Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100563, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33745970

RESUMO

Hematopoietic stem cells (HSCs) and their progeny sustain lifetime hematopoiesis. Aging alters HSC function, number, and composition and increases risk of hematological malignancies, but how these changes occur in HSCs remains unclear. Signaling via p38 mitogen-activated kinase (p38MAPK) has been proposed as a candidate mechanism underlying induction of HSC aging. Here, using genetic models of both chronological and premature aging, we describe a multimodal role for p38α, the major p38MAPK isozyme in hematopoiesis, in HSC aging. We report that p38α regulates differentiation bias and sustains transplantation capacity of HSCs in the early phase of chronological aging. However, p38α decreased HSC transplantation capacity in the late progression phase of chronological aging. Furthermore, codeletion of p38α in mice deficient in ataxia-telangiectasia mutated, a model of premature aging, exacerbated aging-related HSC phenotypes seen in ataxia-telangiectasia mutated single-mutant mice. Overall, these studies provide new insight into multiple functions of p38MAPK, which both promotes and suppresses HSC aging context dependently.


Assuntos
Envelhecimento/patologia , Diferenciação Celular , Senescência Celular , Células-Tronco Hematopoéticas/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Envelhecimento/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Proliferação de Células , Feminino , Hematopoese , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Espécies Reativas de Oxigênio/metabolismo
2.
J Am Soc Nephrol ; 28(3): 823-836, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27620989

RESUMO

An improved understanding of pathogenic pathways in AKI may identify novel therapeutic approaches. Previously, we conducted unbiased liquid chromatography-tandem mass spectrometry-based protein expression profiling of the renal proteome in mice with acute folate nephropathy. Here, analysis of the dataset identified enrichment of pathways involving NFκB in the kidney cortex, and a targeted data mining approach identified components of the noncanonical NFκB pathway, including the upstream kinase mitogen-activated protein kinase kinase kinase 14 (MAP3K14), the NFκB DNA binding heterodimer RelB/NFκB2, and proteins involved in NFκB2 p100 ubiquitination and proteasomal processing to p52, as upregulated. Immunohistochemistry localized MAP3K14 expression to tubular cells in acute folate nephropathy and human AKI. In vivo, kidney expression levels of NFκB2 p100 and p52 increased rapidly after folic acid injection, as did DNA binding of RelB and NFκB2, detected in nuclei isolated from the kidneys. Compared with wild-type mice, MAP3K14 activity-deficient aly/aly (MAP3K14aly/aly) mice had less kidney dysfunction, inflammation, and apoptosis in acute folate nephropathy and less kidney dysfunction and a lower mortality rate in cisplatin-induced AKI. The exchange of bone marrow between wild-type and MAP3K14aly/aly mice did not affect the survival rate of either group after folic acid injection. In cultured tubular cells, MAP3K14 small interfering RNA targeting decreased inflammation and cell death. Additionally, cell culture and in vivo studies identified the chemokines MCP-1, RANTES, and CXCL10 as MAP3K14 targets in tubular cells. In conclusion, MAP3K14 promotes kidney injury through promotion of inflammation and cell death and is a promising novel therapeutic target.


Assuntos
Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/etiologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL
3.
J Neurochem ; 141(1): 37-47, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27973735

RESUMO

Myelination in the peripheral nervous system (PNS) is controlled by both positive and negative regulators within Schwann cells to ensure timely onset and correct myelin thickness for saltatory conduction by neurons. Transcription factors such as Sox10, octamer-binding transcription factor 6 (Oct6) and Krox20 form a positive regulatory network, whereas negative regulators such as cJun and Sox2 oppose myelination in Schwann cells. The role of the p38 MAPK pathway has been studied in PNS myelination, but its precise function remains unclear, with both positive and negative effects of p38 activity reported upon both myelination and processes of nerve repair. To clarify the role of p38 MAPK in the PNS, we have analysed mice with a Schwann cell-specific ablation of the major p38 isoform, p38alpha. In line with previous findings of an inhibitory role for p38 MAPK, we observe acceleration of post-natal myelination in p38alpha null nerves, a delay in myelin down-regulation following injury, together with a small increase in levels of re-myelination following injury. Finally we explored roles for p38alpha in controlling axonal regeneration and functional repair following PNS injury and observe that loss of p38alpha function in Schwann cells does not appear to affect these processes as previously reported. These studies therefore provide further proof for a role of p38 MAPK signalling in the control of myelination by Schwann cells in the PNS, but do not show an apparent role for signalling by this MAP kinase in Schwann cells controlling other elements of Wallerian degeneration and functional repair following injury. Cover Image for this issue: doi: 10.1111/jnc.13793.


Assuntos
Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Fibras Nervosas Mielinizadas/enzimologia , Traumatismos dos Nervos Periféricos/enzimologia , Nervos Periféricos/enzimologia , Recuperação de Função Fisiológica/fisiologia , Células de Schwann/enzimologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Masculino , Camundongos , Fibras Nervosas Mielinizadas/patologia , Traumatismos dos Nervos Periféricos/patologia , Nervos Periféricos/patologia , Ratos , Células de Schwann/patologia
4.
J Biol Chem ; 290(7): 4383-97, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548290

RESUMO

p38 MAPKs regulate migration and invasion. However, the mechanisms involved are only partially known. We had previously identified fibulin 3, which plays a role in migration, invasion, and tumorigenesis, as a gene regulated by p38α. We have characterized in detail how p38 MAPK regulates fibulin 3 expression and its role. We describe here for the first time that p38α, p38γ, and p38δ down-regulate fibulin 3 expression. p38α has a stronger effect, and it does so through hypermethylation of CpG sites in the regulatory sequences of the gene. This would be mediated by the DNA methylase, DNMT3A, which is down-regulated in cells lacking p38α, but once re-introduced represses Fibulin 3 expression. p38α through HuR stabilizes dnmt3a mRNA leading to an increase in DNMT3A protein levels. Moreover, by knocking-down fibulin 3, we have found that Fibulin 3 inhibits migration and invasion in MEFs by mechanisms involving p38α/ß inhibition. Hence, p38α pro-migratory/invasive effect might be, at least in part, mediated by fibulin 3 down-regulation in MEFs. In contrast, in HCT116 cells, Fibulin 3 promotes migration and invasion through a mechanism dependent on p38α and/or p38ß activation. Furthermore, Fibulin 3 promotes in vitro and in vivo tumor growth of HCT116 cells through a mechanism dependent on p38α, which surprisingly acts as a potent inducer of tumor growth. At the same time, p38α limits fibulin 3 expression, which might represent a negative feed-back loop.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Metilação de DNA , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Células Cultivadas , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação para Baixo , Embrião de Mamíferos/citologia , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Nat Genet ; 39(6): 750-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468755

RESUMO

Stem cell function is central for the maintenance of normal tissue homeostasis. Here we show that deletion of p38alpha mitogen-activated protein (MAP) kinase in adult mice results in increased proliferation and defective differentiation of lung stem and progenitor cells both in vivo and in vitro. We found that p38alpha positively regulates factors such as CCAAT/enhancer-binding protein that are required for lung cell differentiation. In addition, p38alpha controls self-renewal of the lung stem and progenitor cell population by inhibiting proliferation-inducing signals, most notably epidermal growth factor receptor. As a consequence, the inactivation of p38alpha leads to an immature and hyperproliferative lung epithelium that is highly sensitized to K-Ras(G12V)-induced tumorigenesis. Our results indicate that by coordinating proliferation and differentiation signals in lung stem and progenitor cells, p38alpha has a key role in the regulation of lung cell renewal and tumorigenesis.


Assuntos
Diferenciação Celular , Proliferação de Células , Pulmão/citologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Células-Tronco/citologia , Animais , Células Cultivadas , Feminino , Genes ras/fisiologia , Técnicas Imunoenzimáticas , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteínas Proto-Oncogênicas c-jun/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
6.
Nat Genet ; 39(6): 741-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468757

RESUMO

The mitogen-activated protein kinase (MAPK) p38alpha controls inflammatory responses and cell proliferation. Using mice carrying conditional Mapk14 (also known as p38alpha) alleles, we investigated its function in postnatal development and tumorigenesis. When we specifically deleted Mapk14 in the mouse embryo, fetuses developed to term but died shortly after birth, probably owing to lung dysfunction. Fetal hematopoietic cells and embryonic fibroblasts deficient in p38alpha showed increased proliferation resulting from sustained activation of the c-Jun N-terminal kinase (JNK)-c-Jun pathway. Notably, in chemical-induced liver cancer development, mice with liver-specific deletion of Mapk14 showed enhanced hepatocyte proliferation and tumor development that correlated with upregulation of the JNK-c-Jun pathway. Furthermore, inactivation of JNK or c-Jun suppressed the increased proliferation of Mapk14-deficient hepatocytes and tumor cells. These results demonstrate a new mechanism whereby p38alpha negatively regulates cell proliferation by antagonizing the JNK-c-Jun pathway in multiple cell types and in liver cancer development.


Assuntos
Proliferação de Células , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Fígado/embriologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Eritrócitos/citologia , Eritrócitos/metabolismo , Eritrócitos/patologia , Feminino , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fígado/citologia , Fígado/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-jun/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-jun/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
7.
J Immunol ; 190(4): 1519-27, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23315073

RESUMO

The MAPK p38α senses environmental stressors and orchestrates inflammatory and immunomodulatory reactions. However, the molecular mechanism how p38α controls immunomodulatory responses in myeloid cells remains elusive. We found that in monocytes and macrophages, p38α activated the mechanistic target of rapamycin (mTOR) pathway in vitro and in vivo. p38α signaling in myeloid immune cells promoted IL-10 but inhibited IL-12 expression via mTOR and blocked the differentiation of proinflammatory CD4(+) Th1 cells. Cellular stress induced p38α-mediated mTOR activation that was independent of PI3K but dependent on the MAPK-activated protein kinase 2 and on the inhibition of tuberous sclerosis 1 and 2, a negative regulatory complex of mTOR signaling. Remarkably, p38α and PI3K concurrently modulated mTOR to balance IL-12 and IL-10 expression. Our data link p38α to mTOR signaling in myeloid immune cells that is decisive for tuning the immune response in dependence on the environmental milieu.


Assuntos
Exposição Ambiental , Imunidade Inata , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Exposição Ambiental/efeitos adversos , Humanos , Imunidade Inata/genética , Interleucina-10/biossíntese , Subunidade p40 da Interleucina-12/biossíntese , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/genética , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
8.
Cancer Cell ; 11(2): 101-3, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17292820

RESUMO

It is established that p38 MAPK can negatively regulate tumorigenesis, but the mechanism is incompletely understood. A new study in this issue of Cancer Cell shows that p38 MAP kinase plays a selective role in tumor initiation mediated by oxidative stress.


Assuntos
Transformação Celular Neoplásica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Neoplasias/enzimologia , Estresse Oxidativo , Animais , Apoptose , Humanos , Proteína Quinase 14 Ativada por Mitógeno/genética , Neoplasias/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
9.
Cancer Cell ; 11(2): 191-205, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17292829

RESUMO

p38alpha is a stress-activated protein kinase that negatively regulates malignant transformation induced by oncogenic H-Ras, although the mechanisms involved are not fully understood. Here, we show that p38alpha is not a general inhibitor of oncogenic signaling, but that it specifically modulates transformation induced by oncogenes that produce reactive oxygen species (ROS). This inhibitory effect is due to the ROS-induced activation of p38alpha early in the process of transformation, which induces apoptosis and prevents the accumulation of ROS and their carcinogenic effects. Accordingly, highly tumorigenic cancer cell lines have developed a mechanism to uncouple p38alpha activation from ROS production. Our results indicate that oxidative stress sensing plays a key role in the inhibition of tumor initiation by p38alpha.


Assuntos
Transformação Celular Neoplásica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Neoplasias/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Células Cultivadas , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Genes ras/genética , Glutationa Transferase/metabolismo , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , NADPH Oxidases/metabolismo , Neoplasias/patologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
Lasers Surg Med ; 47(9): 765-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26414998

RESUMO

BACKGROUND AND OBJECTIVES: As the population ages, osteometabolic diseases and osteoporotic fractures emerge, resulting in substantial healthcare resource utilization and impaired quality of life. Many types of mechanical stimulation have the potential of being recognized by bone cells after a mechanical sign is transformed into a biological one (a process called mechanotransduction). The therapeutic ultrasound (TU) is one of several resources capable of promoting bone cell mechanical stimulation. Therefore, the main purpose of present study was to evaluate the effect of TU on the proliferation of pre-osteoblasts using in vitro bioassays. STUDY DESIGN/MATERIALS AND METHODS: We used MC3T3-E1 pre-osteoblast lineage cells kept in Alpha medium. Cells were treated using pulsed mode therapeutic ultrasound, with frequency of 1 MHz, intensity of 0.2 W/cm(2) (SATA), duty cycle of 20%, for 30 minutes. Nifedipine and rapamycin were used to further investigate the role of L-type Ca(2+) channels and mTOR pathway. Intracellular calcium, TGF-ß1, magnesium, and the mRNA levels of osteopontin, osteonectin, NF-κB1, p38α were evaluated. RESULTS: The results show that TU stimulates the growth of MC3T3-E1 cells and decreases the supernatant calcium and magnesium content. Also, it increases intracellular calcium, activates NF-κB1 and mTOR complex via p38α. Moreover, TU promoted a decrease in the TGF-ß1 synthesis, which is a cell growth inhibitor. CONCLUSIONS: Therapeutic ultrasound, with frequency of 1 MHz, intensity of 0.2 W/cm(2) (SATA) and pulsed mode, for 30 minutes, was able to increase the proliferation of preosteoblast-like bone cells. This effect was mediated by a calcium influx, with a consequent activation of the mTOR pathway, through increased NF-κB1 and p38α.


Assuntos
Proliferação de Células/efeitos da radiação , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , NF-kappa B/fisiologia , Osteoblastos/efeitos da radiação , Serina-Treonina Quinases TOR/fisiologia , Terapia por Ultrassom , Células 3T3 , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Camundongos , Osteoblastos/metabolismo , Osteoblastos/patologia
11.
Blood ; 119(26): 6255-8, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22555972

RESUMO

We report on a forward RNAi screen in primary human hematopoietic stem and progenitor cells, using pooled lentiviral shRNA libraries deconvoluted by next generation sequencing. We identify MAPK14/p38α as a modulator of ex vivo stem cell proliferation and show that pharmacologic inhibition of p38 dramatically enhances the stem cell activity of cultured umbilical cord blood derived hematopoietic cells. p38 inhibitors should thus be considered in strategies aiming at expanding stem cells for clinical benefit.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Descoberta de Drogas , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Proteína Quinase 14 Ativada por Mitógeno/genética , Terapia de Alvo Molecular , Interferência de RNA/fisiologia , RNA Interferente Pequeno/isolamento & purificação
12.
J Neurosci ; 32(36): 12325-36, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956823

RESUMO

Repeated stress releases dynorphins and causes subsequent activation of κ-opioid receptors (KORs) in limbic brain regions. The serotonergic dorsal raphe nucleus (DRN) has previously been found to be an important site of action for the dysphoric effects of dynorphin-κ-opioid receptor system activation during stress-evoked behaviors, and KOR-induced activation of p38α mitogen-activated protein kinase (MAPK) in serotonergic neurons was found to be a critical mediator of the aversive properties of stress. Yet, how dynorphins and KORs functionally regulate the excitability of serotonergic DRN neurons both in adaptive and pathological stress states is poorly understood. Here we report that acute KOR activation by the selective agonist U69,593 [(+)-(5α,7α,8ß)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]benzeneacetamide] inhibits serotonergic neuronal excitability within the DRN through both presynaptic inhibition of excitatory synaptic transmission and postsynaptic activation of G-protein-gated inwardly rectifying potassium channels (GIRKs) electrophysiologically recorded in brain slices. C57BL/6 mice subjected to repeated swim, stress sessions had significantly reduced KOR-mediated GIRK currents recorded in serotonergic neurons in DRN postsynaptically, without significantly affecting presynaptic KOR-mediated regulation of excitatory transmission. This effect was blocked by genetic excision of p38α MAPK selectively from serotonergic neurons. An increase in phospho-immunoreactivity suggests that this functional dysregulation may be a consequence of tyrosine phosphorylation of GIRK (K(IR)3.1) channels. These data elucidate a mechanism for stress-induced dysregulation of the excitability of neurons in the DRN and identify a functional target of stress-induced p38α MAPK activation that may underlie some of the negative effects of pathological stress exposure.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Núcleos da Rafe/enzimologia , Transdução de Sinais/fisiologia , Estresse Psicológico/enzimologia , Animais , Benzenoacetamidas/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fosforilação , Pirrolidinas/farmacologia , Núcleos da Rafe/efeitos dos fármacos , Tempo de Reação/fisiologia , Serotonina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Tirosina/metabolismo
13.
Am J Physiol Renal Physiol ; 304(7): F908-17, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23324178

RESUMO

High NaCl activates the transcription factor nuclear factor of activated T cells 5 (NFAT5), leading to increased transcription of osmoprotective target genes. Kinases PKA, PI3K, AKT1, and p38α were known to contribute to the high NaCl-induced increase of NFAT5 activity. We now identify another kinase, GSK-3ß. siRNA-mediated knock-down of GSK-3ß increases NFAT5 transcriptional and transactivating activities without affecting high NaCl-induced nuclear localization of NFAT5 or NFAT5 protein expression. High NaCl increases phosphorylation of GSK-3ß-S9, which inhibits GSK-3ß. In GSK-3ß-null mouse embryonic fibroblasts transfection of GSK-3ß, in which serine 9 is mutated to alanine, so that it cannot be inhibited by phosphorylation at that site, inhibits high NaCl-induced NFAT5 transcriptional activity more than transfection of wild-type GSK-3ß. High NaCl-induced phosphorylation of GSK-3ß-S9 depends on PKA, PI3K, and AKT, but not p38α. Overexpression of PKA catalytic subunit α or of catalytically active AKT1 reduces inhibition of NFAT5 by GSK-3ß, but overexpression of p38α together with its catalytically active upstream kinase, MKK6, does not. Thus, GSK-3ß normally inhibits NFAT5 by suppressing its transactivating activity. When activated by high NaCl, PKA, PI3K, and AKT1, but not p38α, increase phosphorylation of GSK-3ß-S9, which reduces the inhibitory effect of GSK-3ß on NFAT5, and thus contributes to activation of NFAT5.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cloreto de Sódio/administração & dosagem
14.
J Vasc Res ; 50(2): 145-56, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23258237

RESUMO

Tumor necrosis factor (TNF)-α can alter tissue repair functions in a variety of cells including endothelial cells. However, the mechanism by which TNF-α mediates these functional changes has not fully been studied. We investigated the role of mitogen-activated protein kinases (MAPKs) on mediating the regulatory effect of TNF-α on the tissue repair functions of human pulmonary artery endothelial cells (HPAECs). TNF-α protected HPAECs from undergoing apoptosis induced by serum and growth factor deprivation, augmented collagen gel contraction, and stimulated wound closure. TNF-α activated c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases 1 and 2 (ERK1/2), and p38. Inhibitors of JNK (SP600125, 5 µM) or ERK1/2 (PD98059, 5 µM) significantly inhibited TNF-α-stimulated cell survival, contraction of collagen gels, and wound closure. In contrast, the p38 inhibitor SB203580 (5 µM) further amplified all of the TNF-α effects on HPAECs. TNF-α specifically activated p38α but not other p38 isoforms and suppression of p38α by an siRNA resulted in further amplification of the TNF-α effect. These results suggest that TNF-α stimulates tissue repair functions of HPAECs, and this may be mediated, at least in part, positively via JNK and ERK1/2, and negatively through p38α. MAPKs may play a role in endothelial cell-mediated tissue repair, especially in an inflammatory milieu where TNF-α is present.


Assuntos
Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Artéria Pulmonar/citologia , Fator de Necrose Tumoral alfa/farmacologia , Cicatrização/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Células Cultivadas/fisiologia , Colágeno , Meios de Cultura Livres de Soro/farmacologia , Células Endoteliais/enzimologia , Células Endoteliais/fisiologia , Ativação Enzimática/efeitos dos fármacos , Géis , Humanos , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Vasculite/enzimologia , Vasculite/fisiopatologia , Cicatrização/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
15.
PLoS Pathog ; 6(6): e1000934, 2010 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20532209

RESUMO

Intestinal epithelial cells (IECs) compose the first barrier against microorganisms in the gastrointestinal tract. Although the NF-kappaB pathway in IECs was recently shown to be essential for epithelial integrity and intestinal immune homeostasis, the roles of other inflammatory signaling pathways in immune responses in IECs are still largely unknown. Here we show that p38alpha in IECs is critical for chemokine expression, subsequent immune cell recruitment into the intestinal mucosa, and clearance of the infected pathogen. Mice with p38alpha deletion in IECs suffer from a sustained bacterial burden after inoculation with Citrobacter rodentium. These animals are normal in epithelial integrity and immune cell function, but fail to recruit CD4(+) T cells into colonic mucosal lesions. The expression of chemokines in IECs is impaired, which appears to be responsible for the impaired T cell recruitment. Thus, p38alpha in IECs contributes to the host immune responses against enteric bacteria by the recruitment of immune cells.


Assuntos
Colo/metabolismo , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Linfócitos T/imunologia , Animais , Biomarcadores/metabolismo , Western Blotting , Quimiocinas/metabolismo , Citrobacter rodentium/imunologia , Colo/citologia , Colo/microbiologia , Ensaio de Unidades Formadoras de Colônias , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Integrases/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL/microbiologia , Camundongos Knockout/microbiologia , NF-kappa B , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Linfócitos T/metabolismo , Linfócitos T/microbiologia
16.
Reproduction ; 144(5): 535-46, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22949725

RESUMO

To define signaling pathways that drive FSH- and epidermal growth factor (EGF)-like peptide-induced cumulus expansion and oocyte meiotic resumption, in vitro cultured pig cumulus-oocyte complexes were treated with specific protein kinase inhibitors. We found that FSH-induced maturation of oocytes was blocked in germinal vesicle (GV) stage by protein kinase A (PKA), MAPK14, MAPK3/1, and EGF receptor (EGFR) tyrosine kinase inhibitors (H89, SB203580, U0126, and AG1478 respectively) whereas phosphoinositide-3-kinase/v-akt murine thymoma viral oncogene homolog (PI3K/AKT) inhibitor (LY294002) blocked maturation of oocytes in metaphase I (MI). Amphiregulin (AREG)-induced maturation of oocytes was efficiently blocked in GV by U0126, AG1478, and low concentrations of LY294002; H89, SB203580, and high concentrations of LY294002 allowed the oocytes to undergo breakdown of GV and blocked maturation in MI. Both FSH- and AREG-induced cumulus expansion was incompletely inhibited by H89 and completely inhibited by SB203580, U0126, AG1478, and LY294002. The inhibitors partially or completely inhibited expression of expansion-related genes (HAS2, PTGS2, and TNFAIP6) with two exceptions: H89 inhibited only TNFAIP6 expression and LY294002 increased expression of PTGS2. The results of this study are consistent with the idea that PKA and MAPK14 pathways are essential for FSH-induced transactivation of the EGFR, and synthesis of EGF-like peptides in cumulus cells and MAPK3/1 is involved in regulation of transcriptional and posttranscriptional events in cumulus cells required for meiotic resumption and cumulus expansion. PI3K/AKT signaling is important for regulation of cumulus expansion, AREG-induced meiotic resumption, and oocyte MI/MII transition. The present data also indicate the existence of an FSH-activated and PKA-independent pathway involved in regulation of HAS2 and PTGS2 expression in cumulus cells.


Assuntos
Células do Cúmulo/fisiologia , Hormônio Foliculoestimulante/farmacologia , Glicoproteínas/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Meiose/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sus scrofa/fisiologia , Anfirregulina , Animais , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ciclo-Oxigenase 2/genética , Receptores ErbB/fisiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Glucuronosiltransferase/genética , Meiose/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
Exp Eye Res ; 100: 101-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22564969

RESUMO

The goal of this study was to define the role of p38alpha MAP kinase in VEGF-induced vascular permeability increase. Activation of p38 is correlated with increased permeability in endothelial cells treated with VEGF or high glucose and in retinas of diabetic animal models. We have shown previously that p38 inhibitors preserve endothelial barrier function and block VEGF-induced GSK/beta-catenin signaling. Here, we present data demonstrating that adenoviral vector delivery of a dominant negative p38alpha mutant blocks this signaling pathway and preserves barrier function. This p38alpha mutant was altered on its ATP-binding site, which eliminates its kinase activity. Bovine retinal endothelial (BRE) cells were transduced with recombinant adenovirus containing the p38alpha mutants or empty vector. Successful transduction was confirmed by expression of GFP and p38 increase. Blockade of p38 activity by p38alpha mutant was demonstrated by inhibition of VEGF-induced phosphorylation of a p38 target, MAP kinase activated protein kinase 2 (MK-2). The mutant also prevented VEGF-induced GSK phosphorylation and beta-catenin cytosolic accumulation and nuclear translocation as shown by cell fractionation and Western blotting. Quantitative real-time PCR demonstrated that this mutant inhibited VEGF-induced uPAR gene expression. Importantly, this same mutant also strongly abrogated VEGF-induced endothelial barrier breakdown as determined by measuring transcellular electrical resistance and tracer flux through endothelial cell monolayer. This study indicates a critical role of p38alpha in VEGF-induced permeability and offers a new strategy for developing potent and specific therapies for treatment of retinal diseases associated with vascular barrier dysfunction.


Assuntos
Células Endoteliais/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , beta Catenina/antagonistas & inibidores , Adenoviridae/genética , Animais , Permeabilidade Capilar/fisiologia , Bovinos , Impedância Elétrica , Células Endoteliais/citologia , Regulação Enzimológica da Expressão Gênica , Vetores Genéticos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Vasos Retinianos/citologia , Transdução de Sinais/efeitos dos fármacos , beta Catenina/metabolismo
18.
Biochem J ; 434(3): 549-58, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21226672

RESUMO

p38α MAPK (mitogen-activated protein kinase) plays an important tumour suppressor role, which is mediated by both its negative effect on cell proliferation and its pro-apoptotic activity. Surprisingly, most tumour suppressor mechanisms co-ordinated by p38α have been reported to occur at the post-translational level. This contrasts with the important role of p38α in the regulation of transcription and the profound changes in gene expression that normally occur during tumorigenesis. We have analysed whole-genome expression profiles of Ras-transformed wild-type and p38α-deficient cells and have identified 202 genes that are potentially regulated by p38α in transformed cells. Expression analysis has confirmed the regulation of these genes by p38α in tumours, and functional validation has identified several of them as probable mediators of the tumour suppressor effect of p38α on Ras-induced transformation. Interestingly, approx. 10% of the genes that are negatively regulated by p38α in transformed cells contribute to EGF (epidermal growth factor) receptor signalling. Our results suggest that inhibition of EGF receptor signalling by transcriptional targets of p38α is an important function of this signalling pathway in the context of tumour suppression.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Transcrição Gênica , Animais , Proliferação de Células , Transformação Celular Neoplásica/patologia , Células Cultivadas , Receptores ErbB/fisiologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Genes ras , Humanos , Camundongos , Camundongos Nus , Proteína Quinase 14 Ativada por Mitógeno/genética , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
19.
Artigo em Inglês | MEDLINE | ID: mdl-22575607

RESUMO

The Atlantic killifish (Fundulus heteroclitus) is an environmental sentinel organism used extensively for studies of environmental toxicants and osmoregulation. Previous research in our laboratory has shown that acute acclimation to seawater is mediated by an increase in SGK1. SGK1 promotes the trafficking of CFTR chloride channels from intracellular vesicles to the plasma membrane of the gill within the first hour in seawater resulting in increased chloride secretion. Although we have shown that the increase in gill SGK1 does not require activation of the glucocorticoid receptor, the mechanisms that mediate the rise SGK1 during acute acclimation is unknown. To test the hypothesis that mitogen activated protein kinase (MAPK14) is responsible for the rise in SGK1 we identified the coding sequence of killifish MAPK14-1 and designed a translational blocking vivo-morpholino targeting MAPK14-1. Injection of the MAPK14-1 vivo-morpholino resulted in a 30% reduction of MAPK14-1 and a 45% reduction in phosphorylated-MAPK14-1 protein in the gill of killifish transitioned from freshwater to seawater. Knock down of phosphorlyated-MAPK14-1 completely blocked the rise in SGK1 mRNA and protein in the killifish gill, providing the first direct and in vivo evidence that MAPK14-1 is necessary for acute seawater acclimation.


Assuntos
Aclimatação , Proteínas de Peixes/metabolismo , Fundulidae/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Feminino , Proteínas de Peixes/genética , Fundulidae/metabolismo , Técnicas de Silenciamento de Genes , Brânquias/enzimologia , Masculino , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Morfolinos/genética , Proteínas Serina-Treonina Quinases/genética , Tolerância ao Sal , Água do Mar , Homologia de Sequência de Aminoácidos , ATPase Trocadora de Sódio-Potássio/metabolismo
20.
Int Endod J ; 45(3): 224-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21992459

RESUMO

AIM: To investigate whether the p38α mitogen-activated protein kinases (MAPK) is involved in bone morphogenetic protein (BMP)-2-induced odontoblastic differentiation of human dental pulp cells (HDPCs). METHODOLOGY: Recombinant retrovirus encoding shRNA against p38α MAPK was constructed to investigate the role of p38α MAPK on BMP-2-induced odontoblastic differentiation of HDPCs. HDPCs were transfected with retrovirus expressing sh-p38α. Activation of p38α MAPK was detected by Western blot. The effects of p38α MAPK on BMP-2-induced odontoblastic differentiation of HDPCs were measured by alkaline phosphatase (ALP) activity, and the expression of odontoblastic markers was identified by quantitative real-time polymerase chain reaction analysis. The effect of SD-282, a p38a-specific inhibitor, on BMP-2-induced odontoblastic differentiation was also investigated. RESULTS: BMP-2 dose- and time-dependently upregulated phosphorylation of p38α of HDPCs. Compared with BMP-2-treatment group, gene knock-down of p38α MAPK significantly inhibited ALP activity and the formation of mineralized nodules in HDPCs. Moreover, suppression of p38α MAPK repressed the odontoblastic differentiation in HDPCs. Consistently, inhibition of p38α by SD-282 also decreased odontoblastic differentiation. CONCLUSIONS: p38α MAPK is involved in BMP-2-induced odontoblastic differentiation of HDPCs.


Assuntos
Proteína Morfogenética Óssea 2/fisiologia , Polpa Dentária/citologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Odontoblastos/fisiologia , Adulto , Fosfatase Alcalina/análise , Western Blotting , Proteína Morfogenética Óssea 2/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/enzimologia , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Indóis/farmacologia , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/genética , Odontoblastos/efeitos dos fármacos , Odontoblastos/enzimologia , Fosforilação , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA