher from Mef2D knockout macrophages and this correlated to a reduction in the secretion of TNF, IL-6 and IL-12p40. The use of an IL-10 neutralising antibody showed that this reduction in pro-inflammatory cytokine production in the Mef2D knockouts was IL-10 dependent. As the IL-10 promoter has previously been reported to contain a potential binding site for Mef2D, it is possible that the binding of other Mef2 isoforms in the absence of Mef2D may result in a higher activation of the IL-10 gene. Further studies with compound Mef2 isoforms would be required to address this. We also show that Mef2D is highly expressed in the thymus, but that loss of Mef2D does not affect thymic T-cell development or the production of IFNγ from CD8 T cells.


Assuntos
Interleucina-10/metabolismo , Macrófagos/metabolismo , Receptores Toll-Like/metabolismo , Animais , Células Cultivadas , Mediadores da Inflamação/metabolismo , Interleucina-10/genética , Lipopolissacarídeos/farmacologia , Fatores de Transcrição MEF2/deficiência , Fatores de Transcrição MEF2/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptores Toll-Like/agonistas , Regulação para Cima
8.
J Exp Med ; 215(11): 2737-2747, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30337469

RESUMO

TPL-2 MAP 3-kinase promotes inflammation in numerous mouse disease models and is an attractive anti-inflammatory drug target. However, TPL-2-deficient (Map3k8 -/-) mice develop exacerbated allergic airway inflammation to house dust mite (HDM) compared with wild type controls. Here, we show that Map3k8D270A/D270A mice expressing kinase dead TPL-2 had an unaltered response to HDM, indicating that the severe airway inflammation observed in Map3k8 -/- mice is not due to blockade of TPL-2 signaling and rather reflects a TPL-2 adaptor function. Severe allergic inflammation in TPL-2-deficient mice was likely due to reduced levels of ABIN-2 (TNIP2), whose stability depends on TPL-2 expression. Tnip2E256K knock-in mutation, which reduced ABIN-2 binding to A20, augmented the HDM-induced airway inflammation, but did not affect TPL-2 expression or signaling. These results identify ABIN-2 as a novel negative regulator of allergic airway responses and importantly indicate that TPL-2 inhibitors would not have unwanted allergic comorbidities.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Hipersensibilidade/imunologia , MAP Quinase Quinase Quinases/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Hipersensibilidade/genética , Hipersensibilidade/patologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/imunologia
9.
J Med Chem ; 58(16): 6589-606, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26258521

RESUMO

The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a critical immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently observed in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacological probes and as clinical drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochemical and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compounds 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compound 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacologically interrogate JAK3-dependent biology.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Janus Quinase 3/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Animais , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Masculino , Camundongos , Modelos Moleculares , Inibidores de Proteínas Quinases/farmacocinética , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
10.
FEBS Lett ; 587(10): 1496-503, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23542035

RESUMO

Chemokines, including MCP-1, are crucial to mounting an effective immune response due to their ability to recruit other immune cells. We show that sustained LPS or poly(I:C)-stimulated MCP-1 production requires an IFNß-mediated feedback loop. Consistent with this, exogenous IFNß was able to induce MCP-1 transcription in the absence of other stimuli. Blocking IFNß signaling with Ruxolitinib, a JAK inhibitor, inhibited MCP-1 transcription. The MCP-1 promoter contains potential STAT binding sites and we demonstrate that STAT1 is recruited upon IFNß stimulation. Furthermore we find that IL-10 knockout increases MCP-1 production in response to LPS, which may reflect an ability of IL-10 to repress IFNß production. Overall, these results show the importance of the balance between IFNß and IL-10 in the regulation of MCP-1.


Assuntos
Comunicação Autócrina/fisiologia , Quimiocina CCL2/genética , Retroalimentação Fisiológica/fisiologia , Interferon beta/fisiologia , Macrófagos/metabolismo , Receptores Toll-Like/fisiologia , Animais , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/genética , Células Cultivadas , Quimiocina CCL2/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Interferon beta/metabolismo , Interferon beta/farmacologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-10/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Receptores Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
11.
J Med Chem ; 56(11): 4413-21, 2013 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-23656407

RESUMO

The protein kinase ERK5 (MAPK7) is an emerging drug target for a variety of indications, in particular for cancer where it plays a key role mediating cell proliferation, survival, epithelial-mesenchymal transition, and angiogenesis. To date, no three-dimensional structure has been published that would allow rational design of inhibitors. To address this, we determined the X-ray crystal structure of the human ERK5 kinase domain in complex with a highly specific benzo[e]pyrimido[5,4-b]diazepine-6(11H)-one inhibitor. The structure reveals that specific residue differences in the ATP-binding site, compared to the related ERKs p38s and JNKs, allow for the development of ERK5-specific inhibitors. The selectivity of previously observed ERK5 inhibitors can also be rationalized using this structure, which provides a template for future development of inhibitors with potential for treatment of disease.


Assuntos
Azepinas/química , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/química , Pirimidinas/química , Sequência de Aminoácidos , Azepinas/farmacologia , Cristalografia por Raios X , Células HEK293 , Células HeLa , Humanos , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Conformação Proteica , Pirimidinas/farmacologia , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA