Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 926, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042954

RESUMEN

Lymphangiogenesis is essential for the development of the lymphatic system and is important for physiological processes such as homeostasis, metabolism and immunity. Cellular communication network factor 2 (CCN2, also known as CTGF), is a modular and matricellular protein and a well-known angiogenic factor in physiological and pathological angiogenesis. However, its roles in lymphangiogenesis and intracellular signaling in lymphatic endothelial cells (LECs) remain unclear. Here, we investigated the effects of CCN2 on lymphangiogenesis. In in vivo Matrigel plug assays, exogenous CCN2 increased the number of Podoplanin-positive vessels. Subsequently, we found that CCN2 induced phosphorylation of ERK in primary cultured LECs, which was almost completely inhibited by the blockade of integrin αvß5 and partially decreased by the blockade of integrin αvß3. CCN2 promoted direct binding of ERK to dual-specific phosphatase 6 (DUSP6), which regulated the activation of excess ERK by dephosphorylating ERK. In vitro, CCN2 promoted tube formation in LECs, while suppression of Dusp6 further increased tube formation. In vivo, immunohistochemistry also detected ERK phosphorylation and DUSP6 expression in Podoplanin-positive cells on CCN2-supplemented Matrigel. These results indicated that CCN2 promotes lymphangiogenesis by enhancing integrin αvß5-mediated phosphorylation of ERK and demonstrated that DUSP6 is a negative regulator of excessive lymphangiogenesis by CCN2.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Linfangiogénesis/fisiología , Receptores de Vitronectina/metabolismo , Animales , Movimiento Celular/fisiología , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasa 6 de Especificidad Dual/fisiología , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Femenino , Integrinas/genética , Integrinas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Receptores de Vitronectina/genética , Transducción de Señal/efectos de los fármacos
2.
Cell Stress Chaperones ; 24(6): 1137-1149, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31664697

RESUMEN

MAP kinase phosphatase 3 (MKP3), a member of the dual-specificity protein phosphatase (DUSP) superfamily, has been widely studied for its role in development, cancer, and environmental stress in many organisms. However, the functions of MKP3 in various insects have not been well studied, including honeybees. In this study, we isolated an MKP3 gene from Apis cerana cerana and explored the role of this gene in the resistance to oxidation. We found that AccMKP3 is highly conserved in different species and shares the closest evolutionary relationship with AmMKP3. We determined the expression patterns of AccMKP3 under various stresses. qRT-PCR results showed that AccMKP3 was highly expressed during the pupal stages and in adult muscles. We further found that AccMKP3 was induced in all the stress treatments. Moreover, we discovered that the enzymatic activities of peroxidase, superoxide dismutase, and catalase increased and that the expression levels of several antioxidant genes were affected after AccMKP3 was knocked down. Collectively, these results suggest that AccMKP3 may be associated with antioxidant processes involved in response to various environmental stresses.


Asunto(s)
Abejas , Fosfatasa 6 de Especificidad Dual , Genes de Insecto/fisiología , Proteínas de Insectos , Estrés Oxidativo/fisiología , Estrés Fisiológico/fisiología , Animales , Abejas/genética , Abejas/metabolismo , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/fisiología , Genes de Insecto/genética , Proteínas de Insectos/genética , Proteínas de Insectos/fisiología
3.
FEBS J ; 285(9): 1593-1610, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29493888

RESUMEN

Tumor necrosis factor (TNF)-α activates a diverse array of signaling pathways in vascular endothelial cells (ECs), leading to the inflammatory phenotype that contributes to the vascular dysfunction and neutrophil emigration in patients with sepsis. To date, it is not well understood what key regulator might coordinate signaling pathways to achieve inflammatory response in TNF-α-stimulated ECs. This study investigated the role of dual specificity phosphatase-6 (DUSP6) in the regulation of endothelial inflammation. Using knockout mice, we found that DUSP6 is important for TNF-α-induced endothelial intercellular adhesion molecule-1 (ICAM-1) expression in aorta and in vein. Moreover, genetic deletion of Dusp6 in pulmonary circulation significantly alleviated the susceptibility of mice to lung injury caused by neutrophil recruitment during experimental sepsis induced by TNF-α or lipopolysaccharide (LPS). The role of DUSP6 was further investigated in primary human umbilical vein endothelial cells (HUVECs). Employing RNAi approach in which endogenous DUSP6 was ablated, we showed a critical function of DUSP6 to facilitate TNF-α-induced ICAM-1 expression and endothelial leukocyte interaction. Interestingly, DUSP6-promoted endothelial inflammation is independent of extracellular signaling-regulated kinase (ERK) signaling. On the other hand, inducible DUSP6 leads to activation of canonical nuclear factor (NF)-κB-mediated transcription of ICAM-1 gene in TNF-α-stimulated human ECs. These results are the first to demonstrate a positive role of DUSP6 in endothelial inflammation-mediated pathological process and the underlying mechanism through which DUSP6 promotes NF-κB signaling in the inflamed ECs. Our findings suggest that manipulation of DUSP6 holds great potential for the treatment of acute inflammatory diseases.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Endotelio Vascular/enzimología , Regulación de la Expresión Génica/fisiología , Molécula 1 de Adhesión Intercelular/biosíntesis , Lesión Pulmonar Aguda/fisiopatología , Lesión Pulmonar Aguda/prevención & control , Traslado Adoptivo , Animales , Aorta , Adhesión Celular , Quimiotaxis de Leucocito , Fosfatasa 6 de Especificidad Dual/deficiencia , Fosfatasa 6 de Especificidad Dual/genética , Endotelio Vascular/patología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Genes Reporteros , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Molécula 1 de Adhesión Intercelular/genética , Lipopolisacáridos/farmacología , Ratones , FN-kappa B/metabolismo , Infiltración Neutrófila , Factor de Necrosis Tumoral alfa/farmacología , Células U937 , Vena Cava Inferior , Irradiación Corporal Total
4.
Development ; 145(5)2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29444893

RESUMEN

Zebrafish regenerate cardiac tissue through proliferation of pre-existing cardiomyocytes and neovascularization. Secreted growth factors such as FGFs, IGF, PDGFs and Neuregulin play essential roles in stimulating cardiomyocyte proliferation. These factors activate the Ras/MAPK pathway, which is tightly controlled by the feedback attenuator Dual specificity phosphatase 6 (Dusp6), an ERK phosphatase. Here, we show that suppressing Dusp6 function enhances cardiac regeneration. Inactivation of Dusp6 by small molecules or by gene inactivation increased cardiomyocyte proliferation, coronary angiogenesis, and reduced fibrosis after ventricular resection. Inhibition of Erbb or PDGF receptor signaling suppressed cardiac regeneration in wild-type zebrafish, but had a milder effect on regeneration in dusp6 mutants. Moreover, in rat primary cardiomyocytes, NRG1-stimulated proliferation can be enhanced upon chemical inhibition of Dusp6 with BCI. Our results suggest that Dusp6 attenuates Ras/MAPK signaling during regeneration and that suppressing Dusp6 can enhance cardiac repair.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Corazón/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regeneración/genética , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular/genética , Regulación hacia Abajo/genética , Miocitos Cardíacos/fisiología , Transducción de Señal/genética , Proteínas de Pez Cebra/fisiología
5.
Nat Med ; 21(9): 1038-47, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26301689

RESUMEN

One strategy for combating cancer-drug resistance is to deploy rational polytherapy up front that suppresses the survival and emergence of resistant tumor cells. Here we demonstrate in models of lung adenocarcinoma harboring the oncogenic fusion of ALK and EML4 that the GTPase RAS-mitogen-activated protein kinase (MAPK) pathway, but not other known ALK effectors, is required for tumor-cell survival. EML4-ALK activated RAS-MAPK signaling by engaging all three major RAS isoforms through the HELP domain of EML4. Reactivation of the MAPK pathway via either a gain in the number of copies of the gene encoding wild-type K-RAS (KRAS(WT)) or decreased expression of the MAPK phosphatase DUSP6 promoted resistance to ALK inhibitors in vitro, and each was associated with resistance to ALK inhibitors in individuals with EML4-ALK-positive lung adenocarcinoma. Upfront inhibition of both ALK and the kinase MEK enhanced both the magnitude and duration of the initial response in preclinical models of EML4-ALK lung adenocarcinoma. Our findings identify RAS-MAPK dependence as a hallmark of EML4-ALK lung adenocarcinoma and provide a rationale for the upfront inhibition of both ALK and MEK to forestall resistance and improve patient outcomes.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas de Fusión Oncogénica/fisiología , Proteínas ras/fisiología , Quinasa de Linfoma Anaplásico , Línea Celular Tumoral , Resistencia a Antineoplásicos , Fosfatasa 6 de Especificidad Dual/fisiología , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas ras/genética
6.
Exp Hematol ; 43(7): 524-33.e1, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25846811

RESUMEN

Adenosine monophosphate-activated protein kinase (AMPK) is a sensor for cellular energy status. When the cellular energy level is decreased, AMPK is activated and functions to suppress energy-consuming processes, including protein synthesis. Recently, AMPK has received attention as an attractive molecular target for cancer therapy. Several studies have revealed that the activation of AMPK by chemical stimulators, such as metformin, induces apoptosis in a variety of hematologic malignant cells. From another perspective, these results suggest that the function of AMPK is impaired in hematologic tumor cells. However, the precise mechanisms by which this impairment occurs are not well understood. In melanoma cells, oncogenic BRAF constitutively activates the extracellular signal-regulated kinase (ERK) pathway and phosphorylates liver kinase B1, an upstream activator of 5' adenosine monophosphate-activated protein kinase (AMPK), resulting in the inactivation of liver kinase B1 and AMPK. In this study, we analyzed whether ERK is involved in the suppression of AMPK activity using established and primary human leukemia cells. We found an inverse correlation between the intensity of ERK activity and the degree of AMPK activation after stimulation with either glucose deprivation or metformin. We also found that the inhibition of ERK activity by U0126 restored AMPK activation after metformin treatment. Furthermore, a combined treatment with metformin and U0126 enhanced the antileukemic activity of metformin. Importantly, metformin induced ERK activation by suppressing the protein levels of dual specificity phosphatase 6, a negative regulator of ERK. This crosstalk between AMPK and ERK could diminish the antileukemic activity of metformin. Taken together, our present observations suggest a novel therapeutic strategy for improving the efficacy of metformin in treating leukemia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/genética , Adulto , Anciano , Antineoplásicos/farmacología , Apoptosis , Butadienos/farmacología , Línea Celular Tumoral , Interacciones Farmacológicas , Fosfatasa 6 de Especificidad Dual/fisiología , Activación Enzimática , Retroalimentación Fisiológica , Femenino , Glucosa/farmacología , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mielomonocítica Aguda/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metformina/farmacología , Persona de Mediana Edad , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas
7.
Oncogene ; 33(2): 212-24, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23246965

RESUMEN

Reduced expression of the p53 family member p63 has been suggested to play a causative role in cancer metastasis. Here, we show that ΔNp63α, the predominant p63 isoform, plays a major role in regulation of cell migration, invasion and cancer metastasis. We identified mitogen-activated protein (MAP) kinase phosphatase 3 (MKP3) as a downstream target of ΔNp63α that is required for mediating these effects. We show that ΔNp63α regulates extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) activity via MKP3 in both cancer and non-transformed cells. We further show that exogenous ΔNp63α inhibits cell invasion and is dependent on MKP3 upregulation for repression. Conversely, endogenous pan-p63 ablation results in increased cell migration and invasion, which can be reverted by reintroducing the ΔNp63α isoform alone, but not by other isoforms. Interestingly, these effects require Erk2, but not Erk1 expression, and can be rescued by enforced MKP3 expression. Moreover, MKP3 expression is reduced in invasive cancers, and reduced p63 expression increases metastatic frequency in vivo. Taken together, these results suggest an important role for ΔNp63α in preventing cancer metastasis by inhibition of Erk2 signaling via MKP3.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Metástasis de la Neoplasia/prevención & control , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Ratones , Invasividad Neoplásica
8.
Br J Cancer ; 109(4): 1063-71, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23839489

RESUMEN

BACKGROUND: Dual specificity phosphatase 6 (DUSP6) is a member of a family of mitogen-activated protein kinase phosphatases that dephosphorylates and inhibits activated ERK1/2. Dual specificity phosphatase 6 is dynamically regulated in developmental and pathological conditions such as cancer. METHODS: Cancer cell lines were made deficient in DUSP6 by siRNA and shRNA silencing. Sensitivity to anti-EGFR and chemotherapeutic agents was determined in viability and apoptosis assays, and in xenografts established in SCID mice. Cellular effects of DUSP6 inactivation were analysed by proteomic methods, followed by analysis of markers of DNA damage response (DDR) and cell cycle. RESULTS: We determined that depletion of DUSP6 reduced the viability of cancer cell lines and increased the cytotoxicity of EGFR and other targeted inhibitors, and cytotoxic agents, in vitro and in vivo. Subsequent phosphoproteomic analysis indicated DUSP6 depletion significantly activated CHEK2 and p38, which function in the DDR pathway, and elevated levels of phosphorylated H2AX, ATM, and CHEK2, for the first time identifying a role for DUSP6 in regulating DDR. CONCLUSION: Our results provide a novel insight into the DUSP6 function in regulating genomic integrity and sensitivity to chemotherapy in cancer.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Fosfatasa 6 de Especificidad Dual/fisiología , Receptores ErbB/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa de Punto de Control 2 , Células HEK293 , Humanos , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones SCID , Fosforilación , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Biol Reprod ; 83(5): 807-17, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20650883

RESUMEN

A fully developed, functional epididymis is important for male fertility. In particular, it is apparent that without the most proximal region, the initial segment (IS), infertility results. Therefore, it is important to understand the development and regulation of this crucial epididymal region. We have previously shown that many functions of the IS are regulated by luminal fluid factors/lumicrine factors from the testis. This study provides evidence that lumicrine factors activated the ERK pathway only in epithelial cells of the IS from Postnatal Day (P) 14 to P19 and sustained this activation into adulthood. The activated ERK pathway promoted cell proliferation and differentiation in the developing IS, although in the adult, its role was switched to maintain cell survival. To understand further the regulation of cell proliferation in the IS, we examined the role of DUSP6, an MAPK1/3 (ERK1/2) preferred phosphatase that is also regulated by lumicrine factors in the IS. Utilizing Dusp6(-/-) mice, our studies, surprisingly, revealed that Dusp6 was a major regulator of cell proliferation in the caput and corpus regions, whereas components of the ERK pathway, together with PTEN and SRC, were the major regulators of cell proliferation in the IS. We hypothesize that region-specific regulation of cell proliferation is caused by differences in the balance of activities between pro- and antiproliferation signaling pathway components for each epididymal region. An understanding of the mechanisms of cell proliferation may provide clues as to why the epididymis rarely succumbs to cancer.


Asunto(s)
Proliferación Celular , Fosfatasa 6 de Especificidad Dual/fisiología , Epidídimo/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Envejecimiento , Animales , Supervivencia Celular , Fosfatasa 6 de Especificidad Dual/genética , Epidídimo/citología , Epidídimo/crecimiento & desarrollo , Epidídimo/cirugía , Regulación del Desarrollo de la Expresión Génica , Genes src/fisiología , Ligadura , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Especificidad de Órganos , Fosfohidrolasa PTEN/fisiología , Fosforilación , Análisis por Matrices de Proteínas , ARN Mensajero/metabolismo , Hormonas Testiculares/fisiología
10.
J Leukoc Biol ; 88(1): 9-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20200403

RESUMEN

The role of phosphatases in the impairment of MAPK signaling, which is directly responsible for Leishmania-induced macrophage dysfunction, is still poorly understood. Gene expression profiling revealed that Leishmania donovani infection markedly up-regulated the expression of three phosphatases: MKP1, MKP3, and PP2A. Inhibition of these phosphatases prior to infection points toward preferential induction of the Th2 response through deactivation of p38 by MKP1. On the other hand, MKP3 and PP2A might play significant roles in the inhibition of iNOS expression through deactivation of ERK1/2. Among various PKC isoforms, PKCzeta was associated with induction of MKP3 and PP2A in infected macrophages, whereas PKCepsilon was correlated with MKP1 induction. Inhibition of phosphatases in L. donovani-infected BALB/c mice shifted the cytokine balance in favor of the host by inducing TNF-alpha and iNOS expression. This was validated by cystatin, an immunomodulator and curing agent for experimental visceral leishmaniasis, which showed that inhibition of MKPs and PP2A activity may be necessary for a favorable T cell response and suppression of organ parasite burden. This study, for the first time, suggests the possibility of the involvement of MAPK-directed phosphatases in the establishment of L. donovani infection.


Asunto(s)
Citocinas/biosíntesis , Fosfatasa 1 de Especificidad Dual/fisiología , Fosfatasa 6 de Especificidad Dual/fisiología , Isoenzimas/fisiología , Leishmaniasis Visceral/inmunología , Proteína Quinasa C/fisiología , Proteína Fosfatasa 2/fisiología , Animales , Células Cultivadas , Cistatinas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/genética , Células TH1/inmunología , Células Th2/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Carcinogenesis ; 31(4): 577-86, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20097731

RESUMEN

Mitogen-activated protein kinase (MAPK) pathway signaling plays an important role in the majority of non-small-cell lung cancers (NSCLCs). In a prior microarray analysis of epidermal growth factor receptor (EGFR) inhibition in NSCLC cell lines, we noted that several dual specificity phosphatases (DUSPs) were among the most highly and immediately regulated genes. DUSPs act as natural terminators of MAPK signal transduction and therefore, we hypothesized a tumor suppressive role via feedback mechanisms. In the current study, we focus on the assessment of DUSP6, a cytoplasmic DUSP with high specificity for extracellular signal-regulated kinase (ERK). We demonstrate that DUSP6 expression tracks in tandem with ERK inhibition and that regulation of DUSP6 is mediated at the promoter level by ETS1, a well-known nuclear target of activated ERK. Small interfering RNA knockdown in DUSP6-high H441 lung cancer cells significantly increased ERK activation and cellular proliferation, whereas plasmid-driven overexpression in DUSP6-low H1975 lung cancer cells significantly reduced ERK activation and cellular proliferation and promoted apoptosis. Also, DUSP6 overexpression synergized with EGFR inhibitor treatment in EGFR-mutant HCC827 cells. Our results indicate that DUSP6 expression is regulated by ERK signaling and that DUSP6 exerts antitumor effects via negative feedback regulation, pointing to an important feedback loop in NSCLC. Further studies assessing the tumor suppressive role of DUSP6 and strategies aimed at modulation of its activity are warranted.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Neoplasias Pulmonares/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Proto-Oncogénica c-ets-1/fisiología , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular Tumoral , Receptores ErbB/fisiología , Retroalimentación Fisiológica , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , ARN Interferente Pequeño/genética
12.
J Cell Sci ; 122(Pt 23): 4303-10, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19887586

RESUMEN

Phosphatidylinositol 4-kinases (PI4Ks) catalyze the first committed step in the synthesis of phosphoinositides, important lipid regulators of signaling and trafficking pathways. Here we cloned Pik4a, one of the zebrafish PI4K enzymes, and studied its role(s) in vertebrate development using morpholino oligonucleotide-based gene silencing in zebrafish. Downregulation of Pik4a led to multiple developmental abnormalities, affecting the brain, heart, trunk and most prominently causing loss of pectoral fins. Strikingly similar defects were caused by treatment of the developing embryos with the phosphoinositide 3-kinase (PI3K) inhibitor, LY294002. To investigate the cause of the pectoral fin developmental defect, we focused on fibroblast growth factor (FGF) signaling pathways because vertebrate limb development requires the concerted action of a series of FGF ligands. Using in situ hybridization, the pectoral fin defect was traced to disruption of the early FGF signaling loops that are crucial for the establishment of the sharp signaling center formed by the apical ectodermal ridge and the underlying mesenchyme. This, in turn caused a prominent loss of the induction of one of the mitogen-activated protein kinase (MAPK) phosphatases, Mkp3, an essential intermediate in vertebrate limb development. These changes were associated with impaired proliferation in the developing fin bud due to a loss of balance between the MAPK and PI3K branch of FGF-initiated signals. Our results identify Pik4a as an upstream partner of PI3Ks in the signaling cascade orchestrated by FGF receptors with a prominent role in forelimb development.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/fisiología , Extremidades/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas de Pez Cebra/fisiología , 1-Fosfatidilinositol 4-Quinasa/genética , Animales , Cromonas/farmacología , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Hibridación Fluorescente in Situ , Etiquetado Corte-Fin in Situ , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética
13.
Nat Immunol ; 10(9): 936-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19692992

RESUMEN

Like every metazoan species hosting a gut flora, drosophila tolerate commensal microbiota yet remain able to mount an efficient immune response to food-borne pathogens. New findings explain how the quantity of reactive oxygen species in the gut is 'tuned' to microbial burden and how intestinal immune homeostasis is thereby maintained


Asunto(s)
Drosophila melanogaster/inmunología , Inmunidad Innata , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Proteínas de Drosophila/fisiología , Fosfatasa 6 de Especificidad Dual/fisiología , Homeostasis , Intestinos/inmunología , Intestinos/microbiología , NADPH Oxidasas/fisiología , Fosfolipasa C beta/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
14.
FEBS J ; 276(4): 1024-35, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19154344

RESUMEN

Mitogen-activated protein kinase (MAPK) signaling determines crucial cell fate decisions in most cell types, and mediates cellular transformation in many types of cancer. The activity of MAPK is controlled by reversible phosphorylation, and the quantitative characteristics of MAPK activation determine the cellular response. Many systems biological studies have analyzed the activation kinetics and the dose-response behavior of the MAPK signaling pathway. Here we investigate how the pathway activity is controlled by transcriptional feedback loops. Initially, we predict that MAPK signaling regulates phosphatases, by integrating promoter sequence data and ontology-based classification of gene function. From this, we deduce that MAPK signaling might be controlled by transcriptional negative feedback regulation via dual-specificity phosphatases (DUSPs), and implement a mathematical model to further test this hypothesis. Using time-resolved measurements of pathway activity and gene expression, we employ a model selection approach, and select DUSP6 as a highly likely candidate for shaping the activity of the MAPK pathway during cellular transformation caused by oncogenic RAS. Two predictions from the model were confirmed: first, feedback regulation requires that DUSP6 mRNA and protein are unstable; and second, the activation kinetics of MAPK are ultrasensitive. Taken together, an integrated systems biological approach reveals that transcriptional negative feedback controls the kinetics and the extent of MAPK activation under both physiological and pathological conditions.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica , Genes ras , Modelos Biológicos , Animales , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Estabilidad del ARN , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/fisiología , Biología de Sistemas
15.
Toxicol Appl Pharmacol ; 232(3): 408-17, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18771677

RESUMEN

Mitogen activated protein kinase phosphatase-3 (MKP-3) is a putative tumor suppressor. When transiently overexpressed, MKP-3 dephosphorylates and inactivates extracellular signal regulated kinase (ERK) 1/2. Little is known about the roles of endogenous MKP-3, however. We previously showed that MKP-3 is upregulated in cell lines that express oncogenic Ras. Here we tested the roles of endogenous MKP-3 in modulating ERK1/2 under conditions of chronic stimulation of the Ras/Raf/MEK1/2/ERK1/2 pathway by expression of oncogenic Ras. We used two cell lines: H-ras MCF10A, breast epithelial cells engineered to express H-Ras, and DLD-1, colon cancer cells that express endogenous Ki-Ras. First, we found that MKP-3 acts in a negative feedback loop to suppress basal ERK1/2 when oncogenic Ras stimulates the Ras/Raf/MEK1/2/ERK1/2 cascade. ERK1/2 was required to maintain elevated MKP-3, indicative of a negative feedback loop. Accordingly, knockdown of MKP-3, via siRNA, increased ERK1/2 phosphorylation. Second, by using siRNA, we found that MKP-3 helps establish the sensitivity of ERK1/2 to extracellular activators by limiting the duration of ERK1/2 phosphorylation. Third, we found that the regulation of ERK1/2 by MKP-3 is countered by the complex regulation of MKP-3 by ERK1/2. Potent ERK1/2 activators stimulated the loss of MKP-3 within 30 min due to an ERK1/2-dependent decrease in MKP-3 protein stability. MKP-3 levels recovered within 120 min due to ERK1/2-dependent resynthesis. Preventing MKP-3 resynthesis, via siRNA, prolonged ERK1/2 phosphorylation. Altogether, these results suggest that under the pressure of oncogenic Ras expression, MKP-3 reins in ERK1/2 by serving in ERK1/2-dependent negative feedback pathways.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Línea Celular Tumoral , Genes ras , Humanos , Fosforilación
16.
Mol Biol Cell ; 19(7): 2818-29, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18434601

RESUMEN

The antidepressant and cocaine sensitive plasma membrane monoamine transporters are the primary mechanism for clearance of their respective neurotransmitters and serve a pivotal role in limiting monoamine neurotransmission. To identify molecules in pathways that regulate dopamine transporter (DAT) internalization, we used a genetic complementation screen in Xenopus oocytes to identify a mitogen-activated protein (MAP) kinase phosphatase, MKP3/Pyst1/DUSP6, as a molecule that inhibits protein kinase C-induced (PKC) internalization of transporters, resulting in enhanced DAT activity. The involvement of MKP3 in DAT internalization was verified using both overexpression and shRNA knockdown strategies in mammalian cell models including a dopaminergic cell line. Although the isolation of MKP3 implies a role for MAP kinases in DAT internalization, MAP kinase inhibitors have no effect on internalization. Moreover, PKC-dependent down-regulation of DAT does not correlate with the phosphorylation state of several well-studied MAP kinases (ERK1/2, p38, and SAPK/JNK). We also show that MKP3 does not regulate PKC-induced ubiquitylation of DAT but acts at a more downstream step to stabilize DAT at the cell surface by blocking dynamin-dependent internalization and delaying the targeting of DAT for degradation. These results indicate that MKP3 can act to enhance DAT function and identifies MKP3 as a phosphatase involved in regulating dynamin-dependent endocytosis.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Dopamina/metabolismo , Fosfatasa 6 de Especificidad Dual/genética , Regulación de la Expresión Génica , Animales , Transporte Biológico , Línea Celular Tumoral , Fosfatasa 6 de Especificidad Dual/fisiología , Dinaminas/metabolismo , Endocitosis , Prueba de Complementación Genética , Humanos , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Xenopus laevis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Cancer Metastasis Rev ; 26(3-4): 579-85, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17717636

RESUMEN

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.


Asunto(s)
Fosfatasas de la Proteína Quinasa Activada por Mitógenos/fisiología , Neoplasias/etiología , Animales , Fosfatasa 1 de Especificidad Dual/fisiología , Fosfatasa 6 de Especificidad Dual/fisiología , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...