Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Cesáreo Roncero is active.

Publication


Featured researches published by Cesáreo Roncero.


PLOS ONE | 2013

BMP9 Is a Proliferative and Survival Factor for Human Hepatocellular Carcinoma Cells

Blanca Herrera; María García-Álvaro; Silvia de la Cruz; Peter Walsh; Margarita Fernández; Cesáreo Roncero; Isabel Fabregat; Aránzazu Sánchez; Gareth J. Inman

TGF-β family members play a relevant role in tumorigenic processes, including hepatocellular carcinoma (HCC), but a specific implication of the Bone Morphogenetic Protein (BMP) subfamily is still unknown. Although originally isolated from fetal liver, little is known about BMP9, a BMP family member, and its role in liver physiology and pathology. Our results show that BMP9 promotes growth in HCC cells, but not in immortalized human hepatocytes. In the liver cancer cell line HepG2, BMP9 triggers Smad1,5,8 phosphorylation and inhibitor of DNA binding 1 (Id1) expression up- regulation. Importantly, by using chemical inhibitors, ligand trap and gene silencing approaches we demonstrate that HepG2 cells autocrinely produce BMP9 that supports their proliferation and anchorage independent growth. Additionally, our data reveal that in HepG2 cells BMP9 triggers cell cycle progression, and strikingly, completely abolishes the increase in the percentage of apoptotic cells induced by long-term incubation in low serum. Collectively, our data unveil a dual role for BMP9, both promoting a proliferative response and exerting a remarkable anti-apoptotic function in HepG2 cells, which result in a robust BMP9 effect on liver cancer cell growth. Finally, we show that BMP9 expression is increased in 40% of human HCC tissues compared with normal human liver as revealed by immunohistochemistry analysis, suggesting that BMP9 signaling may be relevant during hepatocarcinogenesis in vivo. Our findings provide new clues for a better understanding of BMPs contribution, and in particular BMP9, in HCC pathogenesis that may result in the development of effective and targeted therapeutic interventions.


Journal of Neurochemistry | 2010

Transient focal cerebral ischemia significantly alters not only EAATs but also VGLUTs expression in rats: relevance of changes in reactive astroglia.

Eduardo Sanchez-Mendoza; María C. Burguete; María Castelló-Ruiz; M.P. González; Cesáreo Roncero; Juan B. Salom; Carmen Arce; Sixta Cañadas; Germán Torregrosa; Enrique Alborch; María Jesús Oset-Gasque

J. Neurochem. (2010) 113, 1343–1355.


Journal of Neurochemistry | 2009

Signaling mechanisms of interferon gamma induced apoptosis in chromaffin cells: involvement of nNOS, iNOS, and NFκB

Rocío Pérez-Rodríguez; Cesáreo Roncero; Ana María Oliván; María del Pilar González; María Jesús Oset-Gasque

Previous work of our group stated that exogenously added and endogenous nitric oxide (NO) generated by cytokines induce apoptosis in chromaffin cells. In this work, we investigate the specific regulation of the NO synthase (NOS) isoforms, inducible NOS (iNOS) and neuronal NOS (nNOS), and their particular participation in cell death induced by interferon gamma (IFNγ). Lipopolysaccharide (LPS) and IFNγ increase iNOS expression, with no effect on nNOS expression. On the other hand, dexamethasone increases basal nNOS expression but decreases LPS + IFNγ‐induced iNOS expression. IFNγ‐induced cell death was abolished by W‐1400, a specific iNOS inhibitor, but only partially by nNOS inhibitors [N‐ω‐propyl‐l‐arginine (N‐PLA), 3‐Bromo‐7‐nitroindazol (7‐NI), l‐methyl thiocitrulline and N‐methyl l‐arginine], indicating the main iNOS participation in chromaffin cell death. IFNγ and LPS induce nuclear factor κB (NFκB) translocation to the nucleus, a process implicated in activation of iNOS expression, as inhibition of NFκB translocation, by SN50, decreased iNOS expression. In addition, IFNγ and LPS induce 847Ser‐nNOS phosphorylation, inhibiting nNOS activity. Both processes, nNOS phosphorylation and iNOS expression induced by LPS + IFNγ, are regulated by Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway, as IFNγ increases 727STAT‐3 phosphorylation and specific inhibitors of JAK/STAT pathway, such as AG490, inhibited both processes. Taken together, these results support the hypothesis of an inactivating phosphorylation of nNOS by IFNγ, via JAK/STAT, in bovine chromaffin cells. Low NO concentrations achieved by this event, would activate NFκB translocation, increasing iNOS expression and generating, this last, high apoptotic NO concentrations.


PLOS ONE | 2013

Mouse Hepatic Oval Cells Require Met-Dependent PI3K to Impair TGF-β-Induced Oxidative Stress and Apoptosis

Adoración Martínez-Palacián; Gaelle del Castillo; Amileth Suárez-Causado; María García-Álvaro; Diego de la Morena-Frutos; Margarita Fernández; Cesáreo Roncero; Isabel Fabregat; Blanca Herrera; Aránzazu Sánchez

We have previously shown that oval cells harboring a genetically inactivated Met tyrosine kinase (Met−/− oval cells) are more sensitive to TGF-β-induced apoptosis than cells expressing a functional Met (Metflx/flx), demonstrating that the HGF/Met axis plays a pivotal role in oval cell survival. Here, we have examined the mechanism behind this effect and have found that TGF-β induced a mitochondria-dependent apoptotic cell death in Metflx/flx and Met−/− oval cells, associated with a marked increase in levels of the BH3-only proteins Bim and Bmf. Bmf plays a key role during TGF-β-mediated apoptosis since knocking down of BMF significantly diminished the apoptotic response in Met−/− oval cells. TGF-β also induced oxidative stress accompanied by NADPH oxidase 4 (Nox4) mRNA up-regulation and decreased protein levels of antioxidant enzymes. Antioxidants inhibit both TGF-β-induced caspase 3 activity and Bmf up-regulation, revealing an oxidative stress-dependent Bmf regulation by TGF-β. Notably, oxidative stress-related events were strongly amplified in Met−/− oval cells, emphasizing the critical role of Met in promoting survival. Pharmacological inhibition of PI3K did impair HGF-driven protection from TGF-β-induced apoptosis and increased sensitivity of Metflx/flx oval cells to TGF-ß by enhancing oxidative stress, reaching apoptotic indices similar to those obtained in Met−/− oval cells. Interestingly, both PI3K inhibition and/or knockdown itself resulted in caspase-3 activation and loss of viability in Metflx/flx oval cells, whereas no effect was observed in Met−/− oval cells. Altogether, results presented here provide solid evidences that both paracrine and autocrine HGF/Met signaling requires PI3K to promote mouse hepatic oval cell survival against TGF-β-induced oxidative stress and apoptosis.


Cellular Signalling | 2012

EGFR is dispensable for c-Met-mediated proliferation and survival activities in mouse adult liver oval cells

Adoración Martínez-Palacián; G. del Castillo; Blanca Herrera; Margarita Fernández; Cesáreo Roncero; Isabel Fabregat; Aránzazu Sánchez

Liver progenitor cells rise as potential critical players in hepatic regeneration but also carcinogenesis. It is therefore mandatory to define the signals controlling their activation and expansion. Recently, by using a novel in vitro model of oval cell lines expressing a mutant tyrosine kinase-inactive form of c-Met we demonstrated that autocrine c-Met signalling plays an essential role in promoting oval cell survival. Here, we investigated the significance of the epidermal growth factor receptor (EGFR) signalling in oval cell proliferation and survival, as well as a potential functional crosstalk between the c-Met and the EGFR pathways. We found an autocrine activation of the EGFR-triggered pathway in Met(flx/flx) and Met(-/-) oval cells as judged by constitutive expression of the EGFR ligands, transforming growth factor-alpha (TGF-α) and heparin-binding EGF like growth factor (HB-EGF), and activation of EGFR. On the other hand, treatment with AG1478, a specific inhibitor of EGFR, effectively blocked endogenous and EGF-induced proliferation, while increased serum withdrawal and transforming growth factor-beta (TGF-β)-induced apoptosis. These results suggest that constitutively activated EGFR might promote oval cell proliferation and survival. We found that hepatocyte growth factor (HGF) does not transactivate EGFR nor EGF transactivates c-Met. Furthermore, treatment with AG1478 or EGFR gene silencing did not interfere with HGF-mediated activation of target signals, such as protein kinase B (AKT/PKB), and extracellular signal-regulated kinases 1/2 (ERK 1/2), nor did it have any effect on HGF-induced proliferative and antiapoptotic activities in Met(flx/flx) cells, showing that HGF does not require EGFR activation to mediate such responses. EGF induced proliferation and survival equally in Met(flx/flx) and Met(-/-) oval cells, proving that EGFR signalling does not depend on c-Met tyrosine kinase activity. Together, our results provide strong evidence that in normal, untransformed oval cells, c-Met and EGFR represent critical molecular players to control proliferation and survival that function independent of one another.


Journal of Biological Chemistry | 2015

p38 MAPK down-regulates fibulin 3 expression through methylation of gene regulatory sequences. Role in migration and invasion

María Arechederra; Neibla Priego; Ana Vázquez-Carballo; Celia Sequera; Álvaro Gutiérrez-Uzquiza; María Isabel Cerezo-Guisado; Sara Ortiz-Rivero; Cesáreo Roncero; Ana Cuenda; Carmen Guerrero; Almudena Porras

Background: p38α MAPK regulates migration/invasion. Results: p38α induces hypermethylation of Fibulin 3 gene regulatory sequences leading to Fibulin 3 down-regulation. This contributes to regulate migration and invasion in MEFs and HCT116 cells. Conclusion: p38α down-regulates fibulin 3 expression through promoter methylation to control p38α-mediated migration and invasion. Significance: To understand the function of new p38α targets in migration/invasion and tumorigenesis. p38 MAPKs regulate migration and invasion. However, the mechanisms involved are only partially known. We had previously identified fibulin 3, which plays a role in migration, invasion, and tumorigenesis, as a gene regulated by p38α. We have characterized in detail how p38 MAPK regulates fibulin 3 expression and its role. We describe here for the first time that p38α, p38γ, and p38δ down-regulate fibulin 3 expression. p38α has a stronger effect, and it does so through hypermethylation of CpG sites in the regulatory sequences of the gene. This would be mediated by the DNA methylase, DNMT3A, which is down-regulated in cells lacking p38α, but once re-introduced represses Fibulin 3 expression. p38α through HuR stabilizes dnmt3a mRNA leading to an increase in DNMT3A protein levels. Moreover, by knocking-down fibulin 3, we have found that Fibulin 3 inhibits migration and invasion in MEFs by mechanisms involving p38α/β inhibition. Hence, p38α pro-migratory/invasive effect might be, at least in part, mediated by fibulin 3 down-regulation in MEFs. In contrast, in HCT116 cells, Fibulin 3 promotes migration and invasion through a mechanism dependent on p38α and/or p38β activation. Furthermore, Fibulin 3 promotes in vitro and in vivo tumor growth of HCT116 cells through a mechanism dependent on p38α, which surprisingly acts as a potent inducer of tumor growth. At the same time, p38α limits fibulin 3 expression, which might represent a negative feed-back loop.


Biochimica et Biophysica Acta | 2015

HGF/c-Met signaling promotes liver progenitor cell migration and invasion by an epithelial-mesenchymal transition-independent, phosphatidyl inositol-3 kinase-dependent pathway in an in vitro model.

A. Suárez-Causado; D. Caballero-Díaz; Esther Bertran; Cesáreo Roncero; Annalisa Addante; María García-Álvaro; Margarita Fernández; Blanca Herrera; Almudena Porras; Isabel Fabregat; Aránzazu Sánchez

Oval cells constitute an interesting hepatic cell population. They contribute to sustain liver regeneration during chronic liver damage, but in doing this they can be target of malignant conversion and become tumor-initiating cells and drive hepatocarcinogenesis. The molecular mechanisms beneath either their pro-regenerative or pro-tumorigenic potential are still poorly understood. In this study, we have investigated the role of the HGF/c-Met pathway in regulation of oval cell migratory and invasive properties. Our results show that HGF induces c-Met-dependent oval cell migration both in normal culture conditions and after in vitro wounding. HGF-triggered migration involves F-actin cytoskeleton reorganization, which is also evidenced by activation of Rac1. Furthermore, HGF causes ZO-1 translocation from cell-cell contact sites to cytoplasm and its concomitant activation by phosphorylation. However, no loss of expression of cell-cell adhesion proteins, including E-cadherin, ZO-1 and Occludin-1, is observed. Additionally, migration does not lead to cell dispersal but to a characteristic organized pattern in rows, in turn associated with Golgi compaction, providing strong evidence of a morphogenic collective migration. Besides migration, HGF increases oval cell invasion through extracellular matrix, a process that requires PI3K activation and is at least partly mediated by expression and activation of metalloproteases. Altogether, our findings provide novel insights into the cellular and molecular mechanisms mediating the essential role of HGF/c-Met signaling during oval cell-mediated mouse liver regeneration.


Journal of Neuroscience Research | 2007

Mechanisms of nitric oxide-induced apoptosis in bovine chromaffin cells: Role of mitochondria and apoptotic proteins.

Rocío Pérez-Rodríguez; María P. Fuentes; Anna M. Oliván; Adoración Martínez-Palacián; Cesáreo Roncero; María Dolores Puga González; María Jesús Oset-Gasque

The aim of this work was to establish the possible involvement of mitochondria in the apoptotic event triggered by nitric oxide (NO) in chromaffin cells. Using bovine chromaffin cells in primary culture and several NO donors (SNP, SNAP, and GSNO) at apoptotic concentrations (50 μM–1 mM), we have shown that NO induces a time‐dependent decrease in the mitochondrial transmembrane potential (ΔΨm), which correlates with the appearance of hypodiploid cells. Disruption in ΔΨm is followed by cytochrome c release to the cytosol, which in turn precedes caspase 3 activation. In this mechanism participates the Bcl‐2 protein family, because NO donors downregulate the expression of anti‐apoptotic members of the family such as Bcl‐2 and Bcl‐XL, and increase the expression of pro‐apoptotic members, Bax and Bcl‐Xs, inductors of cytochrome c release to cytosol. Different cell signaling pathways seem to regulate Bax induction and Bcl‐2 inhibition because decreased Bcl‐2 levels are detected later than enhanced Bax expression. The tumour suppressor protein p53 is also upregulated in a very early phase (30 min) of the NO‐induced apoptosis and may be responsible for the further induction of Bax expression. Finally, the translocation of NF‐κB to the nucleus seems to be another early event in NO‐induced apoptosis and it may be involved in the regulation of p53 expression. These results support strongly the participation of mitochondrial mechanisms in NO‐induced apoptosis in chromaffin cells and suggest that these cells may be good models for the investigation of molecular basis of neurodegeneration and neuroprotection.


Free Radical Biology and Medicine | 2014

Glutamate triggers neurosecretion and apoptosis in bovine chromaffin cells through a mechanism involving NO production by neuronal NO synthase activation.

Rocío Pérez-Rodríguez; A.M. Oliván; Cesáreo Roncero; J. Morón-Oset; M.P. González; María Jesús Oset-Gasque

Previous work from our group stated that nitric oxide (NO), via cytokines, induces apoptosis in chromaffin cells by a mechanism involving iNOS, nNOS, and NF-κB. In this paper the involvement of glutamate as a possible intracellular trigger of neurosecretion and NO-mediated apoptosis has been evaluated. We show that chromaffin cells express different ionotropic and metabotropic glutamate receptors, this exerting different effects on the regulation of basal and glutamate-induced catecholamine secretion, via NO/cGMP. In addition, we studied the effects of endogenously generated NO, both basal and glutamate-stimulated, on apoptosis of chromaffin cells. Our results show that glutamate agonists are able to induce cell death and apoptosis in bovine chromaffin cells, parallel to an increase in NO production. Such effects were reversed by NOS inhibitors and glutamate receptor antagonists. Under basal conditions, iNOS inhibitors did not have any effect on apoptosis, whereas nNOS inhibitors induced apoptosis, indicating a neuroprotective effect of constitutive nNOS-generated NO. In contrast, glutamate-induced apoptosis was strongly reversed by nNOS inhibitors and weakly by iNOS inhibitors, thus indicating nNOS involvement in glutamate-mediated apoptosis. These results were confirmed by the fact that nNOS expression, but not iNOS, is specifically activated by glutamate. Finally, our results suggest the participation of PKG, PKA, PKC, and MAPK pathways in glutamate-mediated nNOS activation in chromaffin cells and point out the involvement of both PKA and PKC signaling pathways in the apoptotic effect of glutamate.


Journal of Neuroscience Research | 2011

Plasma membrane and vesicular glutamate transporter expression in chromaffin cells of bovine adrenal medulla.

A.M. Oliván; Rocío Pérez-Rodríguez; Cesáreo Roncero; Carmen Arce; M.P. González; María Jesús Oset-Gasque

The study of the functional expression of glutamate signaling molecules in peripheral tissues has received relatively little attention. However, evidence is increasing for a role of glutamate as an extracellular signal mediator in endocrine systems, in addition to having an excitatory amino acid neurotransmitter role in the CNS. Chromaffin cells are good models of catecholaminergic neurons, in which previous work from our group demonstrated the existence of both functional glutamate receptors and specific exocytotic and nonexocytotic glutamate release. In this work, the presence of specific plasma membrane (EAATs) and vesicular glutamate (VGLUTs) transporters has been investigated by using confocal microscopy, flow cytometric analysis, Western blot, and qRT‐PCR techniques. We found specific expression of EAAT3, EAAT2, VGLUT1, and VGLUT3 in about 95%, 65%, 55%, and 25%, respectively, of the whole chromaffin cell population. However, chromaffin cells do not express VGLUT2 and have a very low expression of EAAT1. VGLUTs are localized mainly in the membrane fraction, and EAATs share their subcellular location between membrane and cytosolic fractions. Their estimated molecular weights were about 70 kDa for EAAT2, about 65 kDa for EAAT3, about 50 kDa for VGLUT1, and about 60 kDa for VGLUT3. RT‐qPCR techniques confirm the expression of these glutamate transporters at the mRNA level and show a different regulation by cytokines and glucocorticoids between VGLUT1 and ‐3 and EAAT2 and ‐3 subfamilies. These interesting results support the participation of these glutamate transporters in the process of glutamate release in chromaffin cells and in the regulation of their neurosecretory function in adrenal medulla.

Collaboration


Dive into the Cesáreo Roncero's collaboration.

Top Co-Authors

Avatar

Aránzazu Sánchez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Blanca Herrera

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Margarita Fernández

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

María García-Álvaro

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Annalisa Addante

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

María Jesús Oset-Gasque

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Laura Almalé

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Rocío Pérez-Rodríguez

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge