Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Mol Life Sci ; 79(4): 198, 2022 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-35313355

RESUMO

The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH2-terminal mitochondrial targeting sequence. Loss of Dusp26 has a significant impact on mitochondrial function that is associated with increased levels of reactive oxygen species (ROS), reduction in ATP generation, reduction in mitochondria motility and release of mitochondrial HtrA2 protease into the cytoplasm. The mitochondrial dysregulation in dusp26-deficient neuroblastoma cells leads to the inhibition of cell proliferation and cell death. In vivo, Dusp26 is highly expressed in neurons in different brain regions, including cortex and midbrain (MB). Ablation of Dusp26 in mouse model leads to dopaminergic (DA) neuronal cell loss in the substantia nigra par compacta (SNpc), inflammatory response in MB and striatum, and phenotypes that are normally associated with Neurodegenerative diseases. Consistent with the data from our mouse model, Dusp26 expressing cells are significantly reduced in the SNpc of Parkinson's Disease patients. The underlying mechanism of DA neuronal death is that loss of Dusp26 in neurons increases mitochondrial ROS and concurrent activation of MAPK/p38 signaling pathway and inflammatory response. Our results suggest that regulation of mitochondrial-associated protein phosphorylation is essential for the maintenance of mitochondrial homeostasis and dysregulation of this process may contribute to the initiation and development of neurodegenerative diseases.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Fosfatases de Especificidade Dupla/fisiologia , Mitocôndrias/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Animais , Morte Celular/genética , Respiração Celular/genética , Células Cultivadas , Citoproteção/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mitocôndrias/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
2.
Biosci Biotechnol Biochem ; 85(8): 1839-1845, 2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34143206

RESUMO

DUSP4 is considered as an oncogenic gene. However, the effect of DUSP4 on the carcinogenesis of clear cell Renal cell carcinoma (CCRCC) is still unclear. In this study, DUSP4 mRNA levels were significantly increased in CCRCC tissues and cell lines. Furthermore, DUSP4 overexpression promotes the proliferation, migration, and tumorigenicity of CCRCC cells while DUSP4 silencing showed the opposite effects. Importantly, both autophagic activity (LC3 conversion rate and LC3 puncta formation) and total death level promoted by DUSP4 silencing were reversed by treatment with 3-MA in CCRCC cells. Moreover, the proliferation and migration of CCRCC cells inhibited by DUSP4 silencing were also recovered by suppression of autophagy with 3-MA. In conclusion, DUSP4 serves as an oncogenic gene in CCRCC carcinogenesis due to its inhibitory effect on autophagic death, indicating the potential value of DUSP4 in the diagnosis and treatment of CCRCC.


Assuntos
Autofagia/genética , Carcinogênese , Carcinoma de Células Renais/patologia , Morte Celular/genética , Fosfatases de Especificidade Dupla/genética , Neoplasias Renais/patologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Idoso , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Fosfatases de Especificidade Dupla/fisiologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Renais/genética , Pessoa de Meia-Idade , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia
3.
FASEB J ; 33(11): 12941-12959, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31480854

RESUMO

Gliomas are the most common primary brain tumors. Their highly invasive character and the heterogeneity of active oncogenic pathways within single tumors complicate the development of curative therapies and cause poor patient prognosis. Glioma cells express the intermediate filament protein glial fibrillary acidic protein (GFAP), and the level of its alternative splice variant GFAP-δ, relative to its canonical splice variant GFAP-α, is higher in grade IV compared with lower-grade and lower malignant glioma. In this study we show that a high GFAP-δ/α ratio induces the expression of the dual-specificity phosphatase 4 (DUSP4) in focal adhesions. By focusing on pathways up- and downstream of DUSP4 that are involved in the cell-extracellular matrix interaction, we show that a high GFAP-δ/α ratio equips glioma cells to better invade the brain. This study supports the hypothesis that glioma cells with a high GFAP-δ/α ratio are highly invasive and more malignant cells, thus making GFAP alternative splicing a potential therapeutic target.-Van Bodegraven, E. J., van Asperen, J. V., Sluijs, J. A., van Deursen, C. B. J., van Strien, M. E., Stassen, O. M. J. A., Robe, P. A. J., Hol, E. M. GFAP alternative splicing regulates glioma cell-ECM interaction in a DUSP4-dependent manner.


Assuntos
Processamento Alternativo , Neoplasias Encefálicas/patologia , Fosfatases de Especificidade Dupla/fisiologia , Matriz Extracelular/patologia , Proteína Glial Fibrilar Ácida/genética , Glioma/patologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Neoplasias Encefálicas/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Fosfatases de Especificidade Dupla/genética , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Glioma/metabolismo , Humanos , Laminina/metabolismo , MAP Quinase Quinase 4/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosforilação
4.
Exp Cell Res ; 382(1): 111467, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31202710

RESUMO

Hyperglycemia and hyperlipidemia (glycolipotoxicity)-triggered islet ß-cell dysfunction is known to drive the progression of obesity-related type 2 diabetes, however the underlying mechanisms have not been clearly elucidated. The current study aimed to investigate the role of mitogen-activated protein kinase phosphatase 5 (MKP-5) in islet cells under glucolipotoxic conditions. Using gene overexpression and knockdown approaches, we demonstrated that MKP-5 could alleviate glucolipotoxicity-induced apoptosis via the endoplasmic reticulum (ER) stress and mitochondrial apoptosis pathways owing to the altered regulation of caspase family members and ER stress-related molecules in MIN6 and primary islet cells. Overexpression of MKP-5 reversed the glucose and palmitic acid (GP)-induced impairment of insulin secretion as well as the abnormal decreases in the expression of islet functional genes, thereby maintaining the normal insulin secretory functionality, whereas the absence of MKP-5 aggravated islet cell dysfunction. In parallel, the production of ROS and increased inflammation-associated genes in response to GP were also reduced upon MKP-5 overexpression. Further, inhibition of JNK or P38 MAPK pathways resisted to glucolipotoxicity observed in MKP-5 knockdown MIN6 cells. These findings indicate that MKP-5 is an important mediator for glucolipotoxicity-induced islet cell dysfunction and apoptosis, with JNK and P38 as the critical downstream pathways.


Assuntos
Apoptose/fisiologia , Fosfatases de Especificidade Dupla/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Glucose/toxicidade , Ilhotas Pancreáticas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Palmitatos/toxicidade , Animais , Linhagem Celular Tumoral , Dieta Hiperlipídica/efeitos adversos , Fosfatases de Especificidade Dupla/genética , Técnicas de Silenciamento de Genes , Humanos , Insulina/metabolismo , Insulinoma/patologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Neoplasias Pancreáticas/patologia , Proteínas Recombinantes/metabolismo , Regulação para Cima
5.
FASEB J ; 33(6): 7331-7347, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30884976

RESUMO

Osteoblasts are versatile cells involved in multiple whole-body processes, including bone formation and immune response. Secretory amounts and patterns of osteoblast-derived proteins such as osteopontin (OPN) and osteocalcin (OCN) modulate osteoblast function. However, the regulatory mechanism of OPN and OCN expression remains unknown. Here, we demonstrate that p54/p46 c-jun N-terminal kinase (JNK) inhibition suppresses matrix mineralization and OCN expression but increases OPN expression in MC3T3-E1 cells and primary osteoblasts treated with differentiation inducers, including ascorbic acid, bone morphogenic protein-2, or fibroblast growth factor 2. Preinhibition of JNK before the onset of differentiation increased the number of osteoblasts that highly express OPN but not OCN (OPN-OBs), indicating that JNK affects OPN secretory phenotype at the early stage of osteogenic differentiation. Additionally, we identified JNK2 isoform as being critically involved in OPN-OB differentiation. Microarray analysis revealed that OPN-OBs express characteristic transcription factors, cell surface markers, and cytokines, including glycoprotein hormone α2 and endothelial cell-specific molecule 1. Moreover, we found that inhibitor of DNA binding 4 is an important regulator of OPN-OB differentiation and that dual-specificity phosphatase 16, a JNK-specific phosphatase, functions as an endogenous regulator of OPN-OB induction. OPN-OB phenotype was also observed following LPS from Porphyromonas gingivalis stimulation during osteogenic differentiation. Collectively, these results suggest that the JNK-Id4 signaling axis is crucial in the control of OPN and OCN expression during osteoblastic differentiation.-Kusuyama, J., Amir, M. S., Albertson, B. G., Bandow, K., Ohnishi, T., Nakamura, T., Noguchi, K., Shima, K., Semba, I., Matsuguchi, T. JNK inactivation suppresses osteogenic differentiation, but robustly induces osteopontin expression in osteoblasts through the induction of inhibitor of DNA binding 4 (Id4).


Assuntos
Proteínas Inibidoras de Diferenciação/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Osteopontina/biossíntese , Animais , Células Cultivadas , Fosfatases de Especificidade Dupla/deficiência , Fosfatases de Especificidade Dupla/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/deficiência , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Osteocalcina/biossíntese , Osteocalcina/genética , Osteogênese/efeitos dos fármacos , Osteopontina/genética , Isoformas de Proteínas/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia
6.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 124-143, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30401534

RESUMO

It is well established that a family of dual-specificity MAP kinase phosphatases (MKPs) play key roles in the regulated dephosphorylation and inactivation of MAP kinase isoforms in mammalian cells and tissues. MKPs provide a mechanism of spatiotemporal feedback control of these key signalling pathways, but can also mediate crosstalk between distinct MAP kinase cascades and facilitate interactions between MAP kinase pathways and other key signalling modules. As our knowledge of the regulation, substrate specificity and catalytic mechanisms of MKPs has matured, more recent work using genetic models has revealed key physiological functions for MKPs and also uncovered potentially important roles in regulating the pathophysiological outcome of signalling with relevance to human diseases. These include cancer, diabetes, inflammatory and neurodegenerative disorders. It is hoped that this understanding will reveal novel therapeutic targets and biomarkers for disease, thus contributing to more effective diagnosis and treatment for these debilitating and often fatal conditions.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Animais , Catálise , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Diabetes Mellitus/metabolismo , Fosfatases de Especificidade Dupla/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neuropatologia , Obesidade/metabolismo , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Proteínas Tirosina Fosfatases/fisiologia , Transdução de Sinais/fisiologia , Especificidade por Substrato/fisiologia
7.
Plant J ; 95(6): 937-946, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29775492

RESUMO

Grain size and weight are directly associated with grain yield in crops. However, the molecular mechanisms that set final grain size and weight remain largely unknown. Here, we characterize two large grain mutants, large grain8-1 (large8-1) and large grain8-2 (large8-2). LARGE8 encodes the mitogen-activated protein kinase phosphatase1 (OsMKP1). Loss of function mutations in OsMKP1 results in large grains, while overexpression of OsMKP1 leads to small grains. OsMKP1 determines grain size by restricting cell proliferation in grain hulls. OsMKP1 directly interacts with and deactivates the mitogen-activated protein kinase 6 (OsMAPK6). Taken together, we identify OsMKP1 as a crucial factor that influences grain size by deactivating OsMAPK6, indicating that the reversible phosphorylation of OsMAPK6 plays important roles in determining grain size in rice.


Assuntos
Grão Comestível/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Proliferação de Células , Grão Comestível/enzimologia , Grão Comestível/crescimento & desenvolvimento , Genes de Plantas/genética , Genes de Plantas/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Mutação , Oryza/enzimologia , Oryza/genética , Oryza/crescimento & desenvolvimento , Proteínas de Plantas/genética , Proteínas de Plantas/fisiologia
8.
Sci Rep ; 7(1): 5241, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701747

RESUMO

Expression of neuroendocrine-associated phosphatase (NEAP, also named as dual specificity phosphatase 26, [DUSP26]) is restricted to neuroendocrine tissues. We found that NEAP, but not its phosphatase-defective mutant, suppressed nerve growth factor (NGF) receptor TrkA and fibroblast growth factor receptor 1 (FGFR1) activation in PC12 cells upon NGF stimulation. Conversely, suppressing NEAP expression by RNA interference enhanced TrkA and FGFR1 phosphorylation. NEAP was capable of de-phosphorylating TrkA and FGFR1 directly in vitro. NEAP-orthologous gene existed in zebrafish. Morpholino (MO) suppression of NEAP in zebrafish resulted in hyper-phosphorylation of TrkA and FGFR1 as well as abnormal body postures and small eyes. Differentiation of retina in zebrafishes with NEAP MO treatment was severely defective, so were cranial motor neurons. Taken together, our data indicated that NEAP/DUSP26 have a critical role in regulating TrkA and FGFR1 signaling as well as proper development of retina and neuronal system in zebrafish.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Embrião não Mamífero/citologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Doença dos Neurônios Motores/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor trkA/antagonistas & inibidores , Doenças Retinianas/patologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Diferenciação Celular , Fosfatases de Especificidade Dupla/genética , Embrião não Mamífero/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Morfolinos/farmacologia , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/metabolismo , Células PC12 , Fosforilação , Ratos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
9.
Arterioscler Thromb Vasc Biol ; 34(9): 2023-32, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24990230

RESUMO

OBJECTIVE: Reactive oxygen species (ROS) act as signaling molecules during angiogenesis; however, the mechanisms used for such signaling events remain unclear. Stromal cell-derived factor-1α (SDF-1α) is one of the most potent angiogenic chemokines. Here, we examined the role of ROS in the regulation of SDF-1α-dependent angiogenesis. APPROACH AND RESULTS: Bovine aortic endothelial cells were treated with SDF-1α, and intracellular ROS generation was monitored. SDF-1α treatment induced bovine aortic endothelial cell migration and ROS generation, with the majority of ROS generated by bovine aortic endothelial cells at the leading edge of the migratory cells. Antioxidants and nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitors blocked SDF-1α-induced endothelial migration. Furthermore, knockdown of either NOX5 or p22phox (a requisite subunit for NOX1/2/4 activation) significantly impaired endothelial motility and tube formation, suggesting that multiple NOXs regulate SDF-1α-dependent angiogenesis. Our previous study demonstrated that c-Jun N-terminal kinase 3 activity is essential for SDF-1α-dependent angiogenesis. Here, we identified that NOX5 is the dominant NOX required for SDF-1α-induced c-Jun N-terminal kinase 3 activation and that NOX5 and MAP kinase phosphatase 7 (MKP7; the c-Jun N-terminal kinase 3 phosphatase) associate with one another but decrease this interaction on SDF-1α treatment. Furthermore, MKP7 activity was inhibited by SDF-1α, and this inhibition was relieved by NOX5 knockdown, indicating that NOX5 promotes c-Jun N-terminal kinase 3 activation by blocking MKP7 activity. CONCLUSIONS: We conclude that NOX is required for SDF-1α signaling and that intracellular redox balance is critical for SDF-1α-induced endothelial migration and angiogenesis.


Assuntos
Quimiocina CXCL12/fisiologia , Proteínas de Membrana/fisiologia , NADPH Oxidases/fisiologia , Neovascularização Fisiológica/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Aorta , Azóis/farmacologia , Bovinos , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Fosfatases de Especificidade Dupla/fisiologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Técnicas de Silenciamento de Genes , Hiperglicemia/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/fisiologia , Isoindóis , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteína Quinase 10 Ativada por Mitógeno/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Neovascularização Fisiológica/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Oxirredução , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
J Pharmacol Sci ; 122(2): 93-102, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23698110

RESUMO

AMP-activated protein kinase (AMPK) contributes to the acceleration of insulin signaling. However, the mechanism by which AMPK regulates insulin signaling remains unclear. Serine phosphorylation of insulin receptor substrate (IRS)-1 negatively regulates insulin signaling. Here we investigated the role of AMPK in serine phosphorylation of IRS-1 at 636/639 and 307, which is induced by tumor necrosis factor (TNF)-α in 3T3L1 adipocytes. We demonstrated that the AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) significantly inhibited the TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by suppression of extracellular signal-regulated kinase (ERK) phosphorylation but not c-Jun-NH2-terminal kinase (JNK) phosphorylation. In addition, AICAR stimulation resulted in enhanced interaction between ERK and MAP kinase phosphatase-4 (DUSP9/MKP-4) without affecting DUSP9/MPK4 mRNA synthesis. Moreover, intraperitoneal administration (0.25 g/kg) of AICAR to db/db mice improved blood glucose levels and inhibited the phosphorylation of ERK in adipose tissue. In conclusion, we propose a new mechanism in which AICAR suppresses TNF-α-induced serine phosphorylation of IRS-1 at 636/639 and 307 by enhancing the interaction between ERK and DUSP9/MKP-4. Taken together, these findings provide evidence that AMPK plays a crucial role in improving of type 2 diabetes.


Assuntos
Células 3T3-L1/metabolismo , Proteínas Quinases Ativadas por AMP/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Hipoglicemiantes/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Ribonucleotídeos/farmacologia , Serina/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Aminoimidazol Carboxamida/administração & dosagem , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/etiologia , Fosfatases de Especificidade Dupla/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipoglicemiantes/administração & dosagem , Injeções Intraperitoneais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fosforilação/efeitos dos fármacos , Ribonucleotídeos/administração & dosagem , Transdução de Sinais , Estimulação Química , Fator de Necrose Tumoral alfa/antagonistas & inibidores
11.
J Biol Chem ; 288(2): 819-25, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23229544

RESUMO

The transcription factor NF-κB is critically involved in the inflammatory response triggered by the proinflammatory cytokines TNF and IL-1. Various studies have demonstrated that activation of TAK1 (TGF-ß-activated kinase 1) is an essential step in TNF- and IL-1-induced NF-κB activation pathways. In this study, we identified a member of the dual-specificity phosphatase family, DUSP14, as a negative regulator of TNF- and IL-1-triggered NF-κB activation by expression screens. We found that DUSP14 interacted with TAK1 and that this interaction was enhanced by TNF or IL-1 stimulation. Overexpression of DUSP14 dephosphorylated TAK1 at Thr-187, a residue in the activation loop critically involved in TAK1 activation. Knockdown of DUSP14 increased basal as well as TNF- and IL-1-induced TAK1 phosphorylation at Thr-187. Overexpression of DUSP14, but not its phosphatase-deficient mutant, inhibited TNF- and IL-1-induced as well as TAK1-mediated NF-κB activation, whereas knockdown of DUSP14 had opposite effects. These findings suggest that DUSP14 negatively regulates TNF- or IL-1-induced NF-κB activation by dephosphorylating TAK1 at Thr-187. Our study reveals a new post-translational regulatory mechanism of NF-κB activation triggered by the proinflammatory cytokines.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Interleucina-1/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Linhagem Celular , Fosfatases de Especificidade Dupla/genética , Técnicas de Silenciamento de Genes , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosforilação
12.
Cell Signal ; 24(5): 1002-11, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22245064

RESUMO

We have explored the possible role of dual specificity phosphatases (DUSPs) on acute EGF-mediated ERK signalling using high content imaging and a delayed MEK inhibition protocol to distinguish direct and indirect effects of the phosphatases on ERK activity. Using siRNAs, we were unable to find evidence that any of the MAPK phosphatases (MKPs) expressed in HeLa cells acts directly to dephosphorylate ppERK1/2 (dual phosphorylated ERKs 1 and/or 2) in the acute time-frame tested (0-14 min). Nevertheless, siRNAs against two p38/JNK MKPs (DUSPs 10 and 16) inhibited acute EGF-stimulated ERK activation. No such effect was seen for acute effects of the protein kinase C activator PDBu (phorbol 12,13 dibutyrate) on ERK activity, although effects of EGF and PDBu on ERK-dependent transcription (Egr-1 luciferase activity) were both reduced by siRNA targeting DUSPs 10 and 16. Inhibition of EGF-stimulated ERK activity by these siRNAs was reversed by pharmacological inhibition of p38 MAPK and single cell analysis revealed that the siRNAs did not influence the nuclear-cytoplasmic distribution of ppERK1/2. Thus, DUSPs 10 and 16 are positive regulators of activation, apparently acting by modulating cross-talk between the p38 and ERK pathways. A simplified mathematical model of this scenario accurately predicted the experimental data, supporting the conclusion that the major mechanism by which MKPs influence acute EGF-stimulated ERK responses is the negative regulation of p38, resulting in the positive regulation of ERK phosphorylation and activity.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Ativação Enzimática , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Interferência de RNA , Receptor Cross-Talk
13.
Am J Physiol Cell Physiol ; 299(2): C189-202, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20463170

RESUMO

Intracellular signaling by mitogen-activated protein (MAP) kinases (MAPK) is involved in many cellular responses and in the regulation of various physiological and pathological conditions. Tight control of the localization and duration of extracellular-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), or p38 MAPK activity is thus a fundamental aspect of cell biology. Several members of the dual-specificity phosphatase (DUSPs) family are able to dephosphorylate MAPK isoforms with different specificity, cellular, and tissue localization. Understanding how these phosphatases are themselves regulated during development or in physiological and pathological conditions is therefore fundamental. Over the years, gene deletion and knockdown studies have completed initial in vitro studies and shed a new light on the global and specific roles of DUSPs in vivo. Whereas DUSP1, DUSP2, and DUSP10 appear as crucial players in the regulation of immune responses, other members of the family, like the ERK-specific DUSP6, were shown to play a major role in development. Recent findings on the involvement of DUSPs in cancer progression and resistance will also be discussed.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Neoplasias/enzimologia , Neoplasias/etiologia , Animais , Humanos , Isoenzimas/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia
14.
Biochim Biophys Acta ; 1803(9): 1003-12, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20347885

RESUMO

Dual specificity phosphatases are characterised by their ability to dephosphorylate both phosphotyrosine and phosphoserine/threonine residues within the one substrate. The aim of this study was to characterise the phosphatase activity of the atypical dual specificity phosphatase, DUSP26 on MAP kinases, and to determine its expression, regulation and function in cancer cells. Overexpression and knockdown of DUSP26 in epithelial cells and in vitro phosphatase assays were used to demonstrate that, contrary to several published reports, DUSP26 does not act as a dual specificity phosphatase on ERK, JNK or p38 MAPKs. However, overexpression of DUSP26 in MCF10A epithelial cells suppressed colony formation and acinar growth in 3D culture, effects dependent on its phosphatase activity, while knockdown of DUSP26 in HOSE17.1 cells enhanced colony formation and cellular proliferation. DUSP26 mRNA expression was reduced in neuroblastoma, brain and ovarian cancer cell lines. Consistent with epigenetic silencing of DUSP26, expression was enhanced by treatment of cells with 5-aza-2-deoxycitidine and trichostatin A, and a CpG island upstream of the DUSP26 transcriptional start site was variably methylated in cancer cell lines. Together, these results help to clarify confusion in the literature relating to DUSP26 substrate specificity and support recent reports that substrates other than MAPKs are the primary substrates of this phosphatase. In addition, they indicate that DUSP26 may function as a tumour suppressor in particular cancers.


Assuntos
Proliferação de Células , Fosfatases de Especificidade Dupla/fisiologia , Células Epiteliais/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Animais , Células CHO , Células COS , Células Cultivadas , Chlorocebus aethiops , Ilhas de CpG/genética , Cricetinae , Cricetulus , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Genes Supressores de Tumor/fisiologia , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosforilação/fisiologia
15.
Cancer Res ; 70(4): 1689-99, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20124482

RESUMO

Critical tumor suppression pathways in brain tumors have yet to be fully defined. Along with mutational analyses, genome-wide epigenetic investigations may reveal novel suppressor elements. Using differential methylation hybridization, we identified a CpG-rich region of the promoter of the dual-specificity mitogen-activated protein kinase phosphatase-2 gene (DUSP4/MKP-2) that is hypermethylated in gliomas. In 83 astrocytic gliomas and 5 glioma cell lines examined, hypermethylation of the MKP-2 promoter was found to occur relatively more frequently in diffuse or anaplastic astrocytomas and secondary glioblastomas relative to primary glioblastomas. MKP-2 hypermethylation was associated with mutations in TP53 and IDH1, exclusive of EGFR amplification, and with prolonged survival of patients with primary glioblastoma. Expression analysis established that promoter hypermethylation correlated with reduced expression of MKP-2 mRNA and protein. Consistent with a regulatory role, reversing promoter hypermethylation by treating cells with 5-aza-2'-deoxycytidine increased MKP-2 mRNA levels. Furthermore, we found that glioblastoma cell growth was inhibited by overexpression of exogenous MKP-2. Our findings reveal MKP-2 as a common epigenetically silenced gene in glioma, the inactivation of which may play a significant role in glioma development.


Assuntos
Neoplasias Encefálicas/patologia , Proliferação de Células , Fosfatases de Especificidade Dupla/genética , Epigênese Genética/fisiologia , Glioma/patologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo/fisiologia , Fosfatases de Especificidade Dupla/metabolismo , Fosfatases de Especificidade Dupla/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Genes Supressores de Tumor/fisiologia , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia
16.
PLoS One ; 5(12): e15357, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21203456

RESUMO

In plant post-embryonic epidermis mitogen-activated protein kinase (MAPK) signaling promotes differentiation of pavement cells and inhibits initiation of stomata. Stomata are cells specialized to modulate gas exchange and water loss. Arabidopsis MAPKs MPK3 and MPK6 are at the core of the signaling cascade; however, it is not well understood how the activity of these pleiotropic MAPKs is constrained spatially so that pavement cell differentiation is promoted only outside the stomata lineage. Here we identified a PP2C-type phosphatase termed AP2C3 (Arabidopsis protein phosphatase 2C) that is expressed distinctively during stomata development as well as interacts and inactivates MPK3, MPK4 and MPK6. AP2C3 co-localizes with MAPKs within the nucleus and this localization depends on its N-terminal extension. We show that other closely related phosphatases AP2C2 and AP2C4 are also MAPK phosphatases acting on MPK6, but have a distinct expression pattern from AP2C3. In accordance with this, only AP2C3 ectopic expression is able to stimulate cell proliferation leading to excess stomata development. This function of AP2C3 relies on the domains required for MAPK docking and intracellular localization. Concomitantly, the constitutive and inducible AP2C3 expression deregulates E2F-RB pathway, promotes the abundance and activity of CDKA, as well as changes of CDKB1;1 forms. We suggest that AP2C3 downregulates the MAPK signaling activity to help maintain the balance between differentiation of stomata and pavement cells.


Assuntos
Proteínas de Arabidopsis/genética , Regulação da Expressão Gênica de Plantas , Sistema de Sinalização das MAP Quinases , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Epiderme Vegetal/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/fisiologia , Diferenciação Celular , Linhagem da Célula , Núcleo Celular/metabolismo , Proliferação de Células , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fenótipo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Transdução de Sinais , Regulação para Cima
17.
Mol Pain ; 5: 25, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19476641

RESUMO

BACKGROUND: Cannabinoid receptor type 2 (CBR2) inhibits microglial reactivity through a molecular mechanism yet to be elucidated. We hypothesized that CBR2 activation induces an anti-inflammatory phenotype in microglia by inhibiting extracellular signal-regulated kinase (ERK) pathway, via mitogen-activated protein kinase-phosphatase (MKP) induction. MKPs regulate mitogen activated protein kinases, but their role in the modulation of microglial phenotype is not fully understood. RESULTS: JWH015 (a CBR2 agonist) increased MKP-1 and MKP-3 expression, which in turn reduced p-ERK1/2 in LPS-stimulated primary microglia. These effects resulted in a significant reduction of tumor necrosis factor-alpha (TNF) expression and microglial migration. We confirmed the causative link of these findings by using MKP inhibitors. We found that the selective inhibition of MKP-1 by Ro-31-8220 and PSI2106, did not affect p-ERK expression in LPS+JWH015-treated microglia. However, the inhibition of both MKP-1 and MKP-3 by triptolide induced an increase in p-ERK expression and in microglial migration using LPS+JWH015-treated microglia. CONCLUSION: Our results uncover a cellular microglial pathway triggered by CBR2 activation. These data suggest that the reduction of pro-inflammatory factors and microglial migration via MKP-3 induction is part of the mechanism of action of CBR2 agonists. These findings may have clinical implications for further drug development.


Assuntos
Movimento Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Microglia/citologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Animais Recém-Nascidos , Fosfatase 1 de Especificidade Dupla/antagonistas & inibidores , Fosfatase 1 de Especificidade Dupla/genética , Fosfatase 1 de Especificidade Dupla/fisiologia , Fosfatase 6 de Especificidade Dupla/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Regulação Enzimológica da Expressão Gênica , Inflamação/prevenção & controle , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fenótipo , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/metabolismo , Regulação para Cima
18.
Proc Natl Acad Sci U S A ; 105(34): 12337-42, 2008 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-18719100

RESUMO

A major goal for developmental biologists is to define the behaviors and molecular contents of differentiating cells. We have devised a strategy for isolating cells from diverse embryonic regions and stages in the zebrafish, using computer-guided laser photoconversion of injected Kaede protein and flow cytometry. This strategy enabled us to perform a genome-wide transcriptome comparison of germ layer precursor cells. Mesendoderm and ectoderm precursors cells isolated by this method differentiated appropriately in transplantation assays. Microarray analysis of these cells reidentified known genes at least as efficiently as previously reported strategies that relied on artificial mesendoderm activation or inhibition. We also identified a large set of uncharacterized mesendoderm-enriched genes as well as ectoderm-enriched genes. Loss-of-function studies revealed that one of these genes, the MAP kinase inhibitor dusp4, is essential for early development. Embryos injected with antisense morpholino oligonucleotides that targeted Dusp4 displayed necrosis of head tissues. Marker analysis during late gastrulation revealed a specific loss of sox17, but not of other endoderm markers, and analysis at later stages revealed a loss of foregut and pancreatic endoderm. This specific loss of sox17 establishes a new class of endoderm specification defect.


Assuntos
Proteínas de Ligação a DNA/deficiência , Fosfatases de Especificidade Dupla/genética , Camadas Germinativas/citologia , Proteínas de Grupo de Alta Mobilidade/deficiência , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fatores de Transcrição/deficiência , Transcrição Gênica , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Animais , Fosfatases de Especificidade Dupla/fisiologia , Ectoderma/citologia , Ectoderma/embriologia , Embrião não Mamífero , Desenvolvimento Embrionário/genética , Indução Embrionária/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/embriologia , Mesoderma/citologia , Mesoderma/embriologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Fatores de Transcrição SOXF , Proteínas de Peixe-Zebra/fisiologia
19.
Cancer Res ; 67(22): 10711-9, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006813

RESUMO

The extracellular signal-regulated kinase (Erk) is one of the downstream effectors of the Ras pathway whose activation is essential for the proliferation and survival of cancer cells. Erk activation is negatively regulated by mitogen-activated protein kinase (MAPK) phosphatases (MKP), which are generally up-regulated by Erk activation, thus forming a feedback loop for regulation of Erk activity. In searching for early alterations in the Ras pathway in epidermal carcinogenesis, we identified MKP4, a cytosolic MKP with specificity to not only Erk, but also, to a lesser extent, c-jun-NH(2)-kinase and p38. MKP4 is down-regulated at initiation and lost at malignant conversion in a clonal model of epidermal carcinogenesis that lacks Ras mutation. The loss of MKP4 was associated with squamous cell carcinoma (SCC) but not benign papilloma clonal lineages and with independently induced SCC relative to benign tumors in mouse skin. Reconstitution of MKP4 expression in malignant tumor cells leads to cell death and tumor suppression. Unlike Erk inhibition that blocks cell cycle entry, MKP4 reconstitution resulted in G(2)-M associated cell death and microtubule disruption. Thus, microtubule disruption by MKP4 provides a novel mechanism for tumor suppression by a cytosolic MKP and implies a novel therapeutic strategy through combined MAPK inhibitions that mimic the function of MKP4.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Regulação Neoplásica da Expressão Gênica , Microtúbulos/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Neoplasias/patologia , Animais , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Citosol/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microtúbulos/ultraestrutura , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Neoplasias/metabolismo , Papiloma/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Cancer Metastasis Rev ; 26(3-4): 579-85, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17717636

RESUMO

The mitogen-activated protein kinase (MAPK) phosphatases (MKPs) are a family of dual-specificity protein phosphatases that dephosphorylate both phospho-threonine and phospho-tyrosine residues in MAP kinases, including the c-Jun N-terminal protein kinase (JNK)/stress-activated protein kinase (SAPK), the p38 MAPK, and the extracellular signal-related kinase (ERK). Since phosphorylation is required for the activation of MAP kinases, dephosphorylation by MKPs inhibits MAPK activity, thereby negatively regulating MAPK signaling. It is known that deregulation of MAPK signaling is the most common alteration in human cancers. Recent studies have suggested that MKPs play an important role not only in the development of cancers, but also in the response of cancer cells to chemotherapy. Thus, understanding the roles of MKPs in the development of cancer and their impact on chemotherapy can be exploited for therapeutic benefits for the treatment of human cancer.


Assuntos
Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Neoplasias/etiologia , Animais , Fosfatase 1 de Especificidade Dupla/fisiologia , Fosfatase 6 de Especificidade Dupla/fisiologia , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA