Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
1.
J Extracell Vesicles ; 10(8): e12091, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34136107

RESUMEN

Oncogenic RAS impacts communication between cancer cells and their microenvironment, but it is unclear how this process influences cellular interactions with extracellular vesicles (EVs). This is important as intercellular EV trafficking plays a key role in cancer invasion and metastasis. Here we report that overexpression of mutant RAS drives the EV internalization switch from endocytosis (in non-transformed cells) to macropinocytosis (in cancer cells) resulting in enhanced EV uptake. This process depends on the surface proteoglycan, fibronectin and EV engulfment mechanism regulated by CRAF. Both mutant RAS and activated CRAF expression is associated with formation of membrane ruffles to which they colocalize along with actin, sodium-hydrogen exchangers (NHEs) and phosphorylated myosin phosphatase (pMYPT). RAS-transformed cells internalize EVs in the vicinity of ruffled structures followed by apparent trafficking to lysosome and degradation. NHE inhibitor (EIPA) suppresses RAS-driven EV uptake, along with adhesion-independent clonal growth and experimental metastasis in mice. Thus, EV uptake may represent a targetable step in progression of RAS-driven cancers.


Asunto(s)
Vesículas Extracelulares/metabolismo , Metástasis de la Neoplasia/fisiopatología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Transporte Biológico/fisiología , Comunicación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Endocitosis/fisiología , Vesículas Extracelulares/fisiología , Genes ras , Humanos , Ratones , Ratones SCID , Procesos Neoplásicos , Pinocitosis/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Microambiente Tumoral/fisiología , Proteínas ras/metabolismo , Proteínas ras/fisiología
2.
J Neurosci ; 40(34): 6522-6535, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32690613

RESUMEN

Chronic pain caused by spinal cord injury (SCI) is notoriously resistant to treatment, particularly by opioids. After SCI, DRG neurons show hyperactivity and chronic depolarization of resting membrane potential (RMP) that is maintained by cAMP signaling through PKA and EPAC. Importantly, SCI also reduces the negative regulation by Gαi of adenylyl cyclase and its production of cAMP, independent of alterations in G protein-coupled receptors and/or G proteins. Opioid reduction of pain depends on coupling of opioid receptors to Gαi/o family members. Combining high-content imaging and cluster analysis, we show that in male rats SCI decreases opioid responsiveness in vitro within a specific subset of small-diameter nociceptors that bind isolectin B4. This SCI effect is mimicked in nociceptors from naive animals by a modest 5 min depolarization of RMP (15 mm K+; -45 mV), reducing inhibition of cAMP signaling by µ-opioid receptor agonists DAMGO and morphine. Disinhibition and activation of C-Raf by depolarization-dependent phosphorylation are central to these effects. Expression of an activated C-Raf reduces sensitivity of adenylyl cyclase to opioids in nonexcitable HEK293 cells, whereas inhibition of C-Raf or treatment with the hyperpolarizing drug retigabine restores opioid responsiveness and blocks spontaneous activity of nociceptors after SCI. Inhibition of ERK downstream of C-Raf also blocks SCI-induced hyperexcitability and depolarization, without direct effects on opioid responsiveness. Thus, depolarization-dependent C-Raf and downstream ERK activity maintain a depolarized RMP and nociceptor hyperactivity after SCI, providing a self-reinforcing mechanism to persistently promote nociceptor hyperexcitability and limit the therapeutic effectiveness of opioids.SIGNIFICANCE STATEMENT Chronic pain induced by spinal cord injury (SCI) is often permanent and debilitating, and usually refractory to treatment with analgesics, including opioids. SCI-induced pain in a rat model has been shown to depend on persistent hyperactivity in primary nociceptors (injury-detecting sensory neurons), associated with a decrease in the sensitivity of adenylyl cyclase production of cAMP to inhibitory Gαi proteins in DRGs. This study shows that SCI and one consequence of SCI (chronic depolarization of resting membrane potential) decrease sensitivity to opioid-mediated inhibition of cAMP and promote hyperactivity of nociceptors by enhancing C-Raf activity. ERK activation downstream of C-Raf is necessary for maintaining ongoing depolarization and hyperactivity, demonstrating an unexpected positive feedback loop to persistently promote pain.


Asunto(s)
Dolor Crónico/fisiopatología , Nociceptores/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Receptores Opioides mu/fisiología , Transducción de Señal , Traumatismos de la Médula Espinal/fisiopatología , Animales , Células Cultivadas , Dolor Crónico/complicaciones , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Traumatismos de la Médula Espinal/complicaciones
3.
Dev Biol ; 461(2): 172-183, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32061885

RESUMEN

Ras1 (Ras85D) and Ras2 (Ras64B) are the Drosophila orthologs of human H-Ras/N-Ras/K-Ras and R-Ras1-3 genes, respectively. The function of Ras1 has been thoroughly characterised during Drosophila embryonic and imaginal development, and it is associated with coupling activated trans-membrane receptors with tyrosine kinase activity to their downstream effectors. In this capacity, Ras1 binds and is required for the activation of Raf. Ras1 can also interact with PI3K, and it is needed to achieve maximal levels of PI3K signalling in specific cellular settings. In contrast, the function of the unique Drosophila R-Ras member (Ras2/Ras64B), which is more closely related to vertebrate R-Ras2/TC21, has been only studied through the use of constitutively activated forms of the protein. This pioneering work identified a variety of phenotypes that were related to those displayed by Ras1, suggesting that Ras1 and Ras2 might have overlapping activities. Here we find that Ras2 can interact with PI3K and Raf and activate their downstream effectors Akt and Erk. However, and in contrast to mutants in Ras1, which are lethal, null alleles of Ras2 are viable in homozygosis and only show a phenotype of reduced wing size and extended life span that might be related to reduced Insulin receptor signalling.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Insulina/fisiología , Proteínas de la Membrana/fisiología , Proteínas ras/fisiología , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Receptores ErbB , Femenino , Edición Génica , Estudios de Asociación Genética , Longevidad/genética , Masculino , Proteínas de la Membrana/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Péptidos de Invertebrados , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología , Alas de Animales/crecimiento & desarrollo , Alas de Animales/ultraestructura , Proteínas ras/genética
4.
Sci Rep ; 10(1): 824, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31965002

RESUMEN

Protein modifications by phosphorylation or ubiquitylation have been selected throughout evolution as efficient regulatory mechanisms of cellular processes. Cell migration is a complex, highly coordinated process where these mechanisms must participate in an integrated manner to transmit signaling during migration. In this study, we show that the ubiquitin ligase HERC1 regulates the p38 signaling pathway, and that this regulation is mediated by the MAPK kinase MKK3. Moreover, we demonstrate a crosstalk between RAF and MKK3/p38 pathways where RAF acts upstream of MKK3. Mechanistically, HERC1 regulates the protein levels of C-RAF and MKK3. Thus, HERC1 ubiquitylates C-RAF, targeting it for proteasomal degradation, and RAF proteins regulate MKK3 mRNA levels. Accordingly, HERC1 knockdown induces C-RAF stabilization and activation of RAF proteins; in turn, this activation increases MKK3, which phosphorylates and activates p38. The importance of these observations is demonstrated by HERC1 regulation of cell migration through regulation of p38 signaling via a RAF-dependent mechanism. Thus, HERC1 plays an essential role as a regulator of crosstalk between RAF/MKK3/p38 signaling pathways during cell migration.


Asunto(s)
Movimiento Celular/genética , Regulación de la Expresión Génica/genética , MAP Quinasa Quinasa 3/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal/genética , Ubiquitina-Proteína Ligasas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular , Movimiento Celular/fisiología , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación
5.
Exp Dermatol ; 28(9): 1079-1082, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31338879

RESUMEN

Since Drosophila melanogaster has proven to be a useful model system to study phenotypes of oncogenic mutations and to identify new anti-cancer drugs, we generated human BRAFV600E homologous dRaf mutant (dRafA572E ) Drosophila melanogaster strains to use these for characterisation of mutant phenotypes and exploit these phenotypes for drug testing. For mutant gene expression, the GAL4/UAS expression system was used. dRafA572E was expressed tissue-specific in the eye, epidermis, heart, wings, secretory glands and in the whole animal. Expression of dRaf A572E under the control of an eye-specific driver led to semi-lethality and a rough eye phenotype. The vast majority of other tissue-specific and ubiquitous drivers led to a lethal phenotype only. The rough eye phenotype was used to test BRAF inhibitor vemurafenib and MEK1/2 inhibitor cobimetinib. There was no phenotype rescue by this treatment. However, a significant rescue of the lethal phenotype was observed under a gut-specific driver. Here, MEK1/2 inhibitor cobimetinib rescued Drosophila larvae to reach pupal stage in 37% of cases as compared to 1% in control experiments. Taken together, the BRAFV600E homolog dRaf A572E exerts mostly lethal effects in Drosophila. Gut-specific dRaf A572E expression might in future be developed further for drug testing.


Asunto(s)
Azetidinas/farmacología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-raf/genética , Animales , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/fisiología , Evaluación Preclínica de Medicamentos , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Intestinos/enzimología , Larva , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especificidad de Órganos , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/deficiencia , Proteínas Proto-Oncogénicas c-raf/fisiología , Vemurafenib/farmacología
6.
Cancer Cell ; 34(2): 315-330.e7, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30033091

RESUMEN

Platinum-based chemotherapeutics represent a mainstay of cancer therapy, but resistance limits their curative potential. Through a kinome RNAi screen, we identified microtubule-associated serine/threonine kinase 1 (MAST1) as a main driver of cisplatin resistance in human cancers. Mechanistically, cisplatin but no other DNA-damaging agents inhibit the MAPK pathway by dissociating cRaf from MEK1, while MAST1 replaces cRaf to reactivate the MAPK pathway in a cRaf-independent manner. We show clinical evidence that expression of MAST1, both initial and cisplatin-induced, contributes to platinum resistance and worse clinical outcome. Targeting MAST1 with lestaurtinib, a recently identified MAST1 inhibitor, restores cisplatin sensitivity, leading to the synergistic attenuation of cancer cell proliferation and tumor growth in human cancer cells and patient-derived xenograft models.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , MAP Quinasa Quinasa 1/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Activación Enzimática , Femenino , Humanos , Ratones
7.
Lab Invest ; 98(6): 708-714, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29540860

RESUMEN

Interleukin-33 (IL-33) is a potent contributor to antiviral immune responses and antitumor immunity. We recently discovered that IL-33 is overexpressed in dectin-1-activated dendritic cells (DCs). However, mechanisms of dectin-1-induced IL-33 expression in DCs remain elusive. Curdlan, an agonist of dectin-1, was used to mature DCs in this study. We found that dectin-1-induced IL-33 expression in DCs relies on Syk and Raf-1 pathways. By using nuclear factor (NF)-κB inhibitors, we also found that dectin-1-induced IL-33 expression relies on NF-κB signaling. Furthermore, through Syk/Raf-1-NF-κB pathway, dectin-1 signaling stimulates DCs to overexpress interferon regulatory factor 4 (IRF4), which directly upregulates the expression of IL-33 in dectin-1-activated DCs. Thus, our study provides new insights into the mechanisms of dectin-1-induced IL-33 expression in DCs and may provide new targets for improving DC-based cancer immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/fisiología , Interleucina-33/genética , Lectinas Tipo C/fisiología , Animales , Lectinas Tipo C/agonistas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal/fisiología , Quinasa Syk/fisiología , beta-Glucanos/farmacología
8.
Gene ; 656: 53-59, 2018 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-29499332

RESUMEN

C-RAF was the first member of the RAF kinase family to be discovered. Since its discovery, C-RAF has been found to regulate many fundamental cell processes, such as cell proliferation, cell death, and metabolism. However, the majority of these functions are achieved through interactions with different proteins; the genes regulated by C-RAF in its active or inactive state remain unclear. In the work, we used RNA-seq analysis to study the global transcriptomes of C-RAF bearing or C-RAF knockout cells in quiescent or EGF activated states. We identified 3353 genes that are promoted or suppressed by C-RAF. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that these genes are involved in drug addiction, cardiomyopathy, autoimmunity, and regulation of cell metabolism. Our results provide a panoramic view of C-RAF function, including known and novel functions, and have revealed potential targets for elucidating the role of C-RAF.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas Proto-Oncogénicas c-raf/fisiología , Transcriptoma , Secuencia de Bases , Técnicas de Inactivación de Genes , Ontología de Genes , Células HEK293 , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-raf/genética , ARN Mensajero/genética , Análisis de Secuencia de ARN/métodos
9.
J Agric Food Chem ; 66(4): 926-934, 2018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29307174

RESUMEN

In this study, we elucidated the immunomodulatory activity of docosahexaenoic acid (DHA) on protein expression in RAW264.7 cells and its molecular mechanism. The results showed that the proliferation index of RAW264.7 cells at 48 h was about 173.03 ± 7.82% after the treatment of 2.4 µM DHA. DHA could activate RAW264.7 cells by the G-protein coupled cell membrane receptor GPR120-C-Raf- mitogen-activated protein kinases (MAPKs)-nuclear factor κB (NF-κB) p65 pathway. In addition, 2.4 µM of DHA could significantly increase (P < 0.01) the mRNA and protein expression of inducible nitric oxide synthase (iNOS), which is consistent with the result of the NO release. ELISA results revealed that DHA could enhance the protein expression of cytokines IL-1ß, IL-6, IL-10, IL-12, TNF-α, IFN-γ, and TGF-ß. These results indicated that the immunomodulatory mechanism of RAW264.7 cells by DHA was associated with the release of NO and cytokines by stimulating the GPR120, C-Raf, and MAPKs to the NF-κB p65 pathway.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Factores Inmunológicos/farmacología , Activación de Macrófagos/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología , Animales , Citocinas/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/fisiología , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas c-raf/fisiología , Células RAW 264.7 , Transducción de Señal , Factor de Transcripción ReIA/fisiología
10.
J BUON ; 22(1): 258-264, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28365963

RESUMEN

PURPOSE: SOX5 plays important roles in various kinds of cancers. However, the expression and roles of SOX5 in osteosarcoma (OS) have not been investigated well. In the present study we aimed to investigate the mechanism of SOX5 in OS. METHODS: OS and adjacent non-cancerous specimens were obtained from patients with OS. PCR was applied to detect SOX5 mRNA. Then human OS cell lines (U2OS, SoSP-M, SoSP-9607, and MG-63) and one immortalized normal osteoblast hFOB1.19 were investigated. SOX5 knocking with shRNA in U2OS and SOX5 upregulation with recombinant plasmid in MG-63 were applied. Real-time cell monitoring system and invasion assay were used, and Western blot assay was performed to detect the protein level of E-cadherin, N-cadherin, Vimentin and Snail, where Glyceraldehyde3- phosphate dehydrogenase (GAPDH) was presented as control. P<0.05 was considered as statistically significant. RESULTS: Significant upregulation of SOX5 in OS tissues and cell lines was identified. The gain- and loss-of-function studies suggested that OS cell migration and invasion were promoted significantly by SOX5. Additionally, SOX5 promoted epithelial-mesenchymal transition (EMT) by regulation of Snail. CONCLUSION: SOX5 is a novel regulator of EMT in OS, and is a potential target for OS.


Asunto(s)
Neoplasias Óseas/patología , Transición Epitelial-Mesenquimal , Osteosarcoma/patología , Factores de Transcripción SOXD/fisiología , Factores de Transcripción de la Familia Snail/fisiología , Neoplasias Óseas/mortalidad , Línea Celular Tumoral , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Invasividad Neoplásica , Osteosarcoma/mortalidad , Proteínas Proto-Oncogénicas c-raf/fisiología
11.
Leukemia ; 31(7): 1593-1602, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27890930

RESUMEN

Histone deacetylase (HDAC) inhibitors, which are approved for the treatment of cutaneous T-cell lymphoma and multiple myeloma, are undergoing evaluation in other lymphoid neoplasms. How they kill susceptible cells is incompletely understood. Here, we show that trichostatin A, romidepsin and panobinostat induce apoptosis across a panel of malignant B cell lines, including lines that are intrinsically resistant to bortezomib, etoposide, cytarabine and BH3 mimetics. Further analysis traces the pro-apoptotic effects of HDAC inhibitors to increased acetylation of the chaperone heat shock protein 90 (HSP90), causing release and degradation of the HSP90 client proteins RASGRP1 and CRAF, which in turn leads to downregulation of mitogen-activated protein kinase pathway signaling and upregulation of the pro-apoptotic BCL2 family member BIM in vitro and in vivo. Importantly, these pro-apoptotic effects are mimicked by RASGRP1 small interfering RNA (siRNA) or HSP90 inhibition and reversed by overexpression of constitutively active MEK1 or siRNA-mediated downregulation of BIM. Collectively, these observations not only identify a new HSP90 client protein, RASGRP1, but also delineate a complete signaling pathway from HSP90 acetylation through RASGRP1 and CRAF degradation to BIM upregulation that contributes to selective cytotoxicity of HDAC inhibitors in lymphoid malignancies.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Proteínas de Unión al ADN/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas HSP90 de Choque Térmico/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Linfoma de Células B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Células Cultivadas , Resistencia a Antineoplásicos , Genes bcl-2 , Humanos , Linfoma de Células B/patología , Ratones , Regulación hacia Arriba
12.
Oncogene ; 36(14): 2014-2022, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-27841865

RESUMEN

Raf-1 has an important role in cellular antiapoptosis. So far, there is no solid evidence that shows that Raf-1 mutation is associated with cancer development. In the course of further study of Raf-1 signaling, we have reported that Raf-1 hyperphosphorylation inhibits its kinase activity toward its downstream mitogen-activated protein kinase kinase 1/2 (MEK1/2) and proposed a model for negative feedback regulation of Raf-1. Here, we show that there is no hyperphosphorylation in some cancer cells, which results in increased kinase activity and enhances the antiapoptotic ability. Inhibition of either Raf-1 or ALG-2 (apoptosis-linked gene 2) expression results in apoptosis signal-regulating kinase 1/c-Jun N-terminal kinase (ASK1/JNK) signaling activation, and cell sensitivity to chemotherapeutic reagents, indicating that inhibition of ASK1/JNK apoptotic signaling by Raf-1 is mediated by ALG-2. A previous report indicated that extracellular signal-regulated kinase 1/2 (ERK1/2) were responsible for Raf-1 hyperphosphorylation. However, our evidence shows that when ERK1/2 are activated and the Raf-1 gene is not mutated, Raf-1 is not hyperphosphorylated in these cells, indicating that ERK1/2 are not responsible for the Raf-1 hyperphosphorylation in these cancer cell lines. Surprisingly, we also found that Raf-1 is not a necessary kinase for MEK1/2 activation under normal tissue culture conditions, but is required for MEK1/2 activation under apoptosis-inducing conditions. Our research demonstrates that although Raf-1 gene is not mutated, an abnormality of Raf-1 kinase feedback regulation enhances its antiapoptotic function, and Raf-1 can still be a pharmaceutical target to increase chemotherapy or radiotherapy sensitivity in these cancer cells.


Asunto(s)
Apoptosis/genética , Retroalimentación Fisiológica/fisiología , Neoplasias/genética , Neoplasias/patología , Proteínas Proto-Oncogénicas c-raf/fisiología , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células Tumorales Cultivadas
13.
Sci Rep ; 6: 26803, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27222248

RESUMEN

B-Raf inhibitors have been used for the treatment of some B-Raf-mutated cancers. They effectively inhibit B-Raf/MEK/ERK signaling in cancers harboring mutant B-Raf, but paradoxically activates MEK/ERK in Ras-mutated cancers. Death receptor 5 (DR5), a cell surface pro-apoptotic protein, triggers apoptosis upon ligation with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or aggregation. This study focused on determining the effects of B-Raf inhibition on DR5 expression and DR5 activation-induced apoptosis in Ras-mutant cancer cells. Using chemical and genetic approaches, we have demonstrated that the B-Raf inhibitor PLX4032 induces DR5 upregulation exclusively in Ras-mutant cancer cells; this effect is dependent on Ras/c-Raf/MEK/ERK signaling activation. PLX4032 induces DR5 expression at transcriptional levels, largely due to enhancing CHOP/Elk1-mediated DR5 transcription. Pre-exposure of Ras-mutated cancer cells to PLX4032 sensitizes them to TRAIL-induced apoptosis; this is also a c-Raf/MEK/ERK-dependent event. Collectively, our findings highlight a previously undiscovered effect of B-Raf inhibition on the induction of DR5 expression and the enhancement of DR5 activation-induced apoptosis in Ras-mutant cancer cells and hence may suggest a novel therapeutic strategy against Ras-mutated cancer cells by driving their death due to DR5-dependent apoptosis through B-Raf inhibition.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes ras , Indoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Sulfonamidas/farmacología , Apoptosis/efectos de los fármacos , Bencimidazoles/farmacología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Imidazoles/farmacología , Proteínas de Neoplasias/genética , Oximas/farmacología , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Proteínas Recombinantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factor de Transcripción CHOP/fisiología , Transcripción Genética/efectos de los fármacos , Vemurafenib , Proteína Elk-1 con Dominio ets/fisiología
14.
Leuk Res ; 45: 68-74, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27101150

RESUMEN

MEK/ERK signal pathway was required for the differentiation of granulocytes, megakaryocytes and erythrocytes. Recently, MEK/ERK cascade was reported to be involved in all-trans retinoic acid (ATRA) induced differentiation in acute promyelocytic leukemia (APL) cells. However, the upstream and downstream molecules of MEK/ERK signal pathway in this cell model remains to be elucidated. In this work, we showed that RAF-1 was activated and the blockade of RAF-1 activation attenuated MEK/ERK activation as well as ATRA-induced differentiation. ATRA-enhanced protein levels of C/EBPß, C/EBPε and PU.1, which were required for differentiation in APL cells, were suppressed by the specific inhibitor of MEK. However, MEK inhibition had no effect on the degradation of PML-RARα fusion protein or the restoration of PML nuclear bodies by ATRA treatment. Taken together, our study suggested that RAF-1/MEK/ERK cascade was involved in ATRA-induced differentiation in APL cells through enhancing the protein level of C/EBPß, C/EBPε and PU.1.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/patología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Tretinoina/farmacología , Antineoplásicos/farmacología , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/efectos de los fármacos , Línea Celular Tumoral , Granulocitos/efectos de los fármacos , Granulocitos/patología , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/efectos de los fármacos , Transactivadores/biosíntesis , Transactivadores/efectos de los fármacos
15.
Cell Mol Life Sci ; 72(20): 3983-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25975225

RESUMEN

The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K(+) channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf (+/-) mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.


Asunto(s)
Oído Interno/metabolismo , Pérdida Auditiva/genética , Ruido , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Apoptosis/genética , Cóclea/metabolismo , Oído Interno/embriología , Femenino , Pérdida Auditiva/metabolismo , Masculino , Ratones , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal
16.
Curr Opin Oncol ; 26(4): 454-61, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24849046

RESUMEN

PURPOSE OF REVIEW: The modalities of Ras mutation detection, its role as a predictive biomarker, mechanisms of wild-type Ras activation, and the role of Ras-directed targeted therapies will be discussed mainly in colorectal cancer. RECENT FINDINGS: RAS genotype is generally considered to be highly concordant between primary colorectal tumours and metastases. However, recent data show significant discordance between primary tumours and specific metastatic sites, but also heterogeneity within primary tumours. Moreover, the mechanisms of Ras activation expand far beyond mutations through altered expression or function of physiological Ras activators and inhibitors. Accordingly, genomic signatures of Ras or epidermal growth factor receptor (EGFR) activation are being developed and are potential predictive biomarkers of response to anti-EGFR antibodies. Finally, several recent clinical trials targeting Ras or its downstream signalling with statins or Raf inhibitors have shown promising activity in chemorefractory metastatic colorectal cancer. SUMMARY: RAS mutation remains an important biomarker predicting response to anti-EGFR therapies and perhaps clinical outcomes after surgery for metastatic colorectal cancer, but new techniques including genomic signatures need to be validated to take into account the complexity of Ras activation. The importance of Ras signalling as a therapeutic target has recently been outlined by successful clinical trials with Raf inhibitors.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas ras/fisiología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Genes ras , Genotipo , Humanos , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal/fisiología
17.
Ann Pathol ; 33(6): 375-85, 2013 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24331719

RESUMEN

Cutaneous melanoma is a malignant tumor with a high metastatic potential. If an early treatment is associated with a favorable outcome, the prognosis of metastatic melanoma remains poor. Advances in molecular characterization of cancers, notably the discovery of BRAF gene mutations in metastatic melanoma, allowed to the recent development of targeted therapies against mutated BRAF protein. Despite high tumor response rates observed in clinical trials, these new drugs are associated with frequent secondary tumor resistance occurrence and paradoxical carcinogenic side effects. The cellular and molecular mechanisms of these carcinogenic side effects and secondary resistance are not yet fully elucidated and are actually intensely studied. This review of the literature focus on the mechanisms of these carcinogenic side effects and on the tumor resistance associated with anti-BRAF targeted therapies.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Indoles/efectos adversos , Indoles/farmacología , Leucemia/inducido químicamente , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/secundario , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Primarias Secundarias/inducido químicamente , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/inducido químicamente , Sulfonamidas/efectos adversos , Sulfonamidas/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/inducido químicamente , Resistencia a Antineoplásicos/genética , Activación Enzimática/efectos de los fármacos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes ras , Humanos , Indoles/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Queratoacantoma/inducido químicamente , Melanoma/inducido químicamente , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Modelos Biológicos , Terapia Molecular Dirigida , Mutación Missense , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/enzimología , Nevo Pigmentado/enzimología , Nevo Pigmentado/patología , Mutación Puntual , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/fisiología , Sulfonamidas/uso terapéutico , Microambiente Tumoral , Vemurafenib
18.
Am J Pathol ; 183(2): 617-25, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747947

RESUMEN

A number of inhibitors have been used to dissect the functional relevance of Jak2 in endothelial homeostasis, with disparate results. Given that Jak2 deficiency leads to embryonic lethality, the exact role of Jak2 in the regulation of postnatal endothelial function is yet to be fully elucidated. We generated a model in which Jak2 deficiency can be induced by tamoxifen in adult mice. Loss of Jak2 significantly impaired endothelium-dependent response capacity for vasodilators. Matrigel plug assays indicated a notable decrease in endothelial angiogenic function in Jak2-deficient mice. Studies in a hindlimb ischemic model indicated that Jak2 activity is likely to be a prerequisite for prompt perfusion recovery, based on the concordance of temporal changes in Jak2 expression during the course of ischemic injury and perfusion recovery. A remarkable delay in perfusion recovery, along with reduced capillary and arteriole formation, was observed in Jak2-deficient mice. Antibody array studies indicated that loss of Jak2 led to repressed eNOS expression. In mechanistic studies, Jak2 deficiency attenuated Raf-1/MEK1 signaling, which then reduced activity of Sp-1, an essential transcription factor responsible for eNOS expression. These data are important not only for understanding the exact role that Jak2 plays in endothelial homeostasis, but also for assessing Jak2-based therapeutic strategies in a variety of clinical settings.


Asunto(s)
Janus Quinasa 2/deficiencia , MAP Quinasa Quinasa 1/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Miembro Posterior/irrigación sanguínea , Isquemia/enzimología , Janus Quinasa 2/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Tamoxifeno/farmacología , Vasodilatadores/farmacología
19.
Cancer Cell ; 23(5): 594-602, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23680146

RESUMEN

ATP competitive inhibitors of the BRAF(V600E) oncogene paradoxically activate downstream signaling in cells bearing wild-type BRAF (BRAF(WT)). In this study, we investigate the biochemical mechanism of wild-type RAF (RAF(WT)) activation by multiple catalytic inhibitors using kinetic analysis of purified BRAF(V600E) and RAF(WT) enzymes. We show that activation of RAF(WT) is ATP dependent and directly linked to RAF kinase activity. These data support a mechanism involving inhibitory autophosphorylation of RAF's phosphate-binding loop that, when disrupted either through pharmacologic or genetic alterations, results in activation of RAF and the mitogen-activated protein kinase (MAPK) pathway. This mechanism accounts not only for compound-mediated activation of the MAPK pathway in BRAF(WT) cells but also offers a biochemical mechanism for BRAF oncogenesis.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas raf/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/fisiología , Línea Celular Tumoral , Humanos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/fisiología , Quinasas raf/genética , Quinasas raf/metabolismo
20.
J Immunol ; 190(8): 4129-35, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23475217

RESUMEN

The immune system is essential to maintain homeostasis with resident microbial populations, ensuring that the symbiotic host-microbial relationship is maintained. In parallel, commensal microbes significantly shape mammalian immunity at the host mucosal surface, as well as systemically. Candida albicans is an opportunistic pathogen that lives as a commensal on skin and mucosa of healthy individuals. Little is known about its capacity to modulate responses toward other microorganisms, such as colonizing bacteria (e.g., intestinal microorganisms). The aim of this study was to assess the cytokine production of PBMCs induced by commensal bacteria when these cells were primed by C. albicans. We show that C. albicans and ß-1,3-glucan induce priming of human primary mononuclear cells and this leads to enhanced cytokine production upon in vitro stimulation with TLR ligands and bacterial commensals. This priming requires the ß-1,3-glucan receptor dectin-1 and the noncanonical Raf-1 pathway. In addition, although purified mannans cannot solely mediate the priming, the presence of mannosyl residues in the cell wall of C. albicans is nevertheless required. In conclusion, C. albicans is able to modify cytokine responses to TLR ligands and colonizing bacteria, which is likely to impact the inflammatory reaction during mucosal diseases.


Asunto(s)
Candida albicans/inmunología , Citocinas/biosíntesis , Lectinas Tipo C/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal/inmunología , Receptores Toll-Like/fisiología , Bacteroides fragilis/inmunología , Candida albicans/genética , Escherichia coli/inmunología , Humanos , Tolerancia Inmunológica , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Ligandos , Membrana Mucosa/microbiología , Piel/microbiología , Staphylococcus aureus/inmunología , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...