Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36902103

RESUMO

Scleroderma is a chronic fibrotic disease, where proinflammatory and profibrotic events precede collagen accumulation. MKP-1 [mitogen-activated protein kinase (MAPK) phosphatase-1] downregulates inflammatory MAPK pathways suppressing inflammation. MKP-1 also supports Th1 polarization, which could shift Th1/Th2 balance away from profibrotic Th2 profile prevalent in scleroderma. In the present study, we investigated the potential protective role of MKP-1 in scleroderma. We utilized bleomycin-induced dermal fibrosis model as a well-characterized experimental model of scleroderma. Dermal fibrosis and collagen deposition as well as the expression of inflammatory and profibrotic mediators were analyzed in the skin samples. Bleomycin-induced dermal thickness and lipodystrophy were increased in MKP-1-deficient mice. MKP-1 deficiency enhanced collagen accumulation and increased expression of collagens, 1A1 and 3A1, in the dermis. Bleomycin-treated skin from MKP-1-deficient mice also showed enhanced expression of inflammatory and profibrotic factors IL-6, TGF-ß1, fibronectin-1 and YKL-40, and chemokines MCP-1, MIP-1α and MIP-2, as compared to wild-type mice. The results show, for the first time, that MKP-1 protects from bleomycin-induced dermal fibrosis, suggesting that MKP-1 favorably modifies inflammation and fibrotic processes that drive the pathogenesis of scleroderma. Compounds enhancing the expression or activity of MKP-1 could thus prevent fibrotic processes in scleroderma and possess potential as a novel immunomodulative drug.


Assuntos
Fosfatase 1 de Especificidade Dupla , Escleroderma Sistêmico , Pele , Animais , Camundongos , Bleomicina , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrose , Inflamação/patologia , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/patologia , Pele/enzimologia , Pele/patologia , Fosfatase 1 de Especificidade Dupla/deficiência
2.
J Immunol ; 206(12): 2966-2979, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34039638

RESUMO

We have previously shown that Mkp-1-deficient mice produce elevated TNF-α, IL-6, and IL-10 following systemic Escherichia coli infection, and they exhibited increased mortality, elevated bacterial burden, and profound metabolic alterations. To understand the function of Mkp-1 during bacterial infection, we performed RNA-sequencing analysis to compare the global gene expression between E. coli-infected wild-type and Mkp-1 -/- mice. A large number of IFN-stimulated genes were more robustly expressed in E. coli-infected Mkp-1 -/- mice than in wild-type mice. Multiplex analysis of the serum cytokine levels revealed profound increases in IFN-ß, IFN-γ, TNF-α, IL-1α and ß, IL-6, IL-10, IL-17A, IL-27, and GMSF levels in E. coli-infected Mkp-1 -/- mice relative to wild-type mice. Administration of a neutralizing Ab against the receptor for type I IFN to Mkp-1 -/- mice prior to E. coli infection augmented mortality and disease severity. Mkp-1 -/- bone marrow-derived macrophages (BMDM) produced higher levels of IFN-ß mRNA and protein than did wild-type BMDM upon treatment with LPS, E. coli, polyinosinic:polycytidylic acid, and herring sperm DNA. Augmented IFN-ß induction in Mkp-1 -/- BMDM was blocked by a p38 inhibitor but not by an JNK inhibitor. Enhanced Mkp-1 expression abolished IFN-ß induction by both LPS and E. coli but had little effect on the IFN-ß promoter activity in LPS-stimulated RAW264.7 cells. Mkp-1 deficiency did not have an overt effect on IRF3/7 phosphorylation or IKK activation but modestly enhanced IFN-ß mRNA stability in LPS-stimulated BMDM. Our results suggest that Mkp-1 regulates IFN-ß production primarily through a p38-mediated mechanism and that IFN-ß plays a beneficial role in E. coli-induced sepsis.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Infecções por Escherichia coli/metabolismo , Interferon beta/metabolismo , Animais , Células Cultivadas , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/imunologia , Infecções por Escherichia coli/imunologia , Interferon beta/genética , Interferon beta/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células RAW 264.7 , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Immunohorizons ; 4(5): 245-258, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32414764

RESUMO

Sepsis is the leading cause of death in the world. Recent reports suggest that in response to sepsis, metabolism of macrophages switches from oxidative phosphorylation to aerobic glycolysis. MAPK phosphatase (MKP)-1 (also known as DUSP1) localized in the nucleus and preferentially dephosphorylates p38 and JNK. MKP-1 controls the expression of numerous inflammatory genes and transcription factors, thereby regulating innate and adaptive immunity. MKP-1-deficient animals exhibit aberrant metabolic responses following bacterial infections with a markedly increased mortality in response to sepsis. Because metabolic reprogramming modulates immune responses to TLR-4 activation, we investigated the effect of MKP-1 deficiency on mitochondrial electron transport chains involved in oxidative phosphorylation and transcription factors regulating mitochondrial biogenesis. Mitochondrial biogenesis is regulated by three nuclear-encoded proteins, including transcription factor A (TFAM), nuclear respiratory factors (NRF-1), and peroxisome proliferator-activated receptor γ coactivator-1-α (PGC-1α). We show that MKP-1-deficient mice/macrophages exhibit, at baseline, higher expression of oxidative phosphorylation, TFAM, PGC-1α, and NRF-1 associated with increased respiration and production of reactive oxygen species as compared with wild-type mice. Surprisingly, MKP-1-deficient mice/macrophages responded to Escherichia coli sepsis or LPS with an impaired metabolic switch; despite enhanced glycolysis, a preserved mitochondrial function and biogenesis are exhibited. Furthermore, inhibition of p38 MAPK had no significant effect on TFAM and NRF-1 either in MKP-1-deficient macrophages or in wild-type macrophages. These findings support the conclusion that MKP-1 plays an important role in regulating proteins involved in glycolysis and oxidative phosphorylation and modulates expression of mitochondrial transcription factors.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Glicólise , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Fator 1 Nuclear Respiratório/metabolismo , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sepse/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
4.
Elife ; 82019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30938680

RESUMO

Mitogen-activated protein kinases (MAPK) such as p38 and the c-Jun N-terminal kinases (JNKs) are activated during the cellular response to stress signals. Their activity is regulated by the MAPK-phosphatase 1 (DUSP1), a key component of the anti-inflammatory response. Stress kinases are well-described elements of the response to otic injury and the otoprotective potential of JNK inhibitors is being tested in clinical trials. By contrast, there are no studies exploring the role of DUSP1 in hearing and hearing loss. Here we show that Dusp1 expression is age-regulated in the mouse cochlea. Dusp1 gene knock-out caused premature progressive hearing loss, as confirmed by auditory evoked responses in Dusp1-/- mice. Hearing loss correlated with cell death in hair cells, degeneration of spiral neurons and increased macrophage infiltration. Dusp1-/- mouse cochleae showed imbalanced redox status and dysregulated expression of cytokines. These data suggest that DUSP1 is essential for cochlear homeostasis in the response to stress during ageing.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Perda Auditiva/fisiopatologia , Estimulação Acústica , Animais , Cóclea/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Auditivos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução
5.
Int J Mol Sci ; 19(12)2018 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-30563203

RESUMO

Mitogen-activated protein kinase phosphatase (Mkp)-1 exerts its anti-inflammatory activities during Gram-negative sepsis by deactivating p38 and c-Jun N-terminal kinase (JNK). We have previously shown that Mkp-1+/+ mice, but not Mkp-1-/- mice, exhibit hypertriglyceridemia during severe sepsis. However, the regulation of hepatic lipid stores and the underlying mechanism of lipid dysregulation during sepsis remains an enigma. To understand the molecular mechanism underlying the sepsis-associated metabolic changes and the role of Mkp-1 in the process, we infected Mkp-1+/+ and Mkp-1-/- mice with Escherichia coli i.v., and assessed the effects of Mkp-1 deficiency on tissue lipid contents. We also examined the global gene expression profile in the livers via RNA-seq. We found that in the absence of E. coli infection, Mkp-1 deficiency decreased liver triglyceride levels. Upon E. coli infection, Mkp-1+/+ mice, but not Mkp-1-/- mice, developed hepatocyte ballooning and increased lipid deposition in the livers. E. coli infection caused profound changes in the gene expression profile of a large number of proteins that regulate lipid metabolism in wildtype mice, while these changes were substantially disrupted in Mkp-1-/- mice. Interestingly, in Mkp-1+/+ mice E. coli infection resulted in downregulation of genes that facilitate fatty acid synthesis but upregulation of Cd36 and Dgat2, whose protein products mediate fatty acid uptake and triglyceride synthesis, respectively. Taken together, our studies indicate that sepsis leads to a substantial change in triglyceride metabolic gene expression programs and Mkp-1 plays an important role in this process.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Infecções por Escherichia coli/genética , Perfilação da Expressão Gênica/métodos , Metabolismo dos Lipídeos , Sepse/genética , Animais , Infecções por Escherichia coli/metabolismo , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Fígado/química , Redes e Vias Metabólicas , Camundongos , Sepse/metabolismo , Sepse/microbiologia , Análise de Sequência de RNA , Triglicerídeos/metabolismo
6.
Front Immunol ; 9: 569, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29619028

