Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
J Hematol Oncol ; 13(1): 113, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807225

RESUMEN

Cancer is characterized as a complex disease caused by coordinated alterations of multiple signaling pathways. The Ras/RAF/MEK/ERK (MAPK) signaling is one of the best-defined pathways in cancer biology, and its hyperactivation is responsible for over 40% human cancer cases. To drive carcinogenesis, this signaling promotes cellular overgrowth by turning on proliferative genes, and simultaneously enables cells to overcome metabolic stress by inhibiting AMPK signaling, a key singular node of cellular metabolism. Recent studies have shown that AMPK signaling can also reversibly regulate hyperactive MAPK signaling in cancer cells by phosphorylating its key components, RAF/KSR family kinases, which affects not only carcinogenesis but also the outcomes of targeted cancer therapies against the MAPK signaling. In this review, we will summarize the current proceedings of how MAPK-AMPK signalings interplay with each other in cancer biology, as well as its implications in clinic cancer treatment with MAPK inhibition and AMPK modulators, and discuss the exploitation of combinatory therapies targeting both MAPK and AMPK as a novel therapeutic intervention.


Asunto(s)
Adenilato Quinasa/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Terapia Molecular Dirigida , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Aminoácidos/metabolismo , Antineoplásicos/uso terapéutico , Autofagia , Diferenciación Celular/fisiología , División Celular/fisiología , Ensayos Clínicos como Asunto , Sinergismo Farmacológico , Metabolismo Energético , Activación Enzimática , Homeostasis , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Proteínas Supresoras de Tumor/fisiología , Quinasas raf/antagonistas & inhibidores , Quinasas raf/genética , Quinasas raf/fisiología
2.
Hepatology ; 71(3): 929-942, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31692012

RESUMEN

BACKGROUND AND AIMS: Aristolochic acid (AA) exposure has been statistically associated with human liver cancers. However, direct evidence of AA exposure-induced liver cancer is absent. This study aims to establish a direct causal relationship between AA exposure and liver cancers based on a mouse model and then explores the AA-mediated genomic alterations that could be implicated in human cancers with AA-associated mutational signature. APPROACH AND RESULTS: We subjected mice, including phosphatase and tensin homolog (Pten)-deficient ones, to aristolochic acid I (AAI) alone or a combination of AAI and CCl4 . Significantly, AAI exposure induced mouse liver cancers, including hepatocellular carcinoma (HCC) and combined HCC and intrahepatic cholangiocarcinoma, in a dose-dependent manner. Moreover, AAI exposure also enhanced tumorigenesis in these CCl4 -treated or Pten-deficient mice. AAI led to DNA damage and AAI-DNA adduct that could initiate liver cancers through characteristic adenine-to-thymine transversions, as indicated by comprehensive genomic analysis, which revealed recurrent mutations in Harvey rat sarcoma virus oncogene. Interestingly, an AA-associated mutational signature was mainly implicated in human liver cancers, especially from China. Moreover, we detected the AAI-DNA adduct in 25.8% (16/62) of paratumor liver tissues from randomly selected Chinese patients with HCC. Furthermore, based on phylogenetic analysis, the characteristic mutations were found in the initiating malignant clones in the AA-implicated mouse and human liver cancers where the mutations of tumor protein p53 and Janus kinase 1 were prone to be significantly enriched in the AA-affected human tumors. CONCLUSIONS: This study provides evidence for AA-induced liver cancer with the featured mutational processes during malignant clonal evolution, laying a solid foundation for the prevention and diagnosis of AA-associated human cancers, especially liver cancers.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Mutación , Animales , Neoplasias de los Conductos Biliares/inducido químicamente , Neoplasias de los Conductos Biliares/genética , Tetracloruro de Carbono/toxicidad , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Colangiocarcinoma/inducido químicamente , Colangiocarcinoma/genética , Daño del ADN , Relación Dosis-Respuesta a Droga , Humanos , Janus Quinasa 1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/genética , Quinasas raf/fisiología
3.
Br J Cancer ; 121(4): 332-339, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31312030

RESUMEN

BACKGROUND: A phase Ib study of binimetinib and capecitabine for gemcitabine-pretreated biliary tract cancer (BTC) patients was conducted. METHODS: Binimetinib and capecitabine were dosed twice daily on days 1-14, in 3-week cycles. In the dose-escalation (DE) part, three dose levels (DL) were tested (DL1: binimetinib/capecitabine, 15 mg/1000 mg/m2; DL2: 30 mg/1000 mg/m2; DL3: 30 mg/1250 mg/m2). RESULTS: In the DE part, nine patients were recruited and no dose-limiting toxicity was noted. Therefore, the recommended phase 2 dose was determined as DL3. In the expansion part, 25 patients were enrolled. In total, 34 patients, 25 (73.5%) and 9 patients (26.5%) were second-line and third-line settings, respectively. The 3-month progression-free survival (PFS) rate was 64.0%, and the median PFS and overall survival (OS) were 4.1 and 7.8 months. The objective response rate and disease control rate were 20.6% and 76.5%. In total, 68.4% of stable diseases were durable (> 12 weeks). Furthermore, patients with RAS/RAF/MEK/ERK pathway mutations (38.5%) showed significantly better tumour response (p = 0.028), PFS (5.4 vs. 3.5 months, p = 0.010) and OS (10.8 vs. 5.9 months, p = 0.160) than wild type. Most of the adverse events were grade 1/2 and manageable. CONCLUSIONS: A combination of binimetinib and capecitabine shows acceptable tolerability and promising antitumor efficacy for gemcitabine-pretreated BTC, especially in patients with RAS/RAF/MEK/ERK pathway mutations. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov (Identifier: NCT02773459).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Sistema Biliar/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación , Quinasas raf/genética , Proteínas ras/genética , Anciano , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/mortalidad , Neoplasias del Sistema Biliar/psicología , Capecitabina/administración & dosificación , Capecitabina/efectos adversos , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Calidad de Vida , Transducción de Señal , Quinasas raf/fisiología , Proteínas ras/fisiología
4.
Nucleic Acids Res ; 45(18): 10518-10533, 2017 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-29048592

RESUMEN

The yeast 2-µm plasmid is a remarkable genetic parasite, managing efficient maintenance at high-copy number with minimal impact on the host. Equal partitioning of the plasmid upon host cell division requires plasmid proteins Rep1 and Rep2 and the plasmid STB locus. The Rep proteins and the plasmid-encoded Raf protein also regulate plasmid gene transcription. In this study, protein interaction assays, sequence analyses and mutational approaches were used to identify domains and residues in Rep2 and Raf required for association with Rep1 and Rep2 and to delineate the Rep2 DNA-binding domain. Rep2 and Raf displayed similarities in interactions with Rep1 and Rep2, in having Rep1 promote their STB association in vivo, and in stabilizing Rep protein levels. Rep2 mutants impaired for self-association were competent for transcriptional repression while those deficient for Rep1 association were not. Surprisingly, Rep2 mutants impaired for either Rep1 interaction or self-association were able to maintain efficient plasmid inheritance provided Raf was present and competent for Rep protein interaction. Our findings provide insight into the Rep protein complexes required for partitioning and transcriptional repression, and suggest that in addition to its transcriptional function, Raf stabilization of Rep partitioning proteins contributes to the remarkable persistence of the 2-µm plasmid.


Asunto(s)
Plásmidos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Transactivadores/metabolismo , Quinasas raf/metabolismo , Quinasas raf/fisiología , División Celular , Patrón de Herencia , Organismos Modificados Genéticamente , Unión Proteica , Estabilidad Proteica
5.
J Thorac Oncol ; 11(8): 1233-1241, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27133774

RESUMEN

The activity of the RAF/MEK/ERK signaling pathway is critical for the proliferation of normal and cancerous cells. Oncogenic mutations driving the development of lung adenocarcinoma often activate this signaling pathway. In contrast, pathway activity levels and their biological roles are not well established in small cell lung cancer (SCLC), a fast-growing neuroendocrine lung cancer subtype. Here we discuss the function of the RAF/MEK/ERK kinase pathway and the mechanisms leading to its activation in SCLC cells. In particular, we argue that activation of this pathway may be beneficial to the survival, proliferation, and spread of SCLC cells in response to multiple stimuli. We also consider evidence that high levels of RAF/MEK/ERK pathway activity may be detrimental to SCLC tumors, including in part by interfering with their neuroendocrine fate. On the basis of these observations, we examined when small molecules targeting kinases in the RAF/MEK/ERK pathway may be useful therapeutically in patients with SCLC, including in combination with other therapeutic agents.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Quinasas raf/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Humanos , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Carcinoma Pulmonar de Células Pequeñas/patología , Quinasas raf/fisiología
6.
Biochim Biophys Acta ; 1863(4): 770-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26844774

RESUMEN

Melanoma has markedly increased worldwide during the past several decades in the Caucasian population and is responsible for 80% of skin cancer deaths. Considering that metastatic melanoma is almost completely resistant to most current therapies and is linked with a poor patient prognosis, it is crucial to further investigate potential molecular targets. Major cell-autonomous drivers in the pathogenesis of this disease include the classical MAPK (i.e., RAS-RAF-MEK-ERK), WNT, and PI3K signaling pathways. These pathways play a major role in defining the progression of melanoma, and some have been the subject of recent pharmacological strategies to treat this belligerent disease. This review describes the latest advances in the understanding of melanoma progression and the major molecular pathways involved. In addition, we discuss the roles of emerging molecular players that are involved in melanoma pathogenesis, including the functional role of the melanoma tumor antigen, p97/MFI2 (melanotransferrin).


Asunto(s)
Melanoma/genética , Melanoma/patología , Oncogenes/fisiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Animales , Progresión de la Enfermedad , Genes ras/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Vía de Señalización Wnt/fisiología , Quinasas raf/fisiología
7.
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
8.
J Hepatol ; 63(3): 651-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25865556

RESUMEN

BACKGROUND & AIMS: We have previously reported that Shp2, a tyrosine phosphatase previously known as a pro-leukemogenic molecule, suppresses the initiation of hepatocellular carcinoma (HCC). However, the role of Shp2 in HCC progression remains obscure. METHODS: Shp2 expression was determined in human HCC using real-time PCR, immunoblotting and immunohistochemistry. Clinical significance of Shp2 expression was analyzed in 301 HCC tissues with clinico-pathological characteristics and follow-up information. Short hairpin RNA was utilized to investigate the function of Shp2 in hepatoma cell behavior. Role of Shp2 in HCC progression was monitored through nude mice xenograft assay. Kinase activity assay and co-immunoprecipitation were used for mechanism analysis. RESULTS: Elevated expression of Shp2 was detected in 65.9% (394/598) of human HCCs, and its levels were even higher in metastasized foci. Overexpression of Shp2 correlated well with the malignant clinico-pathological characteristics of HCC and predicted the poor prognosis of patients. Interference of Shp2 expression suppressed the proliferation of hepatoma cells in vitro and inhibited the growth of HCC xenografts in vivo. Down-regulation of Shp2 attenuated the adhesion and migration of hepatoma cells and diminished metastasized HCC formation in mice. Our data demonstrated that Shp2 promotes HCC growth and metastasis by coordinately activating Ras/Raf/Erk pathway and PI3-K/Akt/mTOR cascade. Moreover, down-regulation of Shp2 enhanced the sensitivity of hepatoma cells upon sorafenib treatment, and patients with low Shp2 expression exhibited superior prognosis to sorafenib. CONCLUSIONS: Shp2 promotes the progression of HCC and may serve as a prognostic biomarker for patients.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Animales , Carcinoma Hepatocelular/mortalidad , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Humanos , Neoplasias Hepáticas/mortalidad , Sistema de Señalización de MAP Quinasas , Ratones , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Pronóstico , Sorafenib , Quinasas raf/fisiología
10.
Leuk Res ; 38(7): 795-803, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24811390

RESUMEN

Delocalized lipophilic cation dequalinium (DQA) selectively accumulates in mitochondria and displays anticancer activity in different malignancies. Our previous studies indicate a DQA-induced cytotoxicity in human acute promyelocytic leukemia NB4 cells by early disturbance in mitochondrial function and oxidative stress. This study shows the ability of DQA to downregulate Raf/MEK/ERK1/2 and PI3K/Akt signaling pathways in NB4 cells which leads to cell death by apoptosis and/or necrosis. Moreover, DQA potentiates the action of specific inhibitors of these pathways. These DQA effects could be mediated by redox regulation of Akt. Our results contribute to a better understanding of the cytotoxic DQA mechanism on leukemia cells and encourage the performance of further studies in combination with other agents such as kinase inhibitors for improving the efficacy of therapies against acute promyelocytic leukemia.


Asunto(s)
Decualinio/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Leucemia/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Quinasas raf/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Glutatión/metabolismo , Humanos , Leucemia/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Quinasas raf/fisiología
11.
Br J Cancer ; 110(10): 2479-88, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24762959

RESUMEN

BACKGROUND: Mesothelioma is a notoriously chemotherapy-resistant neoplasm, as is evident in the dismal overall survival for patients with those of asbestos-associated disease. We previously demonstrated co-activation of multiple receptor tyrosine kinases (RTKs), including epidermal growth factor receptor (EGFR), MET, and AXL in mesothelioma cell lines, suggesting that these kinases could serve as novel therapeutic targets. Although clinical trials have not shown activity for EGFR inhibitors in mesothelioma, concurrent inhibition of various activated RTKs has pro-apoptotic and anti-proliferative effects in mesothelioma cell lines. Thus, we hypothesised that a coordinated network of multi-RTK activation contributes to mesothelioma tumorigenesis. METHODS: Activation of PI3K/AKT/mTOR, Raf/MAPK, and co-activation of RTKs were evaluated in mesotheliomas. Effects of RTK and downstream inhibitors/shRNAs were assessed by measuring mesothelioma cell viability/growth, apoptosis, activation of signalling intermediates, expression of cell-cycle checkpoints, and cell-cycle alterations. RESULTS: We demonstrate activation of the PI3K/AKT/p70S6K and RAF/MEK/MAPK pathways in mesothelioma, but not in non-neoplastic mesothelial cells. The AKT activation, but not MAPK activation, was dependent on coordinated activation of RTKs EGFR, MET, and AXL. In addition, PI3K/AKT/mTOR pathway inhibition recapitulated the anti-proliferative effects of concurrent inhibition of EGFR, MET, and AXL. Dual targeting of PI3K/mTOR by BEZ235 or a combination of RAD001 and AKT knockdown had a greater effect on mesothelioma proliferation and viability than inhibition of individual activated RTKs or downstream signalling intermediates. Inhibition of PI3K/AKT was also associated with MDM2-p53 cell-cycle regulation. CONCLUSIONS: These findings show that PI3K/AKT/mTOR is a crucial survival pathway downstream of multiple activated RTKs in mesothelioma, underscoring that PI3K/mTOR is a compelling target for therapeutic intervention.


Asunto(s)
Antineoplásicos/farmacología , Mesotelioma/enzimología , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Butadienos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cromonas/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Everolimus , Humanos , Imidazoles/farmacología , Indazoles/farmacología , Sistema de Señalización de MAP Quinasas , Mesotelioma/patología , Terapia Molecular Dirigida , Morfolinas/farmacología , Proteínas de Neoplasias/fisiología , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Quinolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Tirosina Quinasas Receptoras/fisiología , Sirolimus/análogos & derivados , Sirolimus/farmacología , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/fisiología , Quinasas raf/fisiología
12.
Cancer Res ; 73(23): 6926-37, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24129679

RESUMEN

RAF inhibitors achieve unprecedented but mainly transient clinical responses in patients with melanoma whose tumors harbor an activating BRAF mutation. One notable side-effect of RAF inhibitors is the stimulation of cutaneous skin tumors, arising in about 30% of patients receiving these drugs, which are thought to develop as a result of inhibitor-induced activation of wild-type Raf in occult precursor skin lesions. This effect raises the possibility that less manageable tumors might also arise in other epithelial tissues. Here we provide preclinical evidence supporting this disquieting hypothesis by showing that the RAF inhibitors PLX-4032 (vemurafenib) and GDC-0879 precipitate the development of cell-autonomous, Ras-driven tumors in skin and gastric epithelia. The magnitude of the effects correlated with the inhibitors' relative abilities to induce ERK activation. Epidermis-restricted ablation of either B-Raf or C-Raf prevented PLX-4032-induced ERK activation and tumorigenesis. In contrast, GDC-0879 induced ERK activation and tumorigenesis in B-Raf-deficient epidermis, whereas C-Raf ablation blocked GDC-0879-induced tumorigenesis (despite strong ERK activation) by preventing Rokα-mediated keratinocyte dedifferentiation. Thus, inhibitor-induced ERK activation did not require a specific Raf kinase. ERK activation was necessary, but not sufficient for Ras + Raf inhibitor-induced tumorigenesis, whereas C-Raf downregulation of Rokα was essential even in the face of sustained ERK signaling to prevent differentiation and promote tumorigenesis. Taken together, our findings suggest that combination therapies targeting ERK-dependent and -independent functions of Raf may be more efficient but also safer for cancer treatment.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Melanoma/inducido químicamente , Inhibidores de Proteínas Quinasas/efectos adversos , Neoplasias Cutáneas/inducido químicamente , Quinasas raf/antagonistas & inhibidores , Quinasas raf/fisiología , Animales , Células COS , Chlorocebus aethiops , Humanos , Indenos/farmacología , Indoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Transgénicos , Pirazoles/farmacología , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Vemurafenib
13.
Behav Brain Res ; 253: 280-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23911427

RESUMEN

Activation of PKCɛ, an abundant and developmentally regulated PKC isoform in the brain, has been implicated in memory throughout life and across species. Yet, direct evidence for a mechanistic role for PKCɛ in memory is still lacking. Hence, we sought to evaluate this in rats, using short-term treatments with two PKCɛ-selective peptides, the inhibitory ɛV1-2 and the activating ψɛRACK, and the novel object recognition task (NORT). Our results show that the PKCɛ-selective activator ψɛRACK, did not have a significant effect on recognition memory. In the short time frames used, however, inhibition of PKCɛ activation with the peptide inhibitor ɛV1-2 significantly impaired recognition memory. Moreover, when we addressed at the molecular level the immediate proximal signalling events of PKCɛ activation in acutely dissected rat hippocampi, we found that ψɛRACK increased in a time-dependent manner phosphorylation of MARCKS and activation of Src, Raf, and finally ERK1/2, whereas ɛV1-2 inhibited all basal activity of this pathway. Taken together, these findings present the first direct evidence that PKCɛ activation is an essential molecular component of recognition memory and point toward the use of systemically administered PKCɛ-regulating peptides as memory study tools and putative therapeutic agents.


Asunto(s)
Memoria/fisiología , Proteína Quinasa C-epsilon/metabolismo , Reconocimiento en Psicología/fisiología , Animales , Western Blotting , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Productos del Gen tat/farmacología , Hipocampo/citología , Hipocampo/enzimología , Hipocampo/metabolismo , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Neuronas/enzimología , Fosforilación , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Wistar , Reconocimiento en Psicología/efectos de los fármacos , Quinasas raf/fisiología , Familia-src Quinasas/fisiología
14.
Biochem Pharmacol ; 85(7): 1007-17, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23396089

RESUMEN

Fenretinide is significantly more effective in inducing apoptosis in cancer cells than all-trans retinoic acid (ATRA). The current study uses a genome-wide approach to understand the differential role fenretinide and ATRA have in inducing apoptosis in Huh7 cells. Fenretinide and ATRA-induced gene expressions and DNA bindings were profiled using microarray and chromatin immunoprecipitation with anti-RXRα antibody. The data showed that fenretinide was not a strong transcription regulator. Fenretinide only changed the expressions of 1 093 genes, approximately three times less than the number of genes regulated by ATRA (2 811). Biological function annotation demonstrated that both fenretinide and ATRA participated in pathways that determine cell fate and metabolic processes. However, fenretinide specifically induced Fas/TNFα-mediated apoptosis by increasing the expression of pro-apoptotic genes i.e., DEDD2, CASP8, CASP4, and HSPA1A/B; whereas, ATRA induced the expression of BIRC3 and TNFAIP3, which inhibit apoptosis by interacting with TRAF2. In addition, fenretinide inhibited the expression of the genes involved in RAS/RAF/ERK-mediated survival pathway. In contrast, ATRA increased the expression of SOSC2, BRAF, MEK, and ERK genes. Most genes regulated by fenretinide and ATRA were bound by RXRα, suggesting a direct effect. This study revealed that by regulating fewer genes, the effects of fenretinide become more specific and thus has fewer side effects than ATRA. The data also suggested that fenretinide induces apoptosis via death receptor effector and by inhibiting the RAS/RAF/ERK pathway. It provides insight on how retinoid efficacy can be improved and how side effects in cancer therapy can be reduced.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , ADN/metabolismo , Fenretinida/farmacología , Neoplasias Hepáticas/patología , Transcriptoma , Tretinoina/farmacología , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Receptor alfa X Retinoide/metabolismo , Transducción de Señal , Quinasas raf/fisiología , Proteínas ras/fisiología
16.
J Gastroenterol ; 48(3): 350-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23015305

RESUMEN

BACKGROUND: Galectin-3 has been independently correlated with malignant behavior in human colon cancer. The involvement of galectin-3 in the invasiveness of colon cancer cells remains to be determined. We investigated whether galectin-3 was involved in the colon cancer cell migration mediated by certain kinase pathways. METHODS: We studied 2 colon cancer cell lines (DLD-1 and Caco2) and clinical samples. Immunostaining and Western blotting were used to analyze the expression of galectin-3 in vitro and in the clinical samples. Short hairpin RNA and overexpression of galectin-3 were used to study loss- and gain-of-function in a wound-healing assay and a Transwell migration assay, and Western blotting was used to study the Ras-Raf signaling pathway. RESULTS: Galectin-3 was expressed at lower levels in DLD-1 than in Caco2 cells. The lower galectin-3 level in DLD-1 cells was associated with decreased cell migration, in comparison with that of Caco2 cells. Overexpression of galectin-3 increased the migration rate of DLD-1, while knockdown of galectin-3 decreased the migration. Overexpression of galectin-3 was correlated with increased lamellipodia formation and distal lung localization in a mouse model. The galectin-3 enhancement of DLD-1 cell migration was mediated by K-Ras, Raf and Erk1/2 pathway activation, but not the H-Ras, p38, or JNK activation. CONCLUSIONS: Galectin-3 plays an important role in regulating colon cancer cell migration and potential distal localization. The galectin-3 enhancement of cell migration is mediated through the K-Ras-Raf-Erk1/2 pathway. Specific targeting of the K-Ras-Raf-Erk1/2 pathway may be useful for treating colon cancers associated with increased galectin-3 expression.


Asunto(s)
Neoplasias del Colon/metabolismo , Galectina 3/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Células CACO-2 , Movimiento Celular/fisiología , Neoplasias del Colon/patología , Femenino , Galectina 3/biosíntesis , Galectina 3/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas p21(ras)/fisiología , ARN Interferente Pequeño/genética , Trasplante Heterólogo , Células Tumorales Cultivadas , Quinasas raf/fisiología , Proteínas ras/fisiología
17.
Carcinogenesis ; 33(12): 2467-76, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23027619

RESUMEN

Interleukin-11 (IL-11), which belongs to a class of IL6-type cytokines, plays an important role in inflammation, motility and invasion in cancer. The ras mutation is frequently found in human cancer, but little is known regarding the transcriptional activation of the IL-11 gene by the Ras signal pathway in tumour cells. In this study, we investigated the role of Ras in the regulation of IL-11 using two different cell model systems: mouse NIH3T3 cells over-expressing oncogenic Ras with a tet-on system and Capan-1 human pancreatic carcinoma cells harbouring a K-ras mutation. We found that IL-11 expression was up-regulated at the transcriptional level by oncogenic Ras. Activation of the AP-1 response element, located between -153 and -30 in the 5'-regulatory region of the IL-11 gene, was necessary for oncogenic Ras-induced IL-11 promoter activation. AP-1 proteins, including Fra-1 and Fra-2, were up-regulated through the Raf/MEK and phosphatidylinositol 3-kinase (PI3K)/Akt pathways by oncogenic Ras. Knockdown of Fra-1 by siRNA in NIH3T3 or Capan-1 cells strongly attenuated oncogenic Ras-induced IL-11 expression. Additionally, inhibition of JNK, p38 and Stat3 abrogated oncogenic Ras-induced IL-11 expression. These results suggest that both the PI3K and Raf pathways are necessary for the expression of IL-11 in oncogenic Ras-mutated cells, and that JNK, p38 and Stat3 also contribute to oncogenic Ras-induced IL-11 expression.


Asunto(s)
Regulación de la Expresión Génica , Genes ras/fisiología , Interleucina-11/genética , Animales , Células HEK293 , Humanos , Ratones , Mutación , Células 3T3 NIH , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/fisiología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/fisiología , Factor de Transcripción AP-1/fisiología , Transcripción Genética , Quinasas raf/fisiología
19.
Am J Pathol ; 180(6): 2462-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546478

RESUMEN

Lesions displaying a variety of dysplastic changes precede invasive oral and epidermal squamous cell carcinoma (SCC); however, there are no histopathological criteria for either confirming or staging premalignancy. SCCs and dysplasias frequently contain cells that abnormally express the γ2 subunit of laminin-332. We developed cell culture models to investigate γ2 dysregulation. Normal human keratinocytes displayed density-dependent repression of γ2, whereas premalignant keratinocytes and SCC cells overexpressed γ2 and secreted laminin assembly intermediates. Neoplastic cells had hyperactive EGFR/MAPK(ERK) signaling coordinate with overexpressed γ2, and EGFR and MEK inhibitors normalized γ2 expression. Keratinocytes engineered to express HPV16 E6 or activated mutant HRAS, cRAF1, or MEK1 lost density repression of γ2 and shared with neoplastic cells signaling abnormalities downstream of ERK, including increased phosphorylation of S6 and eIF4 translation factors. Notably, qPCR results revealed that γ2 overexpression was not accompanied by increased γ2 mRNA levels, consistent with ERK-dependent, eIF4B-mediated translation initiation of the stem-looped, 5'-untranslated region of γ2 mRNA in neoplastic cells. Inhibitors of MEK, but not of TORC1/2, blocked S6 and eIF4B phosphorylation and γ2 overexpression. Immunostaining of oral dysplasias identified γ2 overexpression occurring within fields of basal cells that had elevated p-S6 levels. These results reveal a causal relationship between ERK-dependent translation factor activation and laminin γ2 dysregulation and identify new markers of preinvasive neoplastic change during progression to SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Laminina/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neoplasias de la Boca/metabolismo , Lesiones Precancerosas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Queratinocitos/metabolismo , Laminina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Modificación Traduccional de las Proteínas/fisiología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Quinasas raf/fisiología , Proteínas ras/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA