450 results on '"Crespo, Piero"'
Search Results
202. An Integrated Global Analysis of Compartmentalized HRAS Signaling.
- Author
-
Santra, Tapesh, Herrero, Ana, Rodriguez, Javier, von Kriegsheim, Alex, Iglesias-Martinez, Luis F., Schwarzl, Thomas, Higgins, Des, Aye, Thin-Thin, Heck, Albert J.R., Calvo, Fernando, Agudo-Ibáñez, Lorena, Crespo, Piero, Matallanas, David, and Kolch, Walter
- Abstract
Modern omics technologies allow us to obtain global information on different types of biological networks. However, integrating these different types of analyses into a coherent framework for a comprehensive biological interpretation remains challenging. Here, we present a conceptual framework that integrates protein interaction, phosphoproteomics, and transcriptomics data. Applying this method to analyze HRAS signaling from different subcellular compartments shows that spatially defined networks contribute specific functions to HRAS signaling. Changes in HRAS protein interactions at different sites lead to different kinase activation patterns that differentially regulate gene transcription. HRAS-mediated signaling is the strongest from the cell membrane, but it regulates the largest number of genes from the endoplasmic reticulum. The integrated networks provide a topologically and functionally resolved view of HRAS signaling. They reveal distinct HRAS functions including the control of cell migration from the endoplasmic reticulum and TP53-dependent cell survival when signaling from the Golgi apparatus. • MiNETi (Mixed Network Integration) enables the integration of multi-omics datasets • MiNETi provides an integrated view of HRAS signaling from different subcellular sites • HRAS controls its interactome, phosphoproteome, and transcriptome site specifically • HRAS regulates cell migration and p53-mediated apoptosis from endomembranes Santra et al. develop MiNETi (Mixed Network Integration) to integrate multi-omics data. Applying MiNETi to analyze the interactome, phosphoproteome, and transcriptome regulated by HRAS signaling from different subcellular compartments shows that HRAS controls phosphorylation-dependent signaling mainly from the cell membrane but regulates a large number of genes from endomembranes. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
203. Vav mediates Ras stimulation by direct activation of the GDP/GTP exchange factor Ras GRP1.
- Author
-
Caloca, María J, Zugaza, José L, Matallanas, David, Crespo, Piero, and Bustelo, Xosé R
- Subjects
GUANINE nucleotide exchange factors ,MOLECULAR biology - Published
- 2019
- Full Text
- View/download PDF
204. Analysis of IncF plasmids evolution: nucleotide sequence of an IncFIII replication region
- Author
-
Lopez, Javier, primary, Crespo, Piero, additional, Rodriguez, Jose C., additional, Andres, Isabel, additional, and Ortiz, Jose M., additional
- Published
- 1989
- Full Text
- View/download PDF
205. The RAS-ERK pathway: A route for couples
- Author
-
Santos, Eugenio and Crespo, Piero
- Published
- 2018
- Full Text
- View/download PDF
206. Mechanisms of action of vitamin D in colon cancer.
- Author
-
Ferrer-Mayorga, Gemma, Larriba, María Jesús, Crespo, Piero, and Muñoz, Alberto
- Subjects
- *
COLON cancer , *VITAMIN D , *VITAMIN D receptors , *FIBROBLASTS , *GUT microbiome - Abstract
Highlights • Colorectal cancer is the neoplasia that is most closely linked to vitamin D deficiency in epidemiological studies. • Calcitriol inhibits the proliferation, migration, invasiveness and angiogenesis of colon carcinoma cells. • Calcitriol promotes differentiation of colon carcinoma cells and sensitizes them to apoptosis. • Calcitriol reduces the protumoral effects of colon cancer-associated fibroblasts. • Calcitriol also regulates the biology of intestinal immune cells and affects the intestinal microbiota. Abstract Colorectal cancer (CRC) is the neoplasia that is most frequently associated with vitamin D deficiency in epidemiological and observational studies in terms of incidence and mortality. Many mechanistic studies show that the active vitamin D metabolite (1α,25-dihydroxyvitamin D 3 or calcitriol) inhibits proliferation and promotes epithelial differentiation of human colon carcinoma cell lines that express vitamin D receptor (VDR) via the regulation of a high number of genes. A key action underlining this effect is the multilevel inhibition of the Wnt/β-catenin signaling pathway, whose abnormal activation in colon epithelial cells initiates and promotes CRC. Recently, our group has shown that calcitriol modulates gene expression and inhibits protumoral properties of patient-derived colon cancer-associated fibroblasts (CAFs). Accordingly, high VDR expression in tumor stromal fibroblasts is associated with longer survival of CRC patients. Moreover, many types of immune cells express VDR and are regulated by calcitriol, which probably contributes to its action against CRC. Given the role attributed to the intestinal microbiota in CRC and the finding that it is altered by vitamin D deficiency, an indirect antitumoral effect of calcitriol is also plausible at this level. In summary, calcitriol has an array of potential protective effects against CRC by acting on carcinoma cells, CAFs, immune cells and probably also the gut microbiota. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
207. Analysis of Rhes Activation State and Effector Function.
- Author
-
Bernal, Juan and Crespo, Piero
- Abstract
An abstract of the article "Analysis of Rhes Activation State and Effector Function," by Juan Bernal and Piero Crespo is presented.
- Published
- 2006
- Full Text
- View/download PDF
208. Scaffold coupling: ERK activation by transphosphorylation across different scaffold protein species.
- Author
-
Martín-Vega, Ana, Ruiz-Peinado, Laura, García-Gómez, Rocío, Herrero, Ana, de la Fuente-Vivas, Dalia, Parvathaneni, Swetha, Caloto, Rubén, Morante, Marta, von Kriegsheim, Alex, Bustelo, Xosé R., Sacks, David B., Casar, Berta, and Crespo, Piero
- Abstract
The article offers information about the activation of extracellular signal-regulated kinase (ERK) by transphosphorylation across different scaffold protein species. It also identifies interactions between scaffold proteins and trans-phosphorylation as an additional level of regulation in the ERK cascade, with broad implications in signaling and the design of scaffold protein–aimed therapeutics.
- Published
- 2023
- Full Text
- View/download PDF
209. Metallothionein-3 promotes cisplatin chemoresistance remodelling in neuroblastoma
- Author
-
Zbynek Heger, Berta Casar, Roman Guran, Vladislav Strmiska, Vivian de los Ríos, Miguel Angel Merlos Rodrigo, Yazan Haddad, Vojtech Adam, Piero Crespo, J. Ignacio Casal, Hana Michalkova, Petra Pokorná, Tomas Eckschlager, European Commission, Czech Health Research Council, Czech Science Foundation, Federation of European Biochemical Societies, Ministry of Education, Youth and Sports (Czech Republic), Merlos Rodrigo, Miguel Angel, Strmiska, Vladislav, Casar, Berta, Crespo, Piero, de los Ríos, Vivian, Casal, J. Ignacio, Haddad, Yazan, Guran, Roman, Adam, Vojtech, Merlos Rodrigo, Miguel Angel [0000-0002-1920-0948], Strmiska, Vladislav [0000-0002-7036-1640], Casar, Berta [0000-0002-3058-5631], Crespo, Piero [0000-0003-2825-7783], de los Ríos, Vivian [0000-0001-5582-6879], Casal, J. Ignacio [0000-0003-1085-2840], Haddad, Yazan [0000-0002-7844-4336], Guran, Roman [0000-0002-2912-714X], and Adam, Vojtech [0000-0002-8527-286X]
- Subjects
medicine.medical_treatment ,Science ,cisplatin ,Chick Embryo ,Biochemistry ,Article ,Biological pathway ,neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neuroblastoma ,medicine ,Animals ,Humans ,cancer ,030304 developmental biology ,Cancer ,Cisplatin ,0303 health sciences ,Chemotherapy ,Multidisciplinary ,Chemistry ,chemoresistance ,medicine.disease ,Metallothionein 3 ,Neoplasm Proteins ,3. Good health ,Gene Expression Regulation, Neoplastic ,Proteasome ,Drug Resistance, Neoplasm ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Metallothionein-3 ,medicine.drug - Abstract
14 p.-6 fig., Metallothionein-3 has poorly characterized functions in neuroblastoma. Cisplatin-based chemotherapy is a major regimen to treat neuroblastoma, but its clinical efficacy is limited by chemoresistance. We investigated the impact of human metallothionein-3 (hMT3) up-regulation in neuroblastoma cells and the mechanisms underlying the cisplatin-resistance. We confirmed the cisplatin-metallothionein complex formation using mass spectrometry. Overexpression of hMT3 decreased the sensitivity of neuroblastoma UKF-NB-4 cells to cisplatin. We report, for the first time, cisplatin-sensitive human UKF-NB-4 cells remodelled into cisplatin-resistant cells via high and constitutive hMT3 expression in an in vivo model using chick chorioallantoic membrane assay. Comparative proteomic analysis demonstrated that several biological pathways related to apoptosis, transport, proteasome, and cellular stress were involved in cisplatin-resistance in hMT3 overexpressing UKF-NB-4 cells. Overall, our data confirmed that up-regulation of hMT3 positively correlated with increased cisplatin-chemoresistance in neuroblastoma, and a high level of hMT3 could be one of the causes of frequent tumour relapses., The authors gratefully acknowledge to the Proteomics and Genomics Facility (CIB-CSIC), a member of ProteoRed-ISCIII network, for the protein identification by nLC-MS/MS. The authors also acknowledge funding from the European Research Council (ERC) under the European Union´s Horizon 2020 Research and Innovation Programme (grant agreement No. 759585), the Czech Health Research Council (project no. 15-28334A), the Czech Science Foundation (project no. 19-13766J), FEBS—Federation of European Biochemical Societies and under the CEITEC 2020 project (LQ1601) by the Ministry of Education, Youth and Sports of the Czech Republic.
- Published
- 2021
210. Inhibiting ERK dimerization ameliorates BRAF-driven anaplastic thyroid cancer.
- Author
-
Zaballos, Miguel A., Acuña-Ruiz, Adrián, Morante, Marta, Riesco-Eizaguirre, Garcilaso, Crespo, Piero, and Santisteban, Pilar
- Abstract
Background: RAS-to-ERK signaling is crucial for the onset and progression of advanced thyroid carcinoma, and blocking ERK dimerization provides a therapeutic benefit in several human carcinomas. Here we analyzed the effects of DEL-22379, a relatively specific ERK dimerization inhibitor, on the activation of the RAS-to-ERK signaling cascade and on tumor-related processes in vitro and in vivo. Methods: We used a panel of four human anaplastic thyroid carcinoma (ATC) cell lines harboring BRAF or RAS mutations to analyze ERK dynamics and tumor-specific characteristics. We also assessed the impact of DEL-22379 on the transcriptional landscape of ATC cell lines using RNA-sequencing and evaluated its therapeutic efficacy in an orthotopic mouse model of ATC. Results: DEL-22379 impaired upstream ERK activation in BRAF- but not RAS-mutant cells. Cell viability and metastasis-related processes were attenuated by DEL-22379 treatment, but mostly in BRAF-mutant cells, whereas in vivo tumor growth and dissemination were strongly reduced for BRAF-mutant cells and mildly reduced for RAS-mutant cells. Transcriptomics analyses indicated that DEL-22379 modulated the transcriptional landscape of BRAF- and RAS-mutant cells in opposite directions. Conclusions: Our findings establish that BRAF- and RAS-mutant thyroid cells respond differentially to DEL-22379, which cannot be explained by the previously described mechanism of action of the inhibitor. Nonetheless, DEL-22379 demonstrated significant anti-tumor effects against BRAF-mutant cells in vivo with an apparent lack of toxicity, making it an interesting candidate for the development of combinatorial treatments. Our data underscore the differences elicited by the specific driver mutation for thyroid cancer onset and progression, which should be considered for experimental and clinical approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
211. The Ras-ERK pathway: Understanding site-specific signaling provides hope of new anti-tumor therapies.
- Author
-
Calvo, Fernando, Agudo-Ibáñez, Lorena, and Crespo, Piero
- Subjects
- *
CYTOLOGY , *CANCER , *CARCINOGENESIS , *RAS proteins , *ONCOGENES - Abstract
The article explains Ras-Extracellularregulated Kinase (ERK) pathway in cancer initiation and progression. Ras protein is described as an oncogene in human cancers. It states that Ras-ERK signals can emanate from distinct subcellular localizations. It contends that interfering with local Ras-ERK subsignals can block cellular transformation and tumor progression. Understanding site-specific regulators can serve as anti-tumor targets.
- Published
- 2010
- Full Text
- View/download PDF
212. Metallothionein-3 promotes cisplatin chemoresistance remodelling in neuroblastoma.
- Author
-
Rodrigo, Miguel Angel Merlos, Michalkova, Hana, Strmiska, Vladislav, Casar, Berta, Crespo, Piero, de los Rios, Vivian, Ignacio Casal, J., Haddad, Yazan, Guran, Roman, Eckschlager, Tomas, Pokorna, Petra, Heger, Zbynek, and Adam, Vojtech
- Subjects
- *
METALLOTHIONEIN , *CISPLATIN , *NEUROBLASTOMA , *DRUG resistance in cancer cells , *MASS spectrometry - Abstract
Metallothionein-3 has poorly characterized functions in neuroblastoma. Cisplatin-based chemotherapy is a major regimen to treat neuroblastoma, but its clinical efficacy is limited by chemoresistance. We investigated the impact of human metallothionein-3 (hMT3) up-regulation in neuroblastoma cells and the mechanisms underlying the cisplatin-resistance. We confirmed the cisplatin-metallothionein complex formation using mass spectrometry. Overexpression of hMT3 decreased the sensitivity of neuroblastoma UKF-NB-4 cells to cisplatin. We report, for the first time, cisplatin-sensitive human UKF-NB-4 cells remodelled into cisplatin-resistant cells via high and constitutive hMT3 expression in an in vivo model using chick chorioallantoic membrane assay. Comparative proteomic analysis demonstrated that several biological pathways related to apoptosis, transport, proteasome, and cellular stress were involved in cisplatin-resistance in hMT3 overexpressing UKF-NB-4 cells. Overall, our data confirmed that up-regulation of hMT3 positively correlated with increased cisplatin-chemoresistance in neuroblastoma, and a high level of hMT3 could be one of the causes of frequent tumour relapses. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
213. Characterisation of HRas local signal transduction networks using engineered site-specific exchange factors.
- Author
-
Herrero, Ana, Reis-Cardoso, Mariana, Jiménez-Gómez, Iñaki, Doherty, Carolanne, Agudo-Ibañez, Lorena, Pinto, Adán, Calvo, Fernando, Kolch, Walter, Crespo, Piero, and Matallanas, David
- Subjects
- *
CELLULAR signal transduction , *CELL membranes , *EXCHANGE , *ONCOGENES - Abstract
Ras GTPases convey signals from different types of membranes. At these locations, different Ras isoforms, interactors and regulators generate different biochemical signals and biological outputs. The study of Ras localisation-specific signal transduction networks has been hampered by our inability to specifically activate each of these Ras pools. Here, we describe a new set of site-specific tethered exchange factors, engineered by fusing the RasGRF1 CDC25 domain to sub-localisation-defining cues, whereby Ras pools at specific locations can be precisely activated. We show that the CDC25 domain has a high specificity for activating HRas but not NRas and KRas. This unexpected finding means that our constructs mainly activate endogenous HRas. Hence, their use enabled us to identify distinct pathways regulated by HRas in endomembranes and plasma membrane microdomains. Importantly, these new constructs unveil different patterns of HRas activity specified by their subcellular localisation. Overall, the targeted GEFs described herein constitute ideal tools for dissecting spatially-defined HRas biochemical and biological functions. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
214. Regulators of the RAS-ERK pathway as therapeutic targets in thyroid cancer.
- Author
-
Zaballos, Miguel A., Acuña-Ruiz, Adrián, Morante, Marta, Crespo, Piero, and Santisteban, Pilar
- Subjects
- *
THYROID cancer , *RAS proteins , *SCAFFOLD proteins , *ANAPLASTIC thyroid cancer , *MITOGEN-activated protein kinase phosphatases , *THERAPEUTICS , *GOVERNORS (Machinery) , *ANIMAL models in research - Abstract
Thyroid cancer is mostly an ERK-driven carcinoma, as up to 70% of thyroid carcinomas are caused by mutations that activate the RAS/ERK mitogenic signaling pathway. The incidence of thyroid cancer has been steadily increasing for the last four decades; yet, there is still no effective treatment for advanced thyroid carcinomas. Current research efforts are focused on impairing ERK signaling with small-molecule inhibitors, mainly at the level of BRAF and MEK. However, despite initial promising r esults in animal models, the clinical success of these inhibitors has been limited by th e emergence of tumor resistance and relapse. The RAS/ERK pathway is an extremely complex signaling cascade with multiple points of control, offering many potential therapeutic targets: from the modulatory proteins regulating the activation state of RAS proteins to the scaffolding proteins of the pathway that provide spatial specificity to the signals, and finally, the negative feedbacks and phosphatases responsible for inactivating the pathway. The aim of this review is to give an overview of the biology of RAS/ERK regulators in human cancer highlighting relevant information on thyroid cancer and future areas of research. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
215. ERK dimerization as a determinant of tumour progression factors
- Author
-
Fuente Vivas, Dalia de la, Crespo, Pedro, Casar Martínez, Berta, Universidad de Cantabria, Crespo, Piero, and Casar, Berta
- Subjects
KSR-1 ,Invasion ,Migración ,Invasión ,ERK dimerization ,ERK dimerización ,Migration ,EGF - Abstract
Tesis doctoral.-- Universidad de Santander., [EN]: Nearly 50% of human malignancies have dysregulated RAS-ERK signalling; inhibiting it is a valid strategy for antineoplastic intervention. ERK dimerization is essential for extranuclear ERK signalling, but not for nuclear signalling. Here we describe the role of ERK dimers in tumour progression in response to different stimuli. ERK dimerization-mediated cell spreading involves actin remodelling and ERK scaffold proteins as potential targets driven during tumorigenesis. In this study, we performed chemotaxis assays and employed 2D migration, 3D organoid formation and chick embryo metastasis models. We have observed the tumorigenic role mediated by ERK dimerization at different stages of tumour progression, monitored with the ERK dimer inhibitor DEL-22379., [ES]: Casi el 50% de las neoplasias humanas presentan una señalización RAS-ERK no regulada; inhibirla es una estrategia válida para la intervención antineoplásica. La dimerización de ERK es esencial para la señalización extranuclear de las ERK, pero no para la nuclear. Aquí describimos el papel de los dímeros de ERK en la progresión tumoral en respuesta a diferentes estímulos. La diseminación celular mediada por la dimerización de ERK implica la remodelación de la actina, y las proteínas andamio de ERK como objetivos potenciales impulsados durante la tumorogénesis. En este estudio se realizan ensayos de quimiotaxis y se emplean modelos de migración 2D, de formación de organoides 3D y de metástasis de embrión de pollo. Hemos observado el papel tumorogénico mediado por la dimerización de ERK en los distintos estados de la progresión tumoral, monitorizado con el inhibidor de dímeros de ERK, DEL-22379.
- Published
- 2021
216. Absence of K-Ras Reduces Proliferation and Migration But Increases Extracellular Matrix Synthesis in Fibroblasts.
- Author
-
Muñoz‐Félix, José M., Fuentes‐Calvo, Isabel, Cuesta, Cristina, Eleno, Nélida, Crespo, Piero, López‐Novoa, José M., and Martínez‐Salgado, Carlos
- Subjects
- *
EXTRACELLULAR matrix , *FIBROBLASTS , *RAS proteins , *SMAD proteins , *CELL proliferation , *PROTEIN expression - Abstract
The involvement of Ras-GTPases in the development of renal fibrosis has been addressed in the last decade. We have previously shown that H- and N-Ras isoforms participate in the regulation of fibrosis. Herein, we assessed the role of K-Ras in cellular processes involved in the development of fibrosis: proliferation, migration, and extracellular matrix (ECM) proteins synthesis. K-Ras knockout (KO) mouse embryonic fibroblasts (K- ras−/−) stimulated with transforming growth factor-β1 (TGF-β1) exhibited reduced proliferation and impaired mobility than wild-type fibroblasts. Moreover, an increase on ECM production was observed in K-Ras KO fibroblasts in basal conditions. The absence of K-Ras was accompanied by reduced Ras activation and ERK phosphorylation, and increased AKT phosphorylation, but no differences were observed in TGF-β1-induced Smad signaling. The MEK inhibitor U0126 decreased cell proliferation independently of the presence of K -ras but reduced migration and ECM proteins expression only in wild-type fibroblasts, while the PI3K-AKT inhibitor LY294002 decreased cell proliferation, migration, and ECM synthesis in both types of fibroblasts. Thus, our data unveil that K-Ras and its downstream effector pathways distinctively regulate key biological processes in the development of fibrosis. Moreover, we show that K-Ras may be a crucial mediator in TGF-β1-mediated effects in this cell type. J. Cell. Physiol. 231: 2224-2235, 2016. © 2016 Wiley Periodicals, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
217. ERK1/2 MAP kinases promote cell cycle entry by rapid, kinase-independent disruption of retinoblastoma--lamin A complexes.
- Author
-
Rodríguez, Javier, Calvo, Fernando, González, José M., Casar, Berta, Andrés, Vicente, and Crespo, Piero
- Subjects
- *
CELL proliferation , *MITOGEN-activated protein kinases , *CELL cycle , *PHOSPHORYLATION , *RETINOBLASTOMA - Abstract
As orchestrators of essential cellular processes like proliferation, ERK1/2 mitogen-activated protein kinase signals impact on cell cycle regulation. A-type lamins are major constituents of the nuclear matrix that also control the cell cycle machinery by largely unknown mechanisms. In this paper, we disclose a functional liaison between ERK1/2 and lamin A whereby cell cycle progression is regulated. We demonstrate that lamin A serves as a mutually exclusive dock for ERK1/2 and the retinoblastoma (Rb) protein. Our results reveal that, immediately after their postactivation entrance in the nucleus, ERK1/2 dislodge Rb from its interaction with lamin A, thereby facilitating its rapid phosphorylation and consequently promoting E2F activation and cell cycle entry. Interestingly, these effects are independent of ERK1/2 kinase activity. We also show that cellular transformation and tumor cell proliferation are dependent on the balance between lamin A and nuclear ERK1/2 levels, which determines Rb accessibility for phosphorylation/inactivation. [ABSTRACT FROM AUTHOR]
- Published
- 2010
218. Fast regulation of AP-1 activity through interaction of lamin A/C, ERK1/2, and c-Fos at the nuclear envelope.
- Author
-
González, José Maria, Navarro-Puche, Ana, Casar, Berta, Crespo, Piero, and Andrés, Vicente
- Subjects
- *
PROTEINS , *PROTEIN-protein interactions , *SEQUESTRATION (Chemistry) , *TRANSCRIPTION factors , *PHOSPHORYLATION - Abstract
Sequestration of c-Fos at the nuclear envelope (NE) through interaction with A-type lamins suppresses AP-1-dependent transcription. We show here that c-Fos accumulation within the extraction-resistant nuclear fraction (ERNF) and its interaction with lamin A are reduced and enhanced by gain-of and loss-of ERK1/2 activity, respectively. Moreover, hindering ERK1/2-dependent phosphorylation of c-Fos attenuates its release from the ERNF induced by serum and promotes its interaction with lamin A. Accordingly, serum stimulation rapidly releases preexisting c-Fos from the NE via ERK1/2-dependent phosphorylation, leading to a fast activation of AP-1 before de nova c-Fos synthesis. Moreover, lamin A-null cells exhibit increased AP-1 activity and reduced levels of c-Fos phosphorylation. We also find that active ERK1/2 interacts with lamin A and colocalizes with c-Fos and A-type lamins at the NE. Thus, NE-bound ERK1/2 functions as a molecular switch for rapid mitogen-dependent AP-1 activation through phosphorylation-induced release of preexisting c-Fos from its inhibitory interaction with lamin A/C. [ABSTRACT FROM AUTHOR]
- Published
- 2008
219. Lysophosphatidic acid rescues RhoA activation and phosphoinositides levels in astrocytes exposed to ethanol.
- Author
-
Martínez, Susana E., Lázaro-Diéguez, Francisco, Selva, Javier, Calvo, Fernando, Piqueras, Jaime-Renau, Crespo, Piero, Claro, Enrique, and Egea, Gustavo
- Subjects
- *
LYSOPHOSPHOLIPIDS , *PHOSPHOINOSITIDES , *NEUROGLIA , *ESTERIFICATION , *CYTOPLASM , *CELLS - Abstract
Long-term ethanol treatment substantially impairs glycosylation and membrane trafficking in primary cultures of rat astrocytes. Our previous studies indicated that these effects were attributable to a primary alteration in the dynamics and organization of the actin cytoskeleton, although the molecular mechanism(s) remains to be elucidated. As small Rho GTPases and phosphoinositides are involved in the actin cytoskeleton organization, we now explore the effects of chronic ethanol treatment on these pathways. We show that chronic ethanol treatment of rat astrocytes specifically reduced endogenous levels of active RhoA as a result of the increase of in the RhoGAP activity. Furthermore, ethanol-treated astrocytes showed reduced phosphoinositides levels. When lysophosphatidic acid was added to ethanol-treated astrocytes, it rapidly reverted actin cytoskeleton reorganization and raised active RhoA levels and phosphoinositides content to those observed in untreated astrocytes. Overall, our results indicate that the harmful effects of chronic exposure to ethanol on a variety of actin dynamics-associated cellular events are primarily because of alterations of activated RhoA and phosphoinositides pools. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
220. Mxi2 promotes stimulus-independent ERK nuclear translocation.
- Author
-
Casar, Berta, Sanz-Moreno, Victoria, Yazicioglu, Mustafa N, Rodríguez, Javier, Berciano, María T, Lafarga, Miguel, Cobb, Melanie H, and Crespo, Piero
- Subjects
- *
CHROMOSOMAL translocation , *MITOGEN-activated protein kinases , *CELL proliferation , *PROTEINS , *CELL nuclei - Abstract
Spatial regulation of ERK1/2 MAP kinases is an essential yet largely unveiled mechanism for ensuring the fidelity and specificity of their signals. Mxi2 is a p38α isoform with the ability to bind ERK1/2. Herein we show that Mxi2 has profound effects on ERK1/2 nucleocytoplasmic distribution, promoting their accumulation in the nucleus. Downregulation of endogenous Mxi2 by RNAi causes a marked reduction of ERK1/2 in the nucleus, accompanied by a pronounced decline in cellular proliferation. We demonstrate that Mxi2 functions in nuclear shuttling of ERK1/2 by enhancing the nuclear accumulation of both phosphorylated and unphosphorylated forms in the absence of stimulation. This process requires the direct interaction of both proteins and a high-affinity binding of Mxi2 to ERK-binding sites in nucleoporins, In this respect, Mxi2 acts antagonistically to PEA15, displacing it from ERK1/2 complexes. These results point to Mxi2 as a key spatial regulator for ERK1/2 and disclose an unprecedented stimulus-independent mechanism for ERK nuclear import. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
221. Myc Antagonizes Ras-mediated Growth Arrest in Leukemia Cells through the Inhibition of the Ras-ERK-p21Cip1 Pathway.
- Author
-
Vaqué, Jose P., Navascues, Joaquin, Shiio, Yuzuru, Laiho, Marikki, Ajenjo, Nuria, Mauleon, Itsaso, Matallanas, David, Crespo, Piero, and León, Javier
- Subjects
- *
GENETIC mutation , *CELLS , *CELL culture , *CELL lines , *MYELOID leukemia , *TISSUE culture , *BIOCHEMISTRY , *NUCLEIC acids - Abstract
Even though RAS usually acts as a dominant transforming oncogene, in primary fibroblasts and some established cell lines Ras inhibits proliferation. This can explain the virtual absence of RAS mutations in some types of tumors, such as chronic myeloid leukemia (CML). We report that in the CML cell line K562 Ras induces p21Cip1 expression through the Raf-MEK-ERK pathway. Because K562 cells are deficient for p15INK4b, p16INK4a, p14ARF, and p53, this would be the main mechanism whereby Ras up-regulates p21 expression in these cells. Accordingly, we also found that Ras suppresses K562 growth by signaling through the Raf-ERK pathway. Because c-Myc and Ras cooperate in cell transformation and c-Myc is up-regulated in CML, we investigated the effect of c-Myc on Ras activity in K562 cells. c-Myc antagonized the induction of p21Cip1 mediated by oncogenic H-, K-, and N-Ras and by constitutively activated Raf and ERK2. Activation of the p21Cip1 promoter by Ras was dependent on Sp⅓ binding sites in K562. However, mutational analysis of the p21 promoter and the use of a Gal4-Sp1 chimeric protein strongly suggest that c-Myc affects Sp1 transcriptional activity but not the binding of Sp1 to the p21 promoter. c-Myc-mediated impairment of Ras activity on p21 expression required a transactivation domain, a DNA binding region, and a Max binding region. Moreover, the effect was independent of Miz1 binding to c-Myc. Consistent with its effect on p21Cip1 expression, c-Myc rescued cell growth inhibition induced by Ras. The data suggest that in particular tumor types, such as those associated with CML, c-Myc contributes to tumorigenesis by inhibiting Ras antiproliferative activity. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
222. The small GTP-binding protein, Rhes, regulates signal transduction from G protein-coupled receptors.
- Author
-
Vargiu, Pierfrancesco, De Abajo, Ricardo, Garcia-Ranea, Juan Antonio, Valencia, Alfonso, Santisteban, Pilar, Crespo, Piero, and Bernal, Juan
- Subjects
- *
THYROTROPIN , *CELLULAR signal transduction , *FIBROBLASTS , *NUCLEOTIDES , *PROTEINS - Abstract
The Ras homolog enriched in striatum, Rhes, is the product of a thyroid hormone-regulated gene during brain development. Rhes and the dexamethasone-induced Dexras1 define a novel distinct subfamily of proteins within the Ras family, characterized by an extended variable domain in the carboxyl terminal region. We have carried this study because there is a complete lack of knowledge on Rhes signaling. We show that in PC12 cells, Rhes is targeted to the plasma membrane by farnesylation. We demonstrate that about 30% of the native Rhes protein is bound to GTP and this proportion is unaltered by typical Ras family nucleotide exchange factors. However, Rhes is not transforming in murine fibroblasts. We have also examined the role of Rhes in cell signaling. Rhes does not stimulate the ERK pathway. By contrast, it binds to and activates PI3K. On the other hand, we demonstrate that Rhes impairs the activation of the cAMP/PKA pathway by thyroid-stimulating hormone, and by an activated ß2 adrenergic receptor by a mechanism that suggests uncoupling of the receptor to its cognate heterotrimeric complex. Overall, our results provide the initial insights into the role in signal transduction of this novel Ras family member.Oncogene (2004) 23, 559-568. doi:10.1038/sj.onc.1207161 [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
223. Ras en el Complejo de Golgi : señales antitumorales mediadas por PTPK
- Author
-
Jiménez Gómez, Iñaki, Crespo Baraja, Pedro, Universidad de Cantabria, Crespo, Piero, and Ministerio de Economía y Competitividad (España)
- Subjects
PTPK and Apoptosis ,Golgi ,Antitumoral ,Ras - Abstract
Tesis Doctoral presentada por Iñaki Jiménez Gómez para optar al Grado de Doctor en Biología Molecular y Biomedicina por la Universidad de Cantabria., [ES]: Las células transforman constantemente los estímulos recibidos del entorno en respuestas biológicas. Este proceso, está mediado por multitud de reacciones químicas en cadena. La fosforilación reversible de proteínas, controlada por la acción contrapuesta de quinasas y fosfatasas, es un mecanismo de regulación fundamental en estas cascadas de señalización. Una de las vías mejor caracterizadas, es la ruta Ras-ERK. Además de en membrana plasmática, las proteínas Ras están presentes y son funcionales en endomembranas. De este modo, debemos entender las señales de Ras como el producto de la integración de distintas sub-señales sito-específicas. En este trabajo, demostramos que Ras activo en Golgi induce apoptosis inhibiendo la activación de ERK. Este efecto es ejercido por PTPK, la molécula responsable de repercutir negativamente sobre los niveles de fosforilación de C-RAF en residuos de tirosina. De este modo, queda establecido que el potencial oncogénico de Ras es estrictamente dependiente de la sublocalización celular., [EN]: Cells respond to changes in their microenvironment eliciting a specific response. This process is mediated by a multitude of chain reactions. Reversible protein phosphorylation, controlled by the opposing activities of kinases and phosphatases, is a fundamental regulatory mechanism in these cascades. One of the most well-known signaling pathways is the Ras/ERK route. In addition to plasma membrane, Ras proteins are present and are functional in endomembranes. In this way, we must understand Ras signaling as the product of the integration of different site-specific sub-signals. In this work, we demonstrate that Ras, activated at Golgi Complex, induces apoptosis by inhibiting ERK activation. This effect is exerted by PTPK, the molecule responsible for negatively affecting the levels of phosphorylation of C-RAF in tyrosine residues. Thus, it is established that the oncogenic potential of Ras is strictly dependent on cellular sublocalization., El autor de esta Tesis ha disfrutado de una beca de Formación del Personal Investigador (FPI) del Ministerio de Economía y Competitividad durante el período de realización de la misma.
- Published
- 2017
224. RAS Subcellular Localization Inversely Regulates Thyroid Tumor Growth and Dissemination.
- Author
-
García-Ibáñez, Yaiza, Riesco-Eizaguirre, Garcilaso, Santisteban, Pilar, Casar, Berta, and Crespo, Piero
- Subjects
- *
ANIMAL experimentation , *BIOLOGICAL models , *CELL membranes , *GENE expression , *GENETIC mutation , *POULTRY , *PROTEINS , *THYROID gland tumors , *TUMOR markers , *VASCULAR endothelial growth factors , *SEQUENCE analysis - Abstract
Simple Summary: RAS mutations occur frequently in thyroid tumors, but the extent to which they are associated to tumor aggressiveness is still uncertain. HRAS proteins occupy different subcellular localizations, from which they regulate distinct biochemical processes. Herein, we demonstrate that the capacity of HRAS-transformed thyroid cells to extravasate and invade distant organs is orchestrated by HRAS subcellular localization, by a mechanism dependent on VEGF-B secretion. Interestingly, aggressiveness inversely correlates with tumor size. Moreover, we have identified the acyl protein thioesterase APT-1, a regulator of HRAS sublocalization, as a determinant of thyroid tumor growth versus dissemination. As such, alterations in APT-1 expression levels can dramatically affect the behavior of thyroid tumors. In this respect, APT-1 levels could serve as a biomarker that may help in the stratification of HRAS mutant thyroid tumors based on their aggressiveness. RAS mutations are the second most common genetic alteration in thyroid tumors. However, the extent to which they are associated with the most aggressive phenotypes is still controversial. Regarding their malignancy, the majority of RAS mutant tumors are classified as undetermined, which complicates their clinical management and can lead to undesired under- or overtreatment. Using the chick embryo spontaneous metastasis model, we herein demonstrate that the aggressiveness of HRAS-transformed thyroid cells, as determined by the ability to extravasate and metastasize at distant organs, is orchestrated by HRAS subcellular localization. Remarkably, aggressiveness inversely correlates with tumor size. In this respect, we also show that RAS site-specific capacity to regulate tumor growth and dissemination is dependent on VEGF-B secretion. Furthermore, we have identified the acyl protein thioesterase APT-1 as a determinant of thyroid tumor growth versus dissemination. We show that alterations in APT-1 expression levels can dramatically affect the behavior of thyroid tumors, based on its role as a regulator of HRAS sublocalization at distinct plasma membrane microdomains. In agreement, APT-1 emerges in thyroid cancer clinical samples as a prognostic factor. As such, APT-1 levels could serve as a biomarker that could help in the stratification of HRAS mutant thyroid tumors based on their aggressiveness. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
225. Regulación de la morfología nuclear por las GTPasas Rho
- Author
-
Colón-Bolea, Paula, Crespo Baraja, Pedro, Universidad de Cantabria, Crespo, Piero, Ministerio de Economía y Competitividad (España), and Consejo Superior de Investigaciones Científicas (España)
- Subjects
Morfología nuclear ,Proteínas ,GTPasas RHo ,Migración celular - Abstract
Tesis doctoral presentada por Paula Colón Bolea para optar al Grado de Doctor por la Universidad de Cantabria y realizada en el Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) y el Departamento de Biología Molecular de la Facultad de Medicina de la Universidad de Cantabria (Santander)., La superfamilia de GTPasas Ras es una familia de proteínas que regulan procesos biológicos muy importantes como la proliferación, la diferenciación y la apoptosis entre muchos. Dentro de esta familia de GTPasas se encuentran las GTPasas Rho. Entre las GTPasas Rho más estudiadas se encuentran Rac1, RhoA y Cdc42. Destacan entre otros aspectos por su papel como reguladoras del citoesqueleto, tanto de actina como los microtúbulos. Es por esta razón por lo que son proteínas estrechamente ligadas a migración celular y por ello a metástasis. Durante la migración celular, una de las limitaciones que tiene una célula es el tamaño de poro a través del cual puede pasar. Esta limitación viene dada por el tamaño del núcleo. Por ello, una mayor plasticidad nuclear puede favorecer una mayor migración celular. Se sabe que la morfología nuclear se determina en parte a través del citoesqueleto, que está en contacto con el nucleoesqueleto a través del complejo LINC. Sin embargo, a pesar de ser las RhoGTPasas unas reguladoras importantes del citoesqueleto, apenas hay estudios que relacionen estas proteínas con la morfología del núcleo. Tras un exhaustivo análisis en varias líneas celulares se verificó que las GTPasas Rac1, RhoA y Cdc42 eran capaces de modificar la morfología nuclear, sobre todo la proteína Rac1. Posteriormente se llevaron a cabo una serie de experimentos que permitieron concluir que dicha influencia sobre el núcleo era debida a una cooperación entre el citoesqueleto de actina (que regula contractilidad) y los microtúbulos. El efector a través del cual regulan la contractilidad actina-miosina está aún por determinar, pero es a través de PAK1 que Rac1 regula los microtúbulos para iniciar el cambio morfológico. Mediante el uso de un modelo animal, el modelo de embrión de pollo, se pudo establecer que asociado al cambio morfológico nuclear inducido por Rac1 se encontraba una mayor intravasación de las células tumorales. Esta mayor intravasación se ve mermada si se fuerza la ruptura del complejo LINC, demostrando así que es a través del complejo que se están ejerciendo estos cambios. A pesar de existir una mayor intravasación, no se asocia ésta a una mayor colonización en otros órganos, dando a entender que Rac1 sólo aporta ventajas invasivas a la hora de intravasar, pero no extravasar. Por tanto, con el trabajo de esta tesis podemos concluir que las RhoGTPasas y sobre todo Rac1 inducen un cambio morfológico nuclear en las células que facilita su intravasación. Este cambio lo ejercen a través del citoesqueleto y la conexión de éste con el complejo LINC., La autora de esta Tesis ha disfrutado de una beca de Formación de Personal Invesitigador (FPI) y de un contrato por el CSIC durante la realización de la misma.
- Published
- 2015
226. Ras and Rap Signal Bidirectional Synaptic Plasticity via Distinct Subcellular Microdomains.
- Author
-
Zhang, Lei, Zhang, Peng, Wang, Guangfu, Zhang, Huaye, Zhang, Yajun, Yu, Yilin, Zhang, Mingxu, Xiao, Jian, Crespo, Piero, Hell, Johannes W., Lin, Li, Huganir, Richard L., and Zhu, J. Julius
- Subjects
- *
NEUROPLASTICITY , *RAS proteins , *CELLULAR signal transduction , *LONG-term synaptic depression , *LYSOSOMES - Abstract
Summary How signaling molecules achieve signal diversity and specificity is a long-standing cell biology question. Here we report the development of a targeted delivery method that permits specific expression of homologous Ras-family small GTPases (i.e., Ras, Rap2, and Rap1) in different subcellular microdomains, including the endoplasmic reticulum, lipid rafts, bulk membrane, lysosomes, and Golgi complex, in rodent hippocampal CA1 neurons. The microdomain-targeted delivery, combined with multicolor fluorescence protein tagging and high-resolution dual-quintuple simultaneous patch-clamp recordings, allows systematic analysis of microdomain-specific signaling. The analysis shows that Ras signals long-term potentiation via endoplasmic reticulum PI3K and lipid raft ERK, whereas Rap2 and Rap1 signal depotentiation and long-term depression via bulk membrane JNK and lysosome p38MAPK, respectively. These results establish an effective subcellular microdomain-specific targeted delivery method and unveil subcellular microdomain-specific signaling as the mechanism for homologous Ras and Rap to achieve signal diversity and specificity to control multiple forms of synaptic plasticity. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
227. Especificidad espacio-temporal de las señales Ras-ERK en la determinación de respuestas biológicas
- Author
-
Herrero, Ana and Crespo, Piero
- Subjects
Señalización ,Cáncer ,MAPK ,Ras - Abstract
Tesis Doctoral presentada en el Departamento de Biología Molecular de la Facultad de Medicina de la Universidad de Cantabria para la obtención del grado de Doctora en Bioquímica., La cascada de señalización de ERK(Extracellular-Signal-Regulated Kinase) es una ruta formada por proteínas quinasas activadas por mitógenos (MAPKs) que regula una amplia variedad de procesos celulares, tan diferentes como la proliferación, diferenciación, supervivencia y el estrés. La activación de la ruta Ras-ERK se ha descrito, predominantemente, desde la membrana plasmática, pero en la última década se ha demostrado que Ras también puede ser activada en otras sublocalizaciones celulares, como por ejemplo en retículo endoplasmático, mitocondria o aparato de Golgi. En este trabajo se demuestra que la localización subcelular de Ras dentro de la célula es determinante en la capacidad de rescate de viabilidad celular en células carentes de Ras endógeno. Por otra parte, se sabe que la duración de la actividad de ERK puede ser determinante en la determinación de respuestas biológicas. En células MCF7 se producen diferentes respuestas en función del estímulo, asociadas con distinta cinética de activación, quedando demostrado que las variaciones temporales de la señal Ras-ERK son responsables de la determinación de una respuesta biológica., El autor de esta Tesis ha disfrutado de una beca de Formación de Profesorado Universitario (FPU) del Ministerio de Educación durante el período de realización de la misma.
- Published
- 2012
228. Relevancia bioquímica y biológica de la dimerización de ERK2
- Author
-
Pinto, Adán and Crespo, Piero
- Abstract
La ruta de Ras-ERK es una de las más conservadas y mejor caracterizadas. La mayoría de los factores de crecimiento activan a las quinasas ERK (quinasas reguladas por señales extracelulares), actuando en primer termino a través de la proteína G pequeña, o monomérica, Ras. La activación de esta ruta es capaz de modular procesos celulares esenciales como proliferación, diferenciación y supervivencia. Se ha analizando el efecto anti-apoptótico y la capacidad transformante de H-Ras, N-Ras y K-Ras constitutivamente activos en sus correspondientes localizaciones subcelulares y en la mayor cantidad de tipos celulares (epiteliales, fibroblastos y tumorales). Se ha estudiado como afecta la capacidad de ERK de formar dímeros, cuando se activa vía Ras-Raf, a la formación de complejos scaffold-ERK-substratos de ERK. Se ha comprobado como influye la capacidad de ERK para formar dímeros en la proliferación de lineas celulares tumorales (expresando de forma estable un mutante de ERK que no dimeriza) y como es capaz de modular la capacidad de formar tumores de estas líneas celulares cuando se inyectan subcutaneamente en ratones inmunodeprimidos. Este trabajo ha sido publicado en la revista Molecular Cell, en su número de Septiembre del año 2008 (Berta Casar, Adán Pinto et al). A la vista de que la inhibición de la dimerización en la célula tiene efectos fisiológicos tan importantes como la inhibición de la formación de tumores en ratones, se está realizando un screening en busca de fármacos que sean capaces de inhibir la dimerización tanto in vivo como in vitro. Estos son los trabajos que están siendo realizados como parte final de la tesis de Adán Pinto Fernández.
- Published
- 2011
229. Regulación de la sublocalización celular de H-Ras y su especificidad funcional
- Author
-
Agudo-Ibáñez, Lorena and Crespo, Piero
- Abstract
Los experimentos llevados a cabo en esta Tesis Doctoral se centraron en el estudio de la proteína H-Ras a diferentes niveles. Comenzamos analizando los efectos que ejerce H-Ras regulando de manera positiva o negativa numerosos genes. Mediante la técnica de microarrays comprobamos que el número total de genes regulados varía enormemente según la localización subcelular desde la que señalizara H-Ras. El mayor número de genes se regula desde membrana desordenada, mientras que desde balsas lipídicas no se controla específicamente ningún gen. Por otro lado quisimos conocer la localización de H-Ras endógeno en la membrana plasmática bajo diferentes condiciones de crecimiento y/o líneas celulares. Mediante técnicas de separación de los microdominios que componen la membrana plasmática conseguimos ubicar a H-Ras en membrana desordenada o balsas lipídicas y seguir su movimiento. Para estos estudios se emplearon análisis por fraccionamientos de gradientes de sacarosa con los que separamos membrana plasmática de diferentes densidades o distinta solubilidad, así como microscopía confocal de doble marcaje. Dando un paso más en este estudio comprobamos si la presencia de H-Ras en un dominio u otro de la membrana plasmática dependía de su grado de palmitilación y esto se traducía en la activación diferencial de unos efectores frente a otros y un efecto biológico cambiante.
- Published
- 2010
230. ERK1/2 mitogen-activated protein kinase dimerization is essential for the regulation of cell motility.
- Author
-
de la Fuente-Vivas D, Cappitelli V, García-Gómez R, Valero-Díaz S, Amato C, Rodriguéz J, Duro-Sánchez S, von Kriegsheim A, Grusch M, Lozano J, Arribas J, Casar B, and Crespo P
- Abstract
ERK1/2 mitogen-activated protein kinases (ERK) are key regulators of basic cellular processes, including proliferation, survival, and migration. Upon phosphorylation, ERK becomes activated and a portion of it dimerizes. The importance of ERK activation in specific cellular events is generally well documented, but the role played by dimerization is largely unknown. Here, we demonstrate that impeding ERK dimerization precludes cellular movement by interfering with the molecular machinery that executes the rearrangements of the actin cytoskeleton. We also show that a constitutively dimeric ERK mutant can drive cell motility per se, demonstrating that ERK dimerization is both necessary and sufficient for inducing cellular migration. Importantly, we unveil that the scaffold protein kinase suppressor of Ras 1 (KSR1) is a critical element for endowing external agonists, acting through tyrosine kinase receptors, with the capacity to induce ERK dimerization and, subsequently, to unleash cellular motion. In agreement, clinical data disclose that high KSR1 expression levels correlate with greater metastatic potential and adverse evolution of mammary tumors. Overall, our results portray both ERK dimerization and KSR1 as essential factors for the regulation of cell motility and mammary tumor dissemination., (© 2024 The Author(s). Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
231. ERK2 stimulates MYC transcription by anchoring CDK9 to the MYC promoter in a kinase activity-independent manner.
- Author
-
Agudo-Ibáñez L, Morante M, García-Gutiérrez L, Quintanilla A, Rodríguez J, Muñoz A, León J, and Crespo P
- Subjects
- Phosphorylation, Cyclin-Dependent Kinases genetics, Promoter Regions, Genetic, Transcription, Genetic, RNA Polymerase II genetics, RNA Polymerase II chemistry, RNA Polymerase II metabolism, Transcription Factors metabolism
- Abstract
The transcription factor MYC regulates cell proliferation, transformation, and survival in response to growth factor signaling that is mediated in part by the kinase activity of ERK2. Because ERK2 can also bind to DNA to modify gene expression, we investigated whether it more directly regulates MYC transcription. We identified ERK2 binding sites in the MYC promoter and detected ERK2 at the promoter in various serum-stimulated cell types. Expression of nuclear-localized ERK2 constructs in serum-starved cells revealed that ERK2 in the nucleus-regardless of its kinase activity-increased MYC mRNA expression and MYC protein abundance. ERK2 bound to the promoter through its amino-terminal insert domain and to the cyclin-dependent kinase CDK9 (which activates RNA polymerase II) through its carboxyl-terminal conserved docking domain. Both interactions were essential for ERK2-induced MYC expression, and depleting ERK impaired CDK9 occupancy and RNA polymerase II progression at the MYC promoter. Artificially tethering CDK9 to the MYC promoter by fusing it to the ERK2 insert domain was sufficient to stimulate MYC expression in serum-starved cells. Our findings demonstrate a role for ERK2 at the MYC promoter acting as a kinase-independent anchor for the recruitment of CDK9 to promote MYC expression.
- Published
- 2023
- Full Text
- View/download PDF
232. Immune Checkpoint Inhibitors and RAS-ERK Pathway-Targeted Drugs as Combined Therapy for the Treatment of Melanoma.
- Author
-
Morante M, Pandiella A, Crespo P, and Herrero A
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, MAP Kinase Signaling System, Immunotherapy methods, Melanoma pathology, Antineoplastic Agents pharmacology
- Abstract
Metastatic melanoma is a highly immunogenic tumor with very poor survival rates due to immune system escape-mechanisms. Immune checkpoint inhibitors (ICIs) targeting the cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and the programmed death-1 (PD1) receptors, are being used to impede immune evasion. This immunotherapy entails an increment in the overall survival rates. However, melanoma cells respond with evasive molecular mechanisms. ERK cascade inhibitors are also used in metastatic melanoma treatment, with the RAF activity blockade being the main therapeutic approach for such purpose, and in combination with MEK inhibitors improves many parameters of clinical efficacy. Despite their efficacy in inhibiting ERK signaling, the rewiring of the melanoma cell-signaling results in disease relapse, constituting the reinstatement of ERK activation, which is a common cause of some resistance mechanisms. Recent studies revealed that the combination of RAS-ERK pathway inhibitors and ICI therapy present promising advantages for metastatic melanoma treatment. Here, we present a recompilation of the combined therapies clinically evaluated in patients.
- Published
- 2022
- Full Text
- View/download PDF
233. The Rho guanosine nucleotide exchange factors Vav2 and Vav3 modulate epidermal stem cell function.
- Author
-
Lorenzo-Martín LF, Menacho-Márquez M, Fernández-Parejo N, Rodríguez-Fdez S, Pascual G, Abad A, Crespo P, Dosil M, Benitah SA, and Bustelo XR
- Subjects
- Animals, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Epidermal Cells cytology, Epidermal Cells metabolism, Epidermis metabolism, Hair Follicle cytology, Hair Follicle metabolism, Mice, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Skin Neoplasms genetics, Skin Neoplasms metabolism, Skin Neoplasms pathology, rho GTP-Binding Proteins genetics, rho GTP-Binding Proteins metabolism, Proto-Oncogene Proteins c-vav genetics, Proto-Oncogene Proteins c-vav metabolism, Skin cytology, Skin metabolism, Stem Cells cytology, Stem Cells metabolism
- Abstract
It is known that Rho GTPases control different aspects of the biology of skin stem cells (SSCs). However, little information is available on the role of their upstream regulators under normal and tumorigenic conditions in this process. To address this issue, we have used here mouse models in which the activity of guanosine nucleotide exchange factors of the Vav subfamily has been manipulated using both gain- and loss-of-function strategies. These experiments indicate that Vav2 and Vav3 regulate the number, functional status, and responsiveness of hair follicle bulge stem cells. This is linked to gene expression programs related to the reinforcement of the identity and the quiescent state of normal SSCs. By contrast, in the case of cancer stem cells, they promote transcriptomal programs associated with the identity, activation state, and cytoskeletal remodeling. These results underscore the role of these Rho exchange factors in the regulation of normal and tumor epidermal stem cells., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
234. Editorial: Ras and Other GTPases in Cancer: From Basic to Applied Research.
- Author
-
Cho KJ, Liang JR, Crespo P, and Aran V
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
235. RAS Dimers: The Novice Couple at the RAS-ERK Pathway Ball.
- Author
-
Herrero A and Crespo P
- Subjects
- Animals, Humans, Signal Transduction, Carcinogenesis metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Protein Multimerization, ras Proteins metabolism
- Abstract
Signals conveyed through the RAS-ERK pathway constitute a pivotal regulatory element in cancer-related cellular processes. Recently, RAS dimerization has been proposed as a key step in the relay of RAS signals, critically contributing to RAF activation. RAS clustering at plasma membrane microdomains and endomembranes facilitates RAS dimerization in response to stimulation, promoting RAF dimerization and subsequent activation. Remarkably, inhibiting RAS dimerization forestalls tumorigenesis in cellular and animal models. Thus, the pharmacological disruption of RAS dimers has emerged as an additional target for cancer researchers in the quest for a means to curtail aberrant RAS activity.
- Published
- 2021
- Full Text
- View/download PDF
236. Editorial overview: Macromolecular assemblies: clues from structural insights.
- Author
-
Walters KJ and Crespo P
- Subjects
- Macromolecular Substances
- Published
- 2021
- Full Text
- View/download PDF
237. ARID2 deficiency promotes tumor progression and is associated with higher sensitivity to chemotherapy in lung cancer.
- Author
-
Moreno T, Monterde B, González-Silva L, Betancor-Fernández I, Revilla C, Agraz-Doblas A, Freire J, Isidro P, Quevedo L, Blanco R, Montes-Moreno S, Cereceda L, Astudillo A, Casar B, Crespo P, Morales Torres C, Scaffidi P, Gómez-Román J, Salido E, and Varela I
- Subjects
- A549 Cells, Animals, Cell Line, Tumor, Disease Progression, Female, High-Throughput Nucleotide Sequencing, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Mice, Nude, Survival Rate, Transcription Factors genetics, Transcription Factors metabolism, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Transcription Factors deficiency
- Abstract
The survival rate in lung cancer remains stubbornly low and there is an urgent need for the identification of new therapeutic targets. In the last decade, several members of the SWI/SNF chromatin remodeling complexes have been described altered in different tumor types. Nevertheless, the precise mechanisms of their impact on cancer progression, as well as the application of this knowledge to cancer patient management are largely unknown. In this study, we performed targeted sequencing of a cohort of lung cancer patients on genes involved in chromatin structure. In addition, we studied at the protein level the expression of these genes in cancer samples and performed functional experiments to identify the molecular mechanisms linking alterations of chromatin remodeling genes and tumor development. Remarkably, we found that 20% of lung cancer patients show ARID2 protein loss, partially explained by the presence of ARID2 mutations. In addition, we showed that ARID2 deficiency provokes profound chromatin structural changes altering cell transcriptional programs, which bolsters the proliferative and metastatic potential of the cells both in vitro and in vivo. Moreover, we demonstrated that ARID2 deficiency impairs DNA repair, enhancing the sensitivity of the cells to DNA-damaging agents. Our findings support that ARID2 is a bona fide tumor suppressor gene in lung cancer that may be exploited therapeutically.
- Published
- 2021
- Full Text
- View/download PDF
238. RAC1 induces nuclear alterations through the LINC complex to enhance melanoma invasiveness.
- Author
-
Colón-Bolea P, García-Gómez R, Shackleton S, Crespo P, Bustelo XR, and Casar B
- Subjects
- Animals, Cell Line, Tumor, Cell Movement genetics, Cell Nucleus Shape physiology, Chick Embryo, Cytoskeleton metabolism, Humans, Membrane Proteins metabolism, Microtubules metabolism, Neoplasm Invasiveness genetics, Nuclear Matrix metabolism, Nuclear Proteins metabolism, p21-Activated Kinases metabolism, rac1 GTP-Binding Protein physiology, rho GTP-Binding Proteins metabolism, Cell Nucleus metabolism, Melanoma metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
RHO GTPases are key regulators of the cytoskeletal architecture, which impact a broad range of biological processes in malignant cells including motility, invasion, and metastasis, thereby affecting tumor progression. One of the constraints during cell migration is the diameter of the pores through which cells pass. In this respect, the size and shape of the nucleus pose a major limitation. Therefore, enhanced nuclear plasticity can promote cell migration. Nuclear morphology is determined in part through the cytoskeleton, which connects to the nucleoskeleton through the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. Here, we unravel the role of RAC1 as an orchestrator of nuclear morphology in melanoma cells. We demonstrate that activated RAC1 promotes nuclear alterations through its effector PAK1 and the tubulin cytoskeleton, thereby enhancing migration and intravasation of melanoma cells. Disruption of the LINC complex prevented RAC1-induced nuclear alterations and the invasive properties of melanoma cells. Thus, RAC1 induces nuclear morphology alterations through microtubules and the LINC complex to promote an invasive phenotype in melanoma cells.
- Published
- 2020
- Full Text
- View/download PDF
239. RAS GTPase-dependent pathways in developmental diseases: old guys, new lads, and current challenges.
- Author
-
Bustelo XR, Crespo P, Fernández-Pisonero I, and Rodríguez-Fdez S
- Subjects
- Embryonic Development, Germ Cells metabolism, Humans, Mutation genetics, Signal Transduction genetics, ras Proteins genetics, Disease, ras Proteins metabolism
- Abstract
Deregulated RAS signaling is associated with increasing numbers of congenital diseases usually referred to as RASopathies. The spectrum of genes and mutant alleles causing these diseases has been significantly expanded in recent years. This progress has triggered new challenges, including the origin and subsequent selection of the mutations driving these diseases, the specific pathobiological programs triggered by those mutations, the type of correlations that exist between the genotype and the clinical features of patients, and the ancillary genetic factors that influence the severity of the disease in patients. These issues also directly impinge on the feasibility of using RAS pathway drugs to treat RASopathy patients. Here, we will review the main developments and pending challenges in this research topic., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
240. RAS at the Golgi antagonizes malignant transformation through PTPRκ-mediated inhibition of ERK activation.
- Author
-
Casar B, Badrock AP, Jiménez I, Arozarena I, Colón-Bolea P, Lorenzo-Martín LF, Barinaga-Rementería I, Barriuso J, Cappitelli V, Donoghue DJ, Bustelo XR, Hurlstone A, and Crespo P
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Gene Knockdown Techniques, Humans, MAP Kinase Signaling System physiology, Mice, NIH 3T3 Cells, RNA, Small Interfering metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 2 genetics, Zebrafish, Zebrafish Proteins metabolism, Cell Transformation, Neoplastic pathology, Golgi Apparatus metabolism, Melanoma pathology, Receptor-Like Protein Tyrosine Phosphatases, Class 2 metabolism, ras Proteins metabolism
- Abstract
RAS GTPases are frequently mutated in human cancer. H- and NRAS isoforms are distributed over both plasma-membrane and endomembranes, including the Golgi complex, but how this organizational context contributes to cellular transformation is unknown. Here we show that RAS at the Golgi is selectively activated by apoptogenic stimuli and antagonizes cell survival by suppressing ERK activity through the induction of PTPRκ, which targets CRAF for dephosphorylation. Consistently, in contrast to what occurs at the plasma-membrane, RAS at the Golgi cannot induce melanoma in zebrafish. Inactivation of PTPRκ, which occurs frequently in human melanoma, often coincident with TP53 inactivation, accelerates RAS-ERK pathway-driven melanomagenesis in zebrafish. Likewise, tp53 disruption in zebrafish facilitates oncogenesis driven by RAS from the Golgi complex. Thus, RAS oncogenic potential is strictly dependent on its sublocalization, with Golgi complex-located RAS antagonizing tumor development.
- Published
- 2018
- Full Text
- View/download PDF
241. Protein-Protein Interactions: Emerging Oncotargets in the RAS-ERK Pathway.
- Author
-
García-Gómez R, Bustelo XR, and Crespo P
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Humans, MAP Kinase Signaling System drug effects, Neoplasms drug therapy, Extracellular Signal-Regulated MAP Kinases metabolism, Neoplasms metabolism, ras Proteins metabolism
- Abstract
Given the implication of aberrant RAS-extracellular signal-regulated kinase (ERK) signaling in the development of a large number of tumor types, this route is under intense scrutiny to identify new anticancer drugs. Most avenues in that direction have been primarily focused on the inhibition of the catalytic activity of the kinases that participate in this pathway. Although promising, the efficacy of these therapies is short lived due to undesired toxicity and/or drug resistance problems. As an alternative path, new efforts are now being devoted to the targeting of protein-protein interactions (PPIs) involved in the flow of RAS-ERK signals. Many of these efforts have shown promising results in preclinical models. In this review, we summarize recent progress made in this area., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
242. Correction for Agudo-Ibáñez et al., "H-Ras Distribution and Signaling in Plasma Membrane Microdomains Are Regulated by Acylation and Deacylation Events".
- Author
-
Agudo-Ibáñez L, Herrero A, Barbacid M, and Crespo P
- Published
- 2018
- Full Text
- View/download PDF
243. Phosphorylation of p38 by GRK2 at the Docking Groove Unveils a Novel Mechanism for Inactivating p38MAPK.
- Author
-
Peregrin S, Jurado-Pueyo M, Campos PM, Sanz-Moreno V, Ruiz-Gomez A, Crespo P, Mayor F Jr, and Murga C
- Published
- 2018
- Full Text
- View/download PDF
244. Analysis of Ras/ERK Compartmentalization by Subcellular Fractionation.
- Author
-
Agudo-Ibañez L, Crespo P, and Casar B
- Subjects
- Animals, Cell Line, Cell Nucleus metabolism, Endoplasmic Reticulum metabolism, Extracellular Signal-Regulated MAP Kinases isolation & purification, Intracellular Space metabolism, Membrane Microdomains metabolism, Protein Transport, Subcellular Fractions, ras Proteins isolation & purification, Cell Fractionation methods, Extracellular Signal-Regulated MAP Kinases metabolism, ras Proteins metabolism
- Abstract
A vast number of stimuli use the Ras/Raf/MEK/ERK signaling cascade to transmit signals from their cognate receptors, in order to regulate multiple cellular functions, including key processes such as proliferation, cell cycle progression, differentiation, and survival. The duration, intensity and specificity of the responses are, in part, controlled by the compartmentalization/subcellular localization of the signaling intermediaries. Ras proteins are found in different plasma membrane microdomains and endomembranes. At these localizations, Ras is subject to site-specific regulatory mechanisms, distinctively engaging effector pathways and switching-on diverse genetic programs to generate a multitude of biological responses. The Ras effector pathway leading to ERKs activation is also subject to space-related regulatory processes. About half of ERK1/2 substrates are found in the nucleus and function mainly as transcription factors. The other half resides in the cytosol and other cellular organelles. Such subcellular distribution enhances the complexity of the Ras/ERK cascade and constitutes an essential mechanism to endow variability to its signals, which enables their participation in the regulation of a broad variety of functions. Thus, analyzing the subcellular compartmentalization of the members of the Ras/ERK cascade constitutes an important factor to be taken into account when studying specific biological responses evoked by Ras/ERK signals. Herein, we describe methods for such purpose.
- Published
- 2017
- Full Text
- View/download PDF
245. Lysine methylation in cancer: SMYD3-MAP3K2 teaches us new lessons in the Ras-ERK pathway.
- Author
-
Colón-Bolea P and Crespo P
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma pathology, Antineoplastic Agents therapeutic use, Histone-Lysine N-Methyltransferase genetics, Humans, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, MAP Kinase Kinase Kinase 2, MAP Kinase Kinase Kinases genetics, Methylation, Mitogen-Activated Protein Kinase 1 genetics, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 genetics, Mitogen-Activated Protein Kinase 3 metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Protein Binding, Protein Kinase Inhibitors therapeutic use, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Signal Transduction, Adenocarcinoma metabolism, Gene Expression Regulation, Neoplastic, Histone-Lysine N-Methyltransferase metabolism, Lung Neoplasms metabolism, Lysine metabolism, MAP Kinase Kinase Kinases metabolism, Pancreatic Neoplasms metabolism
- Abstract
Lysine methylation has been traditionally associated with histones and epigenetics. Recently, lysine methyltransferases and demethylases - which are involved in methylation of non-histone substrates - have been frequently found deregulated in human tumours. In this realm, a new discovery has unveiled the methyltransferase SMYD3 as an enhancer of Ras-driven cancer. SMYD3 is up-regulated in different types of tumours. SMYD3-mediated methylation of MAP3K2 increases mutant K-Ras-induced activation of ERK1/2. Methylation of MAP3K2 prevents it from binding to the phosphatase PP2A, thereby impeding the impact of this negative regulator on Ras-ERK1/2 signals, leading to the formation of lung and pancreatic adenocarcinomas. Furthermore, depletion of SMYD3 synergises with a MEK inhibitor, currently in clinical trials, to block Ras-driven pancreatic neoplasia. These results underscore the importance of lysine methylation in the regulation of signalling pathways relevant for tumourigenesis and endorse the development of drugs targeting unregulated lysine methylation as therapeutic agents in the struggle against cancer., (© 2014 WILEY Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
246. Mxi2 sustains ERK1/2 phosphorylation in the nucleus by preventing ERK1/2 binding to phosphatases.
- Author
-
Casar B, Rodríguez J, Gibor G, Seger R, and Crespo P
- Subjects
- Animals, COS Cells, Cell Line, Cell Nucleus metabolism, Chlorocebus aethiops, Cytoplasm metabolism, Dogs, HEK293 Cells, Humans, Phosphorylation, Protein Tyrosine Phosphatases metabolism, Signal Transduction, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 14 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Phosphoric Monoester Hydrolases metabolism
- Abstract
ERK1/2 (extracellular-signal-regulated kinase 1/2) MAPKs (mitogen-activated protein kinases) are tightly regulated by the cellular microenvironment in which they operate. Mxi2 is a p38α splice isoform capable of binding to ERK1/2 and ensuring their translocation to the nucleus. Therein Mxi2 sustains ERK1/2 phosphorylation levels and, as a consequence, ERK1/2 nuclear signals are enhanced. However, the molecular mechanisms underlying this process are still unclear. In the present study, we show that Mxi2 prevents nuclear but not cytoplasmic phosphatases from binding to and dephosphorylating ERK1/2, disclosing an unprecedented mechanism for the spatial regulation of ERK1/2 activation. We also demonstrate that the kinetics of ERK1/2 extranuclear signals can be significantly altered by artificially tethering Mxi2 to the cytoplasm. In this case, Mxi2 abolishes ERK1/2 inactivation by cytoplasmic phosphatases and potentiates ERK1/2 functions at this compartment. These results highlight Mxi2 as a key spatial regulator of ERK1/2 functions, playing a pivotal role in the balance between ERK1/2 nuclear and cytoplasmic signals.
- Published
- 2012
- Full Text
- View/download PDF
247. Ras, an actor on many stages: posttranslational modifications, localization, and site-specified events.
- Author
-
Arozarena I, Calvo F, and Crespo P
- Abstract
Among the wealth of information that we have gathered about Ras in the past decade, the introduction of the concept of space in the field has constituted a major revolution that has enabled many pieces of the Ras puzzle to fall into place. In the early days, it was believed that Ras functioned exclusively at the plasma membrane. Today, we know that within the plasma membrane, the 3 Ras isoforms-H-Ras, K-Ras, and N-Ras-occupy different microdomains and that these isoforms are also present and active in endomembranes. We have also discovered that Ras proteins are not statically associated with these localizations; instead, they traffic dynamically between compartments. And we have learned that at these localizations, Ras is under site-specific regulatory mechanisms, distinctively engaging effector pathways and switching on diverse genetic programs to generate different biological responses. All of these processes are possible in great part due to the posttranslational modifications whereby Ras proteins bind to membranes and to regulatory events such as phosphorylation and ubiquitination that Ras is subject to. As such, space and these control mechanisms act in conjunction to endow Ras signals with an enormous signal variability that makes possible its multiple biological roles. These data have established the concept that the Ras signal, instead of being one single, homogeneous entity, results from the integration of multiple, site-specified subsignals, and Ras has become a paradigm of how space can differentially shape signaling.
- Published
- 2011
- Full Text
- View/download PDF
248. New druggable targets in the Ras pathway?
- Author
-
Matallanas D and Crespo P
- Subjects
- Cell Proliferation drug effects, Humans, Models, Biological, Mutation, Neoplasms genetics, Neoplasms metabolism, Signal Transduction genetics, ras Proteins genetics, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Signal Transduction drug effects, ras Proteins metabolism
- Abstract
Ras proteins are key elements in the regulation of cellular proliferation, differentiation and survival. Mutational activation of Ras or of components of its effector pathways are detected in one-third of human cancers and are essential for the genesis and maintenance of the tumoral phenotype. Research efforts have been dedicated to the development of therapeutic agents that inhibit aberrant Ras signals and, subsequently, tumor progression. However, many of these initiatives have proven less successful than expected. This review summarizes the current status of developments in Ras research, the challenges that have arisen during preclinical and clinical stages, and how novel approaches to targeting Ras pathways have introduced new strategies toward the development of antitumoral agents that are alternative or complementary to those currently in use. These new approaches would be aimed at disrupting key protein-protein interactions that are essential for the conveyance of Ras aberrant signals or would be directed against new proteins recently demonstrated to be critical participants in Ras-regulated pathways.
- Published
- 2010
249. Activation of Ras and Rho GTPases and MAP Kinases by G-protein-coupled receptors.
- Author
-
Chiariello M, Vaqué JP, Crespo P, and Gutkind JS
- Subjects
- Amino Acid Motifs, Antibodies immunology, Antibody Specificity, Blotting, Western, Cell Line, Conserved Sequence, Enzyme Activation, Guanosine Triphosphate metabolism, Humans, Mitogen-Activated Protein Kinases chemistry, Mitogen-Activated Protein Kinases immunology, Phosphoproteins immunology, Phosphorylation, Mitogen-Activated Protein Kinases metabolism, Receptors, G-Protein-Coupled metabolism, ras Proteins metabolism, rho GTP-Binding Proteins metabolism
- Abstract
A complex intracellular signaling network mediates the multiple biological activities of G-protein-coupled receptors (GPCRs). Among them, monomeric GTPases and a family of closely related proline-targeted serine-threonine kinases, collectively known as Mitogen-Activated Protein Kinases (MAPKs), appears to play central roles in orchestrating the proliferative responses to multiple mitogens that act on GPCRs. Upon GDP/GTP exchange, monomeric GTPases control the phosphorylation of conserved threonine and tyrosine residues in MAPKs by their immediate upstream kinases, increasing their enzymatic activity and inducing their translocation to the nucleus where they phosphorylate transcription factors, thereby regulating the expression of genes playing a key role in normal and aberrant cell growth. Recently, a number of GPCRs have been engineered to provide exclusive activation by synthetic drug-like compounds while becoming insensitive to endogenous ligands. These engineered receptors, named Receptors Activated Solely by Synthetic Ligands (RASSLs), promise better understanding of GPCRs signaling in vitro and in vivo, thus representing ideal tools to selectively modulate MAPK signaling routes controlling a wide range of biological functions, from proliferation to differentiation, migration, invasion, and cell survival or death by apoptosis.
- Published
- 2010
- Full Text
- View/download PDF
250. Analysis of ERKs' dimerization by electrophoresis.
- Author
-
Pinto A and Crespo P
- Subjects
- Blotting, Western, Electrophoresis, Polyacrylamide Gel, HEK293 Cells, HeLa Cells, Humans, Protein Structure, Quaternary, Electrophoresis methods, Extracellular Signal-Regulated MAP Kinases chemistry, Extracellular Signal-Regulated MAP Kinases metabolism, Protein Multimerization
- Abstract
Signals transmitted by ERK MAP Kinases regulate the functions of multiple substrates present in the nucleus and the cytoplasm. Once phosphorylated, ERKs dimerize. The functions of these dimers had remained elusive until recently when we demonstrated that ERK dimers are assembled using scaffolds proteins as platforms. Dimerization is critical for connecting the scaffolded ERK complex to cognate cytoplasmic substrates. Contrarily, nuclear substrates associate to ERK monomers. These results identify dimerization as a key determinant of the spatial specificity of ERK signals. Moreover, we showed that preventing ERK dimerization, without affecting ERK phosphorylation, is sufficient for attenuating cellular proliferation, transformation, and tumor development. Thus, analyzing ERK dimerization will be an important factor in the future for determining, for example, the real impact on the ERK pathway of some drugs that do not affect ERK phosphorylation. Herein, we describe user-friendly methods for such purpose.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.