RESUMO

Persistent activation of mitogen-activated protein kinase (MAPK) is believed to be involved in psoriasis pathogenesis. MAPK phosphatase-1 (MKP-1) is an important negative regulator of MAPK activity, but the cellular and molecular mechanisms of MKP-1 in psoriasis development are largely unknown. In this study, we found that the expression of MKP-1 was decreased in the imiquimod (IMQ)-induced psoriasiform mouse skin. MKP-1-deficient (MKP-1-/-) mice were highly susceptible to IMQ-induced skin inflammation, which was associated with increased production of inflammatory cytokines and chemokines. MKP-1 acted on both hematopoietic and non-hematopoietic cells to regulate psoriasis pathogenesis. MKP-1 deficiency in macrophages led to enhanced p38 activation and higher expression of interleukin (IL)-1ß, CXCL2, and S100a8 upon R848 stimulation. Moreover, MKP-1 deficiency in the non-hematopoietic compartments led to an enhanced IL-22 receptor signaling and higher expression of CXCL1 and CXCL2 upon IMQ treatment. Collectively, our data suggest a critical role for MKP-1 in the regulation of skin inflammation.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Psoríase/enzimologia , Dermatopatias/enzimologia , Pele/enzimologia , Animais , Citocinas/genética , Citocinas/metabolismo , Fosfatase 1 de Especificidade Dupla/genética , Imiquimode , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Psoríase/induzido quimicamente , Psoríase/genética , Índice de Gravidade de Doença , Pele/metabolismo , Pele/patologia , Dermatopatias/induzido quimicamente , Dermatopatias/genética
7.
Mol Cell Biol ; 37(11)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28265004

RESUMO

The mRNA-destabilizing factor tristetraprolin (TTP) binds in a sequence-specific manner to the 3' untranslated regions of many proinflammatory mRNAs and recruits complexes of nucleases to promote rapid mRNA turnover. Mice lacking TTP develop a severe, spontaneous inflammatory syndrome characterized by the overexpression of tumor necrosis factor and other inflammatory mediators. However, TTP also employs the same mechanism to inhibit the expression of the potent anti-inflammatory cytokine interleukin 10 (IL-10). Perturbation of TTP function may therefore have mixed effects on inflammatory responses, either increasing or decreasing the expression of proinflammatory factors via direct or indirect mechanisms. We recently described a knock-in mouse strain in which the substitution of 2 amino acids of the endogenous TTP protein renders it constitutively active as an mRNA-destabilizing factor. Here we investigate the impact on the IL-10-mediated anti-inflammatory response. It is shown that the gain-of-function mutation of TTP impairs IL-10-mediated negative feedback control of macrophage function in vitro However, the in vivo effects of TTP mutation are uniformly anti-inflammatory despite the decreased expression of IL-10.


Assuntos
Retroalimentação Fisiológica , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Mutação/genética , Tristetraprolina/genética , Animais , Células da Medula Óssea/metabolismo , Citocinas/metabolismo , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/metabolismo , Perfilação da Expressão Gênica , Inflamação/genética , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Transcrição Gênica
8.
Cell Signal ; 34: 1-10, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28238855

RESUMO

Interleukin 1 beta (IL-1ß) is a pro-inflammatory cytokine that plays a major role in inflammatory diseases as well as cancer. The inflammatory response after Toll-like receptor (TLR) 4 activation is tightly regulated through phosphorylation of MAP kinases, including p38 and JNK pathways. The activation of MAP kinases is negatively regulated by MAPK phosphatases (MKPs). MKP-1 preferentially dephosphorylates p38 and JNK. IL-1ß is regulated through the activation of MAPK, including p38 as well as several transcription factors. The oxygen-sensitive transcription factor HIF-1α is a known transcription factor for several inflammatory cytokines including IL-1ß and IL-6. Here, we report that MKP-1 regulates HIF-1α expression in response to LPS. MKP-1 deficient bone marrow derived macrophages (BMDMs) exhibited increased reactive oxygen species (ROS) production and higher HIF-1α expression. In contrast, the expression of all three isoforms of prolyl hydroxylases (PHDs), which are important in destabilizing HIF-1α through hydroxylation, were significantly decreased in MKP-1 deficient BMDMs. LPS challenge of MKP-1 deficient BMDMs led to a substantial increase in IL-1ß production. An inhibitor of HIF-1α significantly decreased LPS mediated IL-1ß production both at the transcript and protein levels. Similarly, inhibition of p38 MAP kinase reduced LPS mediated pro-IL-1ß and HIF-1α protein levels as well as ROS production in MKP-1 deficient BMDMs. These findings demonstrate a regulatory function for MKP-1 in modulating IL-1ß expression through p38 activation, ROS production and HIF-1α expression.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipopolissacarídeos/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antracenos/farmacologia , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Equinomicina/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Interleucina-1beta/análise , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
10.
Atherosclerosis ; 235(2): 613-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24956536

RESUMO

OBJECTIVE: The c-Jun N-terminal kinase (JNK) family regulates fundamental physiological processes including apoptosis and metabolism. Although JNK2 is known to promote foam cell formation during atherosclerosis, the potential role of JNK1 is uncertain. We examined the potential influence of JNK1 and its negative regulator, MAP kinase phosphatase-1 (MKP-1), on endothelial cell (EC) injury and early lesion formation using hypercholesterolemic LDLR(-/-) mice. METHODS AND RESULTS: To assess the function of JNK1 in early atherogenesis, we measured EC apoptosis and lesion formation in LDLR(-/-) or LDLR(-/-)/JNK1(-/-) mice exposed to a high fat diet for 6 weeks. En face staining using antibodies that recognise active, cleaved caspase-3 (apoptosis) or using Sudan IV (lipid deposition) revealed that genetic deletion of JNK1 reduced EC apoptosis and lesion formation in hypercholesterolemic mice. By contrast, although EC apoptosis was enhanced in LDLR(-/-)/MKP-1(-/-) mice compared to LDLR(-/-) mice, lesion formation was unaltered. CONCLUSION: We conclude that JNK1 is required for EC apoptosis and lipid deposition during early atherogenesis. Thus pharmacological inhibitors of JNK may reduce atherosclerosis by preventing EC injury as well as by influencing foam cell formation.


Assuntos
Células Endoteliais/patologia , Hipercolesterolemia/fisiopatologia , Proteína Quinase 8 Ativada por Mitógeno/deficiência , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Dieta Hiperlipídica , Fosfatase 1 de Especificidade Dupla/deficiência , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo
11.
Exp Neurol ; 261: 404-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24842488

RESUMO

BACKGROUND AND PURPOSE: Activation of mitogen-activated protein kinases (MAPKs), particularly c-jun-N-terminal kinases (JNK) and p38 exacerbates stroke injury by provoking pro-apoptotic and pro-inflammatory cellular signaling. MAPK phosphatase-1 (MKP-1) restrains the over-activation of MAPKs via rapid de-phosphorylation of the MAPKs. We therefore examined the role of MKP-1 in stroke and studied its inhibitory effects on MAPKs after experimental stroke. METHODS: Male mice were subjected to transient middle cerebral artery occlusion (MCAO). MKP-1 knockout (KO) mice and a MKP-1 pharmacological inhibitor were utilized. We utilized flow cytometry, immunohistochemistry (IHC), and Western blots analysis to explore MKP-1 signaling and its effects on apoptosis/inflammation in the brain and specifically in microglia after stroke. RESULTS: MKP-1 was highly expressed in the nuclei of both neurons and microglia after stroke. MKP-1 genetic deletion exacerbated stroke outcome by increasing infarct, neurological deficits and hemorrhagic transformation. Additionally, delayed treatment of the MKP-1 pharmacological inhibitor worsened stroke outcome in wild type (WT) mice but had no effect in MKP-1 KO mice. Furthermore, MKP-1 deletion led to increased c-jun-N-terminal kinase (JNK) activation and microglial p38 activation after stroke. Finally, MKP-1 deletion or inhibition increased inflammatory and apoptotic response as evidenced by the increased levels of interleukin-6 (IL-6), tumor necrosis factor α (TNFα), ratio of p-c-jun/c-jun and cleaved caspase-3 following ischemia. CONCLUSIONS: We have demonstrated that MKP-1 signaling is an endogenous protective mechanism in stroke. Our data imply that MKP-1 possesses anti-apoptotic and anti-inflammatory properties by simultaneously controlling the activities of JNK and microglial p38.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Encefalite/etiologia , Regulação Enzimológica da Expressão Gênica/genética , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/etiologia , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Cicloexilaminas/efeitos adversos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fosfatase 1 de Especificidade Dupla/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Indenos/efeitos adversos , Infarto da Artéria Cerebral Média/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos Knockout , Exame Neurológico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 34(5): 1020-31, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24578378

RESUMO

OBJECTIVE: Angiogenesis is the formation of new blood vessels through endothelial cell sprouting. This process requires the mitogen-activated protein kinases, signaling molecules that are negatively regulated by the mitogen-activated protein kinase phosphatase-1 (MKP-1). The purpose of this study was to evaluate the role of MKP-1 in neovascularization in vivo and identify associated mechanisms in endothelial cells. APPROACH AND RESULTS: We used murine hindlimb ischemia as a model system to evaluate the role of MKP-1 in angiogenic growth, remodeling, and arteriogenesis in vivo. Genomic deletion of MKP-1 blunted angiogenesis in the distal hindlimb and microvascular arteriogenesis in the proximal hindlimb. In vitro, endothelial MKP-1 depletion/deletion abrogated vascular endothelial growth factor-induced migration and tube formation, and reduced proliferation. These observations establish MKP-1 as a positive mediator of angiogenesis and contrast with the canonical function of MKP-1 as a mitogen-activated protein kinase phosphatase, implying an alternative mechanism for MKP-1-mediated angiogenesis. Cloning and sequencing of MKP-1-bound chromatin identified localization of MKP-1 to exonic DNA of the angiogenic chemokine fractalkine, and MKP-1 depletion reduced histone H3 serine 10 dephosphorylation on this DNA locus and blocked fractalkine expression. In vivo, MKP-1 deletion abrogated ischemia-induced fractalkine expression and macrophage and T-lymphocyte infiltration in distal hindlimbs, whereas fractalkine delivery to ischemic hindlimbs rescued the effect of MKP-1 deletion on neovascular hindlimb recovery. CONCLUSIONS: MKP-1 promoted angiogenic and arteriogenic neovascular growth, potentially through dephosphorylation of histone H3 serine 10 on coding-region DNA to control transcription of angiogenic genes, such as fractalkine. These observations reveal a novel function for MKP-1 and identify MKP-1 as a potential therapeutic target.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Células Endoteliais/enzimologia , Isquemia/enzimologia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Animais , Sítios de Ligação , Movimento Celular , Proliferação de Células , Células Cultivadas , Quimiocina CX3CL1/administração & dosagem , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Modelos Animais de Doenças , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Éxons , Regulação da Expressão Gênica , Membro Posterior , Histonas/metabolismo , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Isquemia/genética , Isquemia/fisiopatologia , Isquemia/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/genética , Fosforilação , Interferência de RNA , Serina , Transdução de Sinais , Fatores de Tempo , Transfecção
13.
Bone ; 60: 16-25, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24269279

RESUMO

Cytokine-directed osteoclastogenesis is initiated in response to macrophage colony stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) to drive formation of osteoclasts (OC), large bone resorptive cells of hematopoietic origin. RANKL-induced signaling activates the MAPK pathways, which initiates nuclear translocation of the master regulator of osteoclast formation, transcription factor NFATc1. Proper control over these signaling events is essential to normal OC formation response to stimuli. MAPK phosphatase 1 (MKP-1), a serine and tyrosine phosphatase encoded by the gene Dusp1, functions to dephosphorylate and subsequently inactivate MAPK (p38 and JNK) signaling essential in osteoclastogenesis. Here, we explored the role of MKP-1 during RANKL-driven osteoclastogenesis from defined (B220/CD45(-)GR1(-)CD11b(lo/-)CD115(+)) OC progenitor (dOCP) populations using WT and Dusp1(-/-) global knockout mice. Sorted cells were driven to OC by M-CSF pre-treatment followed by RANKL stimulation for 3days. OC formation and qPCR products were analyzed for maturation. Results indicate that Dusp1(-/-) dOCP form less numerous, significantly smaller and less functional OC compared to WT controls. These data were corroborated by mRNA expression of the key OC genes, Nfatc1 and Tm7sf4 (DC-STAMP), which were significantly reduced in early osteoclastogenesis in OC progenitor from Dusp1(-/-) mice. Intriguingly, our data reveals that MKP-1 may positively control OC formation in response to RANKL by regulating NFATc1 nuclear translocation. Collectively, this report supports the idea that MKP-1 signaling is essential in early osteoclastogenesis in response to RANKL-induced signaling.


Assuntos
Linhagem da Célula/efeitos dos fármacos , Núcleo Celular/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/enzimologia , Osteogênese/efeitos dos fármacos , Ligante RANK/farmacologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Fusão Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Fosfatase 1 de Especificidade Dupla/deficiência , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/genética , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
BMC Genomics ; 14: 104, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23414127

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are short non-coding regulatory RNAs that control gene expression usually producing translational repression and gene silencing. High-throughput sequencing technologies have revealed heterogeneity at length and sequence level for the majority of mature miRNAs (IsomiRs). Most isomiRs can be explained by variability in either Dicer1 or Drosha cleavage during miRNA biogenesis at 5' or 3' of the miRNA (trimming variants). Although isomiRs have been described in different tissues and organisms, their functional validation as modulators of gene expression remains elusive. Here we have characterized the expression and function of a highly abundant miR-101 5'-trimming variant (5'-isomiR-101). RESULTS: The analysis of small RNA sequencing data in several human tissues and cell lines indicates that 5'-isomiR-101 is ubiquitously detected and a highly abundant, especially in the brain. 5'-isomiR-101 was found in Ago-2 immunocomplexes and complementary approaches showed that 5'-isomiR-101 interacted with different members of the silencing (RISC) complex. In addition, 5'-isomiR-101 decreased the expression of five validated miR-101 targets, suggesting that it is a functional variant. Both the binding to RISC members and the degree of silencing were less efficient for 5'-isomiR-101 compared with miR-101. For some targets, both miR-101 and 5'-isomiR-101 significantly decreased protein expression with no changes in the respective mRNA levels. Although a high number of overlapping predicted targets suggest similar targeted biological pathways, a correlation analysis of the expression profiles of miR-101 variants and predicted mRNA targets in human brains at different ages, suggest specific functions for miR-101- and 5'-isomiR-101. CONCLUSIONS: These results suggest that isomiRs are functional variants and further indicate that for a given miRNA, the different isomiRs may contribute to the overall effect as quantitative and qualitative fine-tuners of gene expression.


Assuntos
Regulação da Expressão Gênica/genética , Inativação Gênica , MicroRNAs/genética , Isoformas de RNA/genética , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Proteínas Argonautas/metabolismo , Linhagem Celular , Ciclo-Oxigenase 2/deficiência , Ciclo-Oxigenase 2/genética , RNA Helicases DEAD-box/metabolismo , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , MicroRNAs/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , Complexo Repressor Polycomb 2/deficiência , Complexo Repressor Polycomb 2/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/deficiência , Proteínas Proto-Oncogênicas c-bcl-2/genética , Isoformas de RNA/metabolismo
15.
Circ Res ; 112(1): 48-56, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22993413

RESUMO

RATIONALE: Mitogen-activated protein kinases (MAPKs) are activated in the heart by disease-inducing and stress-inducing stimuli, where they participate in hypertrophy, remodeling, contractility, and heart failure. A family of dual-specificity phosphatases (DUSPs) directly inactivates each of the MAPK terminal effectors, potentially serving a cardioprotective role. OBJECTIVE: To determine the role of DUSP1 and DUSP4 in regulating p38 MAPK function in the heart and the effect on disease. METHODS AND RESULTS: Here, we generated mice and mouse embryonic fibroblasts lacking both Dusp1 and Dusp4 genes. Although single nulls showed no molecular effects, combined disruption of Dusp1/4 promoted unrestrained p38 MAPK activity in both mouse embryonic fibroblasts and the heart, with no change in the phosphorylation of c-Jun N-terminal kinases or extracellular signal-regulated kinases at baseline or with stress stimulation. Single disruption of either Dusp1 or Dusp4 did not result in cardiac pathology, although Dusp1/4 double-null mice exhibited cardiomyopathy and increased mortality with aging. Pharmacological inhibition of p38 MAPK with SB731445 ameliorated cardiomyopathy in Dusp1/4 double-null mice, indicating that DUSP1/4 function primarily through p38 MAPK in affecting disease. At the cellular level, unrestrained p38 MAPK activity diminished cardiac contractility and Ca2+ handling, which was acutely reversed with a p38 inhibitory compound. Poor function in Dusp1/4 double-null mice also was partially rescued by phospholamban deletion. CONCLUSIONS: Our data demonstrate that Dusp1 and Dusp4 are cardioprotective genes that play a critical role in the heart by dampening p38 MAPK signaling that would otherwise reduce contractility and induce cardiomyopathy.


Assuntos
Cardiomiopatias/enzimologia , Fosfatase 1 de Especificidade Dupla/deficiência , Miócitos Cardíacos/enzimologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Cardiomiopatias/diagnóstico , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Cardiomiopatias/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Fosfatase 1 de Especificidade Dupla/genética , Ativação Enzimática , Fibroblastos/enzimologia , Regulação da Expressão Gênica , Hemodinâmica , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Fosfatases , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
16.
Toxicol Pathol ; 40(8): 1095-105, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22623522

RESUMO

c-Jun N-terminal kinase (JNK) activation promotes hepatocyte death during acetaminophen overdose, a common cause of drug-induced liver failure. While mitogen-activated protein kinase (MAPK) phosphatase (Mkp)-1 is a critical negative regulator of JNK MAPK, little is known about the role of Mkp-1 during hepatotoxicity. In this study, we evaluated the role of Mkp-1 during acute acetaminophen toxicity. Mkp-1⁺/⁺ and Mkp-1⁻/⁻ mice were dosed ip with vehicle or acetaminophen at 300 mg/kg (for mechanistic studies) or 400 mg/kg (for survival studies). Tissues were collected 1-6 hr post 300 mg/kg dosing to assess glutathione levels, organ damage, and MAPK activation. Mkp-1⁻/⁻ mice exhibited more rapid plasma clearance of acetaminophen than did Mkp-1⁺/⁺ mice, indicated by a quicker decline of plasma acetaminophen level. Moreover, Mkp-1⁻/⁻ mice suffered more severe liver injury, indicated by higher plasma alanine transaminase activity and more extensive centrilobular apoptosis and necrosis. Hepatic JNK activity in Mkp-1⁻/⁻ mice was higher than in Mkp-1⁺/⁺ mice. Finally, Mkp-1⁻/⁻ mice displayed a lower overall survival rate and shorter median survival time after dosing with 400 mg/kg acetaminophen. The more severe phenotype exhibited by Mkp-1⁻/⁻ mice indicates that Mkp-1 plays a protective role during acute acetaminophen overdose, potentially through regulation of JNK.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Fosfatase 1 de Especificidade Dupla/metabolismo , Acetaminofen/farmacocinética , Alanina Transaminase/sangue , Analgésicos não Narcóticos/farmacocinética , Animais , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/genética , Modelos Animais de Doenças , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Regulação Enzimológica da Expressão Gênica , Glutationa/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Testes de Função Hepática , Longevidade/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Necrose/induzido quimicamente , Necrose/patologia
17.
Cell Death Differ ; 19(10): 1634-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22522596

RESUMO

The dual specificity phosphatase MAPK phosphatase-1 (MKP-1) feeds back on MAP kinase signaling to regulate metabolic, inflammatory and survival responses. MKP-1 is widely expressed in the central nervous system (CNS) and induced after ischemic stress, although its function in these contexts remains unclear. Here we report that MKP-1 activated several cell death factors, including BCL2 and adenovirus E1B 19 kDa interacting protein 3, and caspases 3 and 12 culminating in apoptotic cell death in vitro. MKP-1 also exerted inhibitory effects on the bZIP transcription factor CCAAT/enhancer-binding protein (C/EBPß), previously shown to have neuroprotective properties. These effects included reduced expression of the full-length C/EBPß variant and hypo-phosphorylation at the MEK-ERK1/2-sensitive Thr(188) site. Notably, enforced expression C/EBPß rescued cells from MKP-1-induced toxicity. Studies performed in knock-out mice indicate that the MKP-1 activity is required to exclude C/EBPß from the nucleus basally, and that MKP-1 antagonizes C/EBPß expression after global forebrain ischemia, particularly within the vulnerable CA1 sector of the hippocampus. Overall, MKP-1 appears to lower the cellular apoptotic threshold by inhibiting C/EBPß and enhancing both BH3 protein expression and cellular caspase activity. Thus, although manipulation of the MKP-1-C/EBPß axis could have therapeutic value in ischemic disorders, our observations using MKP-1 catalytic mutants suggest that approaches geared towards inhibiting MKP-1's phosphatase activity alone may be ineffective.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Animais , Apoptose , Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Caspase 12/metabolismo , Caspase 3/metabolismo , Hipóxia Celular , Linhagem Celular , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mitocondriais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Ativação Transcricional
18.
Cell Cycle ; 11(5): 877-86, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22361726

RESUMO

Re-establishing tissue homoeostasis in response to injury requires infiltration of inflammatory cells and activation of resident stem cells. However, full tissue recovery also requires that the inflammation is resolved. While it is known that disturbing the interactions between inflammatory cells and tissue resident cells prevents successful healing, the molecular mechanisms underlying the paracrine interactions between these cell types are practically unknown. Here, and in a recent study, we provide mechanistic evidence that macrophages control stem cell-dependent tissue repair. In particular, we found that the temporal spacing of the pro- to anti-inflammatory macrophage polarization switch is controlled by the balance of p38 MAPK (termed here p38) and the MAPK phosphatase MKP-1 during the muscle healing process. Moreover, we demonstrate a new function for MKP-1-regulated p38 signaling in deactivating macrophages during inflammation resolution after injury. Specifically, at advanced stages of regeneration, MKP-1 loss caused an unscheduled "exhaustion-like" state in muscle macrophages, in which neither pro- nor anti-inflammatory cytokines are expressed despite persistent tissue damage, leading to dysregulated reparation by the tissue stem cells. Mechanistically, we demonstrate that p38 and MKP-1 control the AKT pathway through a miR-21-dependent PTEN regulation. Importantly, both genetic and pharmacological interference with the individual components of this pathway restored inflammation-dependent tissue homeostasis in MKP-1-deficient mice and delayed inflammation resolution and tissue repair dysregulation in wild-type mice. Because the process of tolerance to bacterial infection involves a progressive attenuation of pro-inflammatory gene expression, we discuss here the potential similarities between the mechanisms underlying inflammation resolution during tissue repair and those controlling endotoxin tolerance.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Macrófagos/metabolismo , Células-Tronco/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/imunologia , Camundongos , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células-Tronco/imunologia , Cicatrização , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
J Immunol ; 188(5): 2127-35, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22301548

RESUMO

It is estimated that 1 billion people around the world are vitamin D deficient. Vitamin D deficiency has been linked to various inflammatory diseases. However, the mechanism by which vitamin D reduces inflammation remains poorly understood. In this study, we investigated the inhibitory effects of physiologic levels of vitamin D on LPS-stimulated inflammatory response in human blood monocytes and explored potential mechanisms of vitamin D action. We observed that two forms of the vitamin D, 1,25(OH)(2)D(3), and 25(OH)D(3), dose dependently inhibited LPS-induced p38 phosphorylation at physiologic concentrations, IL-6 and TNF-α production by human monocytes. Upon vitamin D treatment, the expression of MAPK phosphatase-1 (MKP-1) was significantly upregulated in human monocytes and murine bone marrow-derived macrophages (BMM). Increased binding of the vitamin D receptor and increased histone H4 acetylation at the identified vitamin D response element of the murine and human MKP-1 promoters were demonstrated. Moreover, in BMM from MKP1(-/-) mice, the inhibition of LPS-induced p38 phosphorylation by vitamin D was completely abolished. Vitamin D inhibition of LPS-induced IL-6 and TNF-α production by BMM from MKP-1(-/-) mice was significantly reduced as compared with wild-type mice. In conclusion, this study identified the upregulation of MKP-1 by vitamin D as a novel pathway by which vitamin D inhibits LPS-induced p38 activation and cytokine production in monocytes/macrophages.


Assuntos
Citocinas/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Fosfatase 1 de Especificidade Dupla/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Vitamina D/farmacologia , Adulto , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/enzimologia , Células da Medula Óssea/imunologia , Células Cultivadas , Citocinas/biossíntese , Sistemas de Liberação de Medicamentos/métodos , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Humanos , Mediadores da Inflamação/metabolismo , Células L , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/genética , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Cell Signal ; 24(2): 560-568, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22037550

RESUMO

The dual-specificity mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1) inactivates MAP kinases by dephosphorylation. Here we show that the proinflammatory cytokine interleukin (IL)-17A induces adult mouse primary cardiac fibroblast (CF) proliferation and migration via IL-17 receptor A//IL-17 receptor C-dependent MKP-1 suppression, and activation of p38 MAPK and ERK1/2. IL-17A mediated p38 MAPK and ERK1/2 activation is inhibited by MKP-1 overexpression, but prolonged by MKP-1 knockdown. IL-17A induced miR-101 expression via PI3K/Akt, and miR-101 inhibitor reversed MKP-1 down regulation. Importantly, MKP-1 knockdown, pharmacological inhibition of p38 MAPK and ERK1/2, or overexpression of dominant negative MEK1, each markedly attenuated IL-17A-mediated CF proliferation and migration. Similarly, IL-17F and IL-17A/F heterodimer that also signal via IL-17RA/IL-17RC, stimulated CF proliferation and migration. These results indicate that IL-17A stimulates CF proliferation and migration via Akt/miR-101/MKP-1-dependent p38 MAPK and ERK1/2 activation. These studies support a potential role for IL-17 in cardiac fibrosis and adverse myocardial remodeling.


Assuntos
Movimento Celular/genética , Fosfatase 1 de Especificidade Dupla/genética , Fibroblastos/metabolismo , Fibrose/metabolismo , Interleucina-17/metabolismo , MicroRNAs/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fosfatase 1 de Especificidade Dupla/antagonistas & inibidores , Fosfatase 1 de Especificidade Dupla/deficiência , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibrose/genética , Fibrose/patologia , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Interleucina-17/genética , Camundongos , MicroRNAs/genética , Miocárdio/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica , Transfecção , Remodelação Ventricular , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA