Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cell Rep ; 38(2): 110223, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021072

RESUMEN

MEK1 and MEK2, the only known activators of ERK, are attractive therapeutic candidates for both cancer and autoimmune diseases. However, how MEK signaling finely regulates immune cell activation is only partially understood. To address this question, we specifically delete Mek1 in hematopoietic cells in the Mek2 null background. Characterization of an allelic series of Mek mutants reveals the presence of distinct degrees of spontaneous B cell activation, which are inversely proportional to the levels of MEK proteins and ERK activation. While Mek1 and Mek2 null mutants have a normal lifespan, 1Mek1 and 1Mek2 mutants retaining only one functional Mek1 or Mek2 allele in hematopoietic cell lineages die from glomerulonephritis and lymphoproliferative disorders, respectively. This establishes that the fine-tuning of the ERK/MAPK pathway is critical to regulate B and T cell activation and function and that each MEK isoform plays distinct roles during lymphocyte activation and disease development.


Asunto(s)
Activación de Linfocitos/fisiología , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Alelos , Animales , Linfocitos B/metabolismo , Femenino , Humanos , Activación de Linfocitos/genética , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/fisiología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosforilación , Transducción de Señal/fisiología , Linfocitos T/metabolismo
2.
Chembiochem ; 18(16): 1593-1598, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28561913

RESUMEN

Interference from endogenous signaling enzymes represents a major hurdle for building orthogonal signaling cascades inside cells, particularly among closely related isoforms within an enzyme family. Here, we employed a genetically encoded chemical decaging strategy to build orthogonally activated kinase isoforms, with the endogenous counterparts temporally disabled by an extracellularly delivered bacterial effector. This approach eliminated any potential interference from other kinase isoforms as well as endogenous kinases, which allowed the specific, gain-of-function report of mitogen-activated protein kinase kinase 1 (MEK1) activity as opposed to MEK2 with high temporal resolution. Our study dissected the distinct enzymatic activity, feedback regulation and signal outputs between these closely related kinase isoforms.


Asunto(s)
MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Reacción de Cicloadición , Retroalimentación Fisiológica , Células HEK293 , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Interleucina-8/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/fisiología , Lisina/análogos & derivados , Lisina/química , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/química , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/química , MAP Quinasa Quinasa 2/genética , Redes y Vías Metabólicas , Fosforilación , Ingeniería de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 42(4): 380-388, 2017 Apr 28.
Artículo en Chino | MEDLINE | ID: mdl-28490694

RESUMEN

OBJECTIVE: To observe effect of acupuncture combined with hypothermia therapy on MAPK/ERK pathway and apoptosis related factorsin rats suffered cerebral ischemia reperfusion and to explore underlying mechanisms.
 Methods: Middle cerebral artery ischemia model were established.Ninety SD rats were randomly assigned into a blank group, a control group, a model group, an acupuncture group, a mild hypothermia group, and an acupuncture with hypothermia group. After 72 h treatment, nerve function defect scores were observed, and infarction area percent was detected by 2, 3, 5-triphenyl-2H-tetrazolium chloride (TTC) staining; expressions of Bcl-2 and Bax were examined by immunohistochemistry; apoptotic cells were detected by TUNEL assay; and expression levels of phospho-mitogen-activated protein kinase(p-MEK2) and phospho-extracellular signal regulated kinase 1/2 (p-ERK1/2) in the rats' hippocampus ischemic side were determined by Western blot.
 Results: In the rats of the model group, the neural function defect scores, the infarction area percent, the expression level of Bax, and apoptotic cells increased, while the level of Bcl-2 decreased significantly. The level of p-MEK2 and p-ERK1/2 increased obviously compared with the blank and control groups (P<0.05 or P<0.01). After treatment with acupuncture and hypothermia, the neural function defect scores, infarction area percent, and the level of Bax, apoptotic cells and the levels of p-MEK2 and p-ERK1/2 were significantly decreased, while the level of Bcl-2 in the treatment group was significantly elevated (P<0.05 or P<0.01) compared with the model group. Compared with the acupuncture group or the hypothermia group, the neural function defect scores and the levels of p-MEK2 and p-ERK1/2 in the acupuncture combined with hypothermia group were significantly reduced (P<0.05 or P<0.01).
 Conclusion: Acupuncture and hypothermia therapy can improve cerebral function, and reduce the cerebral injury through down-regulation of Bax level, and up-regulation of Bcl-2 level, which is related to reducing the levels of p-MEK2 and p-ERK1/2. The therapeutic effects on cerebral ischemia reperfusion injury for combination of acupuncture with hypothermia are better than those with single application of acupuncture or hypothermia.


Asunto(s)
Terapia por Acupuntura , Hipotermia Inducida , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Apoptosis/fisiología , Infarto Encefálico/terapia , Lesiones Encefálicas/terapia , Isquemia Encefálica/terapia , Regulación hacia Abajo , MAP Quinasa Quinasa 2/fisiología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/terapia , Regulación hacia Arriba
4.
Respir Res ; 16: 93, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242183

RESUMEN

BACKGROUND: Polo-like kinase 1 (Plk1) is a serine/threonine protein kinase that has been implicated in the regulation of mitosis. In addition, the activation of mitogen-activated protein kinase (MAPK) is a key event in the early stage of the growth factor response. The role of Plk1 in MAPK phosphorylation in cells has not been investigated. METHODS: Immunoblot analysis was used to evaluate Plk1 and MAPK phosphorylation in cells upon stimulation with platelet-derived growth factor (PDGF). We also generated stable Plk1 knockdown (KD) cells to assess the role of Plk1 in MAPK activation and cell proliferation. Furthermore, we used a non-phosphorylatable Plk1 mutant to determine the function of Plk1 phosphorylation in these processes. RESULTS: Treatment with PDGF increased Plk1 phosphorylation at Thr-210 (an indication of Plk1 activation) in human airway smooth muscle cells. Plk1 KD attenuated the PDGF-induced phosphorylation of MEK1/2 and ERK1/2 as well as cell proliferation. However, phosphorylation of Raf-1 and AKT upon stimulation with PDGF was not reduced in Plk1 KD cells. Furthermore, the expression of T210A Plk1 (alanine substitution at Thr-210) inhibited the PDGF-stimulated MEK1/2 phosphorylation, ERK1/2 phosphorylation and cell proliferation. CONCLUSIONS: Together, these findings suggest that Plk1 is activated upon growth factor stimulation, which may control the activation of MEK1/2 and ERK1/2, and smooth muscle cell proliferation.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proliferación Celular/fisiología , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Miocitos del Músculo Liso/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Bronquios/citología , Bronquios/fisiología , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Tráquea/citología , Tráquea/fisiología , Quinasa Tipo Polo 1
5.
Arch Pharm Res ; 38(12): 2153-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26119076

RESUMEN

It has been reported that alkaloids derived from Coptis chinensis exert anti-adipogenic activity on 3T3-L1 adipocytes by downregulating peroxisome proliferation-activity receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein-α (C/EBP-α). However, the signaling-based mechanism of the inhibitory role of epiberberine in the early stages of 3T3-L1 adipocyte differentiation is uncharacterized. Here, we show that epiberberine had inhibitory effects on adipocyte differentiation and significantly decreased lipid accumulation by downregulating an adipocyte-specific transcription factor, sterol regulatory element-binding protein-1 (SREBP-1). Furthermore, we observed that epiberberine markedly suppressed the differentiation-mediated phosphorylation of components of both the Raf/mitogen-activated protein kinase 1 (MEK1)/extracellular signal-regulated protein kinase 1/2 (ERK1/2) and AMP-activated protein kinase-α1 (AMPKα)/Akt pathways. In addition, gene expression of fatty acid synthase (FAS) was significantly inhibited by treatment with epiberberine during adipogenesis. These results indicate that the anti-adipogenic mechanism of epiberberine is associated with inhibition of phosphorylation of Raf/MEK1/ERK1/2 and AMPKα/Akt, followed by downregulation of the major transcription factors of adipogenesis, such as PPAR-γ, C/EBP-α, and SREBP-1, and FAS. Taken together, this study suggests that the anti-adipogenic effect of epiberberine is mediated by downregulation of the Raf/MEK1/ERK1/2 and AMPKα/Akt pathways during 3T3-L1 adipocyte differentiation. Moreover, the anti-adipogenic effects of epiberberine were not accompanied by modulation of ß-catenin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Adipogénesis/fisiología , Berberina/análogos & derivados , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína Oncogénica v-akt/fisiología , Quinasas raf/fisiología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Adipogénesis/efectos de los fármacos , Animales , Fármacos Antiobesidad/farmacología , Berberina/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteína Oncogénica v-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Quinasas raf/antagonistas & inhibidores
6.
Development ; 141(16): 3197-211, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25100655

RESUMEN

The mammalian genome contains two ERK/MAP kinase genes, Mek1 and Mek2, which encode dual-specificity kinases responsible for ERK/MAP kinase activation. In order to define the function of the ERK/MAPK pathway in the lung development in mice, we performed tissue-specific deletions of Mek1 function on a Mek2 null background. Inactivation of both Mek genes in mesenchyme resulted in several phenotypes, including giant omphalocele, kyphosis, pulmonary hypoplasia, defective tracheal cartilage and death at birth. The absence of tracheal cartilage rings establishes the crucial role of intracellular signaling molecules in tracheal chondrogenesis and provides a putative mouse model for tracheomalacia. In vitro, the loss of Mek function in lung mesenchyme did not interfere with lung growth and branching, suggesting that both the reduced intrathoracic space due to the dysmorphic rib cage and the omphalocele impaired lung development in vivo. Conversely, Mek mutation in the respiratory epithelium caused lung agenesis, a phenotype resulting from the direct impact of the ERK/MAPK pathway on cell proliferation and survival. No tracheal epithelial cell differentiation occurred and no SOX2-positive progenitor cells were detected in mutants, implying a role for the ERK/MAPK pathway in trachea progenitor cell maintenance and differentiation. Moreover, these anomalies were phenocopied when the Erk1 and Erk2 genes were mutated in airway epithelium. Thus, the ERK/MAPK pathway is required for the integration of mesenchymal and epithelial signals essential for the development of the entire respiratory tract.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Sistema de Señalización de MAP Quinasas , Sistema Respiratorio/embriología , Animales , Apoptosis , Secuencia de Bases , Cartílago/embriología , Diferenciación Celular , Proliferación Celular , Perfilación de la Expresión Génica , Pulmón/embriología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Fenotipo , Células Madre/citología , Tráquea/embriología
7.
Gynecol Oncol ; 133(1): 128-37, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24434059

RESUMEN

Mitogen-activated protein kinases (MAPKs) are a family of ubiquitous eukaryotic signal transduction enzymes which link extracellular stimuli to intracellular gene expression pathways. While several three-tiered MAPK cascades have been elucidated in mammals, the prototypical pathway involves a network of proteins and kinases including the Rat sarcoma protein (Ras), mitogen-activated protein kinase kinase kinase (Raf or MAP3K), mitogen-activated protein kinase kinase (MEK or MAP2K), and extracellular signal regulated protein kinase (ERK or MAPK). This MAPK cascade (the Ras/Raf/MEK/ERK pathway) is a receptor tyrosine kinase mediated signaling pathway that regulates cell proliferation, cell cycle progression, and cell migration. There are multiple molecular mechanisms of interaction and activation between the upstream nodes of the Ras/Raf/MEK/ERK cascade and other cell signaling pathways, all ultimately leading to the activation of the nuclear transcription factor ERK. Important downstream targets include MEK1/2, which comprise the final step leading to ERK transcription factor activation. While multiple conduits exist to activate ERK upstream of MEK, there is little redundancy downstream. Located at this pivotal intersection between a limited number of upstream activators and its exclusive downstream targets, MEK is an appealing molecular target of novel cancer therapies. MEK inhibitors are small molecules that inhibit MEK phosphorylation by binding to a pocket adjacent to the ATP binding site, decreasing both the amount of MEK activity, and the quantity of activated ERK in the cell. Unique allosteric noncompetitive binding sites of MEK inhibitors allow specific targeting of MEK enzymes and prevent cross-activation of other serine/threonine protein kinases through the conserved ATP binding site. This paper reviews the translational evidence in favor of MEK inhibitors in cancer, their role in gynecologic malignancies, and details regarding the status of the fourteen MEK inhibitors currently being clinically tested: trametinib, selumetinib, pimasertib, refametinib, PD-0325901, MEK162, TAK733, RO5126766, WX-554, RO4987655, cobimetinib, AZD8330, MSC2015103B, and ARRY-300.


Asunto(s)
Carcinoma/tratamiento farmacológico , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Carcinoma/enzimología , Neoplasias Colorrectales/tratamiento farmacológico , Femenino , Humanos , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias Ováricas/enzimología
8.
Gastroenterology ; 146(3): 822-834.e7, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24315826

RESUMEN

BACKGROUND & AIMS: Kras signaling via mitogen-activated protein kinase (MAPK) is highly up-regulated in pancreatic cancer cells. We investigated whether MAPK signaling is required for the initiation and maintenance of pancreatic carcinogenesis in mice. METHODS: We studied the formation and maintenance of pancreatic intraepithelial neoplasia (PanINs) in p48Cre; TetO-KrasG12D; Rosa26(rtTa-IRES-EGFP) (iKras*) mice and LSL-KrasG12D mice bred with p48Cre mice (KC). Mice were given oral PD325901, a small-molecule inhibitor of MEK1 and MEK2 (factors in the MAPK signaling pathway), along with injections of cerulein to induce pancreatitis. Other mice were given PD325901 only after PanINs developed. Pancreatic tissues were collected and evaluated using histologic, immunohistochemical, immunofluorescence, and electron microscopy analyses. Acinar cells were isolated from the tissues and the effects of MEK1 and 2 inhibitors were assessed. RESULTS: PD325901 prevented PanIN formation, but not pancreatitis, in iKras* and KC mice. In iKras* or KC mice given PD325901 at 5 weeks after PanINs developed, PanINs regressed and acinar tissue regenerated. The regression occurred through differentiation of the PanIN cells to acini, accompanied by re-expression of the acinar transcription factor Mist1. CONCLUSIONS: In iKras* and KC mice, MAPK signaling is required for the initiation and maintenance of pancreatic cancer precursor lesions. MAPK signaling promotes formation of PanINs by enabling dedifferentiation of acinar cells into duct-like cells that are susceptible to transformation.


Asunto(s)
Células Acinares/patología , Carcinoma in Situ/fisiopatología , Desdiferenciación Celular/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neoplasias Pancreáticas/fisiopatología , Transducción de Señal/fisiología , Células Acinares/fisiología , Animales , Carcinoma in Situ/patología , Modelos Animales de Enfermedad , Femenino , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Mutantes , Neoplasias Pancreáticas/patología , Pancreatitis/fisiopatología , Regulación hacia Arriba/fisiología
9.
Neuropharmacology ; 67: 78-87, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23154302

RESUMEN

We investigated whether combinatorial post-injury treatment with progesterone (P4) and vitamin D hormone (VDH) would reduce ischemic injury more effectively than P4 alone in an oxygen glucose deprivation (OGD) model in primary cortical neurons and in a transient middle cerebral artery occlusion (tMCAO) model in rats. In the OGD model, P4 and VDH each showed neuroprotection individually, but combination of the "best" doses did not show substantial efficacy; instead, the lower dose of VDH in combination with P4 was the most effective. In the tMCAO model, P4 and VDH were given alone or in combination at different times post-occlusion for 7 days. In vivo data confirmed the in vitro findings and showed better infarct reduction at day 7 and functional outcomes (at 3, 5 and 7 days post-occlusion) after combinatorial treatment than when either agent was given alone. VDH, but not P4, upregulated heme oxygenase-1, suggesting a pathway for the neuroprotective effects of VDH differing from that of P4. The combination of P4 and VDH activated brain-derived neurotrophic factor and its specific receptor, tyrosine kinase receptor-B. Under specific conditions VDH potentiates P4's neuroprotective efficacy and should be considered as a potential partner of P4 in a low-cost, safe and effective combinatorial treatment for stroke.


Asunto(s)
Isquemia Encefálica/prevención & control , Factor Neurotrófico Derivado del Encéfalo/fisiología , MAP Quinasa Quinasa 2/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Progesterona/administración & dosificación , Receptor trkB/fisiología , Accidente Cerebrovascular/prevención & control , Vitamina D/administración & dosificación , Animales , Células Cultivadas , Quimioterapia Combinada , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Resultado del Tratamiento
10.
APMIS ; 121(4): 299-310, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23030595

RESUMEN

Interaction between the immune system and cancer cells allows for the use of biological response modifiers, like OK-432, in cancer therapy. We have studied the involvement of monocytes (MOs) in the immune response to OK-432 by examining MCP-1, MIP-1α and MIP-1ß secretion, in vitro. OK-432-induced IL-6/TNF-α secretion has previously been shown to depend on mitogen-activated protein kinases (MAPKs) ERK1/2 and p38, and we therefore investigated the role of these MAPKs in OK-432-induced chemokine secretion. Here we demonstrate that pharmacological MEK1/2 kinase inhibition generally impaired chemokine secretion from MOs, whereas p38 MAPK inhibition in particular reduced MIP-1α production. Furthermore, simultaneous inhibition of MEK1/2 and Syk kinase was seen to have an additive impact on reduced MCP-1, MIP-1α and MIP-1ß secretion. Based on single cell flow cytometry analyses, OK-432, lipoteichoic acid (LTA) and lipopolysaccharide (LPS) were seen to induce p38 MAPK and NF-κB phosphorylation in MOs with different time kinetics. LTA and LPS have been shown to induce ERK1/2 phosphorylation, whereas the levels of phosphorylated ERK1/2 remained constant following OK-432 treatment at the time points tested. Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns, and we demonstrate increased TLR2 cell surface levels on the MO population, most profoundly following stimulation with LTA and OK-432. Together these results indicate that modulation of MEK1/2 and p38 MAPK signalling could affect the response to OK-432 treatment, having the potential to improve its therapeutic potential within cancer and lymphangioma treatment.


Asunto(s)
Quimiocinas/metabolismo , Factores Inmunológicos/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/efectos de los fármacos , FN-kappa B/metabolismo , Picibanil/farmacología , Butadienos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/fisiología , Monocitos/inmunología , Monocitos/metabolismo , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinasa Syk , Ácidos Teicoicos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Cell Cycle ; 11(17): 3237-49, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22895183

RESUMEN

The p53R2 protein, a newly identified member of the ribonucleotide reductase family that provides nucleotides for DNA damage repair, is directly regulated by p53. We show that p53R2 is also regulated by a MEK2 (ERK kinase 2/MAP kinase kinase 2)-dependent pathway. Increased MEK1/2 phosphorylation by serum stimulation coincided with an increase in the RNR activity in U2OS and H1299 cells. The inhibition of MEK2 activity, either by treatment with a MEK inhibitor or by transfection with MEK2 siRNA, dramatically decreased the serum-stimulated RNR activity. Moreover, p53R2 siRNA, but not R2 siRNA, significantly inhibits serum-stimulated RNR activity, indicating that p53R2 is specifically regulated by a MEK2-dependent pathway. Co-immunoprecipitation analyses revealed that the MEK2 segment comprising amino acids 65-171 is critical for p53R2-MEK2 interaction, and the binding domain of MEK2 is required for MEK2-mediated increased RNR activity. Phosphorylation of MEK1/2 was greatly augmented by ionizing radiation, and RNR activity was concurrently increased. Ionizing radiation-induced RNR activity was markedly attenuated by transfection of MEK2 or p53R2 siRNA, but not R2 siRNA. These data show that MEK2 is an endogenous regulator of p53R2 and suggest that MEK2 may associate with p53R2 and upregulate its activity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reparación del ADN/genética , Regulación Enzimológica de la Expresión Génica/fisiología , MAP Quinasa Quinasa 2/metabolismo , Ribonucleótido Reductasas/metabolismo , Anticuerpos Monoclonales , Western Blotting , Línea Celular Tumoral , Rayos gamma , Vectores Genéticos/genética , Humanos , Inmunoprecipitación , MAP Quinasa Quinasa 2/fisiología , Fosforilación , Interferencia de ARN , Conteo por Cintilación
12.
Med Sci (Paris) ; 28(4): 409-15, 2012 Apr.
Artículo en Francés | MEDLINE | ID: mdl-22549869

RESUMEN

The ERK/MAPK signaling pathway is involved in several cellular functions. Inactivation in mice of genes encoding members of this pathway is often associated with embryonic death resulting from abnormal placental development. The placenta is essential for nutritional and gaseous exchanges between maternal and embryonic circulations, as well as for the removal of metabolic wastes. These exchanges take place without direct contact between the two circulations. In mice, the hematoplacental barrier consists in a triple layer of trophoblast cells and endothelial cells of the embryo. MEK1 and MEK2 are double specificity serine-threonine/tyrosine kinases responsible for the activation of ERK1 and ERK2. Mek1 inactivation results in placental anomalies due to trophoblast cell proliferation and differentiation defects leading to severe delays in the development of placenta and causing the death of the embryo. Although Mek2(-/-) mutant mice survived without any apparent phenotype, double heterozygous Mek1(+/-)Mek2(+/-) mutants die during gestation from placental malformations. Together, these data emphasize the crucial role of the ERK/MAPK cascade in the formation of extraembryonic structures.


Asunto(s)
MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Placenta/metabolismo , Circulación Placentaria/genética , Animales , Barrera Hematoacuosa/metabolismo , Barrera Hematoacuosa/fisiología , Femenino , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Circulación Placentaria/fisiología , Embarazo
13.
Neurochem Res ; 36(7): 1304-13, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21369758

RESUMEN

Caspase plays an important role in apoptosis and physiological processes such as synaptic plasticity. However, the caspase substrate at the synapse is still unknown. Here we used an in vitro cleavage assay with a small-pool human brain cDNA library. We identified the presynaptic protein Caytaxin as a substrate of caspase-3 and caspase-7. Deficiency in Caytaxin causes Cayman ataxia, a disorder characterized by cerebellar dysfunction and mental retardation. Caytaxin cleavage in cerebellar granule neurons is dependent on caspase-3 activation. The cleavage site is upstream of the cellular retinal and the TRIO guanine exchange factor domain, producing a C-terminal fragment that may play an alternative role in inhibiting MEK2 signaling. Thus, we concluded that Caytaxin is a novel substrate of caspase-3 at the presynapse.


Asunto(s)
Caspasa 3/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Caspasa 7/metabolismo , Línea Celular , Humanos , MAP Quinasa Quinasa 2/fisiología , Ratones , Datos de Secuencia Molecular , Alineación de Secuencia , Transducción de Señal , Sinapsis/metabolismo
14.
PLoS One ; 6(2): e17165, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21365009

RESUMEN

Mitogen-activated protein kinase kinases (MKK or MEK) 1 and 2 are usually treated as redundant kinases. However, in assessing their relative contribution towards ERK-mediated biologic response investigators have relied on tests of necessity, not sufficiency. In response we developed a novel experimental model using lethal toxin (LeTx), an anthrax toxin-derived pan-MKK protease, and genetically engineered protease resistant MKK mutants (MKKcr) to test the sufficiency of MEK signaling in melanoma SK-MEL-28 cells. Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK1cr. Microarray analysis revealed non-overlapping downstream transcriptional targets of MEK1 and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL-28 cells expressing MKK1cr but not MEK2cr. These results indicate in SK-MEL-28 cells MEK1 and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. We conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK1 conditionally compensates for loss of MEK2 only in the presence of other MKK.


Asunto(s)
Proliferación Celular , MAP Quinasa Quinasa 2/fisiología , Melanoma/patología , Neoplasias Cutáneas/patología , Animales , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/farmacología , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Células CHO , Dominio Catalítico/efectos de los fármacos , Dominio Catalítico/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Cricetinae , Cricetulus , Perfilación de la Expresión Génica , Humanos , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Melanoma/genética , Análisis por Micromatrices , Invasividad Neoplásica , Mutación Puntual/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias Cutáneas/genética , Células Tumorales Cultivadas
15.
J Periodontal Res ; 46(3): 327-37, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21338358

RESUMEN

BACKGROUND AND OBJECTIVE: The antimicrobial peptide LL-37, derived from human neutrophils, can directly chemoattract leukocytes and up-regulate the expression of several immune-related genes in various cell types. In this study, we wanted to determine the immunoregulatory effect of LL-37 on interleukin-8 (IL-8) expression in human gingival fibroblasts (HGFs) and to characterize intracellular signaling pathway(s) and receptor(s) involved in IL-8 induction. MATERIAL AND METHODS: Cultured fibroblasts were treated with different concentrations of LL-37 or interleukin-1ß (IL-1ß), as a positive control, for specific periods of time in the presence or absence of various inhibitors. RT-PCR and real-time PCR were conducted to analyze the expression of IL-8 mRNA, and the IL-8 levels in cell-free culture media were measured using ELISAs. The MTT assay was performed to determine the cytotoxicity of LL-37. RESULTS: Nontoxic concentrations of LL-37 (up to 10 µm) and IL-1ß significantly up-regulated the expression of IL-8 mRNA in a dose-dependent manner (p < 0.05). The IL-8 protein levels were consistently significantly elevated in conditioned media of LL-37-treated HGFs (p < 0.05). IL-8 up-regulation by LL-37 was completely abrogated by 20 µm U0126, consistent with transient phosphorylation of p44/42 MAP kinases. Moreover, pretreatment with Brilliant Blue G (a selective antagonist of the P2X(7) receptor) and the neutralizing antibody against P2X(7) blocked IL-8 up-regulation in a dose-dependent manner, consistent with expression of the P2X(7) receptor in HGFs. CONCLUSION: These findings indicate that LL-37 induces IL-8 expression via the P2X(7) receptor and the MEK1/2-dependent p44/42 MAP kinases in HGFs, suggesting both direct and indirect involvement of LL-37 in neutrophil recruitment into an inflammatory site within diseased periodontal tissues.


Asunto(s)
Antibacterianos/farmacología , Catelicidinas/farmacología , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Interleucina-8/efectos de los fármacos , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Receptores Purinérgicos P2X7/fisiología , Regulación hacia Arriba/efectos de los fármacos , Anticuerpos Neutralizantes/farmacología , Péptidos Catiónicos Antimicrobianos , Butadienos/farmacología , Células Cultivadas , Medios de Cultivo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Encía/citología , Humanos , Interleucina-1beta/farmacología , Interleucina-8/metabolismo , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Infiltración Neutrófila/fisiología , Nitrilos/farmacología , Fosforilación , Antagonistas del Receptor Purinérgico P2X/farmacología , Colorantes de Rosanilina/farmacología
16.
J Leukoc Biol ; 88(6): 1191-200, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20837746

RESUMEN

IFN-ß and sIL-1Ra play crucial roles in the regulation of innate immunity and inflammation. IFN-ß, which is widely used to improve the course of relapsing, remitting multiple sclerosis, induces the production of sIL-1Ra in human monocytes through mechanisms that remain largely unknown. In this study, we identified PI3Kδ and MEK2 as key elements that control sIL-1Ra production in isolated human monocytes activated by IFN-ß. Blockade of MEK2, but not of MEK1, by inhibitors and siRNA prevented IFN-ß-induced PI3Kδ recruitment to the membrane, Akt phosphorylation, and sIL-1Ra production, suggesting that MEK2 acted upstream of PI3Kδ. Furthermore, ERK1/2, the only identified substrates of MEK1/2 to date, are dispensable for sIL-1Ra production in response to IFN-ß stimulation. Upon IFN-ß activation, MEK2 and PI3Kδ are translocated to monocyte membranes. These data suggest that MEK1 and MEK2 display different, nonredundant functions in IFN-ß signaling. That neither MEK1 nor ERK1/2 play a part in this mechanism is also an unexpected finding that gives rise to a better understanding of the MAPK signaling network. Together, these findings demonstrate that IFN-ß triggers an atypical MEK2/PI3Kδ signaling cascade to regulate sIL-1Ra expression in monocytes. The premise that MEK1 and MEK2 play a part in the induction of the proinflammatory cytokine, IL-1ß in human monocytes provides a rationale for an alternative, IFN-ß-mediated pathway to induce/enhance sIL-1Ra production and thus, to dampen inflammation.


Asunto(s)
Interferón beta/farmacología , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , MAP Quinasa Quinasa 2/fisiología , Monocitos/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/fisiología , Fosfatidilinositol 3-Quinasa Clase I , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , MAP Quinasa Quinasa 1/fisiología , Monocitos/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Oncol Rep ; 24(1): 251-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20514469

RESUMEN

The mitogen-activated protein kinase kinase 1/2 (MEK1/2) signalling pathway plays a central role in tumour progression. Small molecules that inhibit MEK1/2 are therefore considered attractive candidates for anti-cancer drugs. However, the exact contributions of MEK1 and MEK2 to the development of pancreatic cancer remain to be established. To differentiate the functions of MEK1 and MEK2 in a cultured pancreatic cancer cell line, we utilised shRNA-mediated knockdown of their two mRNAs individually. We studied the effects of MEK1 and MEK2 knockdown on cell morphology, proliferation, mitotic arrest, and in vitro invasion capability in PC-1.0 cells. The results showed that inhibition of MEK1 expression was an effective and specific approach to inhibit cell proliferation and induce G0/G1 arrest. On the other hand, MEK2 knockdown specially altered cell morphology and inhibited the invasive ability of pancreatic cancer cells. Therefore, MEK1 and MEK2 mediate different biological responses in cultured pancreatic cancer cells. These proteins could become distinct targets for the inhibition of specific cellular functions in the treatment of pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Neoplasias Pancreáticas/enzimología , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetinae , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Mesocricetus , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Interferente Pequeño/farmacología
18.
Aktuelle Urol ; 41(3): 178-83, 2010 May.
Artículo en Alemán | MEDLINE | ID: mdl-20446247

RESUMEN

PURPOSE: Obesity is considered to be a risk factor for prostate cancer. Mitogenic actions of leptin, an adipocyte-derived hormone in a variety of cancer cell types have been identified. We have investigated the proliferative effects of leptin on human prostate cancer cells and assessed the role of tyrosine kinase signalling in mediating these actions. MATERIALS AND METHODS: Two human androgen-resistant prostate cancer cell lines and one androgen-sensitive human prostate adenocarcinoma cell line were treated with leptin (5-100 ng / mL) for up to 48 hours. Under serum-free conditions, cell proliferation was measured using an enzyme-linked colorimetric assay. Furthermore, phosphorylation of a downstream component of MAPK (ERK1 / 2) was detected by Western blotting and a specific inhibitor of MAPK (PD98059; 40 microM) was used to evaluate the role of this signalling pathway. RESULTS: Leptin dose-dependently increased the cell number in both androgen-resistant cell lines after 24 h and 48 h of incubation (percent of control: DU145 = 194.6 +/- 5.9 %, PC-3 = 177.9 +/- 6.8 %; 100 ng / mL leptin; 48 h; p < 0.001). Conversely, leptin's proliferative effect on the androgen-sensitive cell line was less pronounced (percent of control: LNCaP = 112.3 +/- 6.1 %; 100 ng / mL leptin; 48 h). Leptin also caused dose-dependent ERK1 / 2 phosphorylation in both androgen-resistant cell lines. In addition, pre-treatment with PD98059 inhibited these responses and attenuated leptin's mitogenic action. CONCLUSIONS: Data from this in vitro study suggest an association between obesity-associated hyperleptinemia and an increased risk for prostate cancer. Further investigations are necessary to clarify whether these data have clinical relevance regarding the use as a prognostic marker for predicting the timing of the occurrence of androgen resistency.


Asunto(s)
Adenocarcinoma/fisiopatología , Adipoquinas/fisiología , Neoplasias Hormono-Dependientes/fisiopatología , Obesidad/fisiopatología , Neoplasias de la Próstata/fisiopatología , Proteínas Tirosina Quinasas/fisiología , Transducción de Señal/fisiología , Adenocarcinoma/patología , Western Blotting , División Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Humanos , Técnicas In Vitro , Leptina/fisiología , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Masculino , Neoplasias Hormono-Dependientes/patología , Obesidad/patología , Fosforilación , Neoplasias de la Próstata/patología , Factores de Riesgo
19.
Biochemistry ; 49(23): 4766-78, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20469933

RESUMEN

Translocator protein TSPO is an 18 kDa protein implicated in numerous cell functions and is highly expressed in secretory and glandular tissues, especially in steroidogenic cells. TSPO expression is altered in pathological conditions such as certain cancers and neurological diseases. In search of the factors regulating Tspo expression, we recently showed that high levels of TSPO in steroidogenic cells may be due to high constitutive expression of protein kinase Cepsilon (PKCepsilon), while phorbol 12-myristate 13-acetate (PMA) activation of PKCepsilon drives inducible TSPO expression in nonsteroidogenic cells, likely through activator protein 1 (AP1). In this study, we aimed to identify the signal transduction pathway through which PKCepsilon regulates Tspo gene expression. The MEK1/2 specific inhibitor U0126, but not NFkappaB inhibitors, reduced basal Tspo promoter activity in TSPO-rich steroidogenic cells (MA-10 Leydig), as well as basal and PMA-induced Tspo promoter levels in TSPO-poor nonsteroidogenic cells (NIH-3T3 fibroblasts). AP1 and signal transducer and activation of transcription 3 (STAT3) have binding sites in the Tspo promoter and are downstream targets of PKCepsilon and MAPK (Raf-1-ERK1/2) pathways. PKCepsilon overexpression induced STAT3 phosphorylation in NIH-3T3 cells, while PKCepsilon knockdown reduced STAT3 and c-Jun phosphorylation in Leydig cells. MEK1/2, ERK2, c-Jun, and STAT3 knockdown reduced Tspo mRNA and protein levels in Leydig cells. Additionally, Raf-1 reduced Tspo mRNA levels in the same cells. MEK1/2, c-Jun, and STAT3 knockdown also reduced basal as well as PMA-induced Tspo mRNA levels in NIH-3T3 cells. Together, these results demonstrate that PKCepsilon regulates Tspo gene expression through a MAPK (Raf-1-MEK1/2-ERK1/2) signal transduction pathway, acting at least in part through c-Jun and STAT3 transcription factors.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa C-epsilon/fisiología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptores de GABA/biosíntesis , Receptores de GABA/genética , Factor de Transcripción STAT3/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Marcación de Gen/métodos , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Receptores de GABA/deficiencia , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/fisiología
20.
Mol Cancer Ther ; 9(3): 606-16, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20179161

RESUMEN

The HER-2 oncogene, a member of the erythroblastosis oncogene B (ERBB)-like oncogene family, has been shown to be amplified in many types of cancer, including breast cancer. However, the molecular mechanism of HER-2 overexpression is not completely understood. The phosphorylation of proteins on the serine or threonine residues that immediately precede proline (pSer/Thr-Pro) is specifically catalyzed by the prolyl isomerase Pin1 and is a key signaling mechanism in cell proliferation and transformation. Here, we found that Pin1 interacts with mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) protein kinase 1, resulting in the induction of HER-2 expression. Pin1(-/-) mouse embryonic fibroblasts exhibited a decrease in epidermal growth factor (EGF)-induced MEK1/2 phosphorylation compared with Pin1(+/+) mouse embryonic fibroblast. In addition, a knockdown of Pin1 resulted in the inhibition of MEK1/2 phosphorylation induced by EGF in MCF-7 cells. Furthermore, PD98059, a specific inhibitor of MEK1/2, and Juglone, a potent Pin1 inhibitor, markedly suppressed the expression of activator protein-2alpha and the HER-2 promoter activity induced by EGF or 12-O-tetradecanoylphorbol-13-acetate in MCF-7 cells. Importantly, these inhibitors inhibited the neoplastic cell transformation induced by EGF in Pin1-overexpressing JB6 Cl41 cells, which showed enhanced cellular formation compared with the control cells. Therefore, Juglone and PD98059 inhibited the colony formation of MCF-7 breast cancer cells in soft agar. These results indicate that Pin1 amplifies EGF signaling in breast cancer cells through its interaction with MEK1 and then enhances HER-2 expression, suggesting that Pin1 plays an important role in the overexpression of HER-2 through Pin1-MEK1-activator protein-2alpha signaling in breast cancer.


Asunto(s)
Transformación Celular Neoplásica , MAP Quinasa Quinasa 1/metabolismo , Isomerasa de Peptidilprolil/fisiología , Receptor ErbB-2/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Factor de Crecimiento Epidérmico/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/metabolismo , MAP Quinasa Quinasa 2/fisiología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , Fosforilación , Unión Proteica/fisiología , Receptor ErbB-2/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...