Network


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

Hotspot


Dive into the research topics where Jaime Renart is active.

Publication


Featured researches published by Jaime Renart.


Infection and Immunity | 2000

Predictive Value of Nuclear Factor κB Activity and Plasma Cytokine Levels in Patients with Sepsis

Francisco Arnalich; Esther García-Palomero; Julia López; Manuel Jiménez; Rosario Madero; Jaime Renart; Juan José Vázquez; Carmen Montiel

ABSTRACT The relationship between fluctuating cytokine concentrations in plasma and the outcome of sepsis is complex. We postulated that early measurement of the activation of nuclear factor κB (NF-κB), a transcriptional regulatory protein involved in proinflammatory cytokine expression, may help to predict the outcome of sepsis. We determined NF-κB activation in peripheral blood mononuclear cells of 34 patients with severe sepsis (23 survivors and 11 nonsurvivors) and serial concentrations of inflammatory cytokines (interleukin-6, interleukin-1, and tumor necrosis factor) and various endogenous antagonists in plasma. NF-κB activity was significantly higher in nonsurvivors and correlated strongly with the severity of illness (APACHE II score), although neither was related to the cytokine levels. Apart from NF-κB activity, the interleukin-1 receptor antagonist was the only cytokine tested whose level in plasma was of value in predicting mortality by logistic regression analysis. These results underscore the prognostic value of early measurement of NF-κB activity in patients with severe sepsis.


Journal of Biological Chemistry | 2006

Interleukin-1β Enhances GABAA Receptor Cell-surface Expression by a Phosphatidylinositol 3-Kinase/Akt Pathway RELEVANCE TO SEPSIS-ASSOCIATED ENCEPHALOPATHY

Rocío Serantes; Francisco Arnalich; Maria E. Figueroa; Marta Salinas; Eva Andrés-Mateos; Rosa Codoceo; Jaime Renart; Carlos Matute; Carmen Cavada; Antonio Cuadrado; Carmen Montiel

Sepsis-associated encephalopathy (SAE) is a frequent but poorly understood neurological complication in sepsis that negatively influences survival. Here we present clinical and experimental evidence that this brain dysfunction may be related to altered neurotransmission produced by inflammatory mediators. Compared with septic patients, SAE patients had higher interleukin-1β (IL-1β) plasma levels; interestingly, these levels decreased once the encephalopathy was resolved. A putative IL-1β effect on type A γ-aminobutyric acid receptors (GABAARs), which mediate fast synaptic transmission in most cerebral inhibitory synapses in mammals, was investigated in cultured hippocampal neurons and in Xenopus oocytes expressing native or foreign rat brain GABAARs, respectively. Confocal images in both cell types revealed that IL-1β increases recruitment of GABAARs to the cell surface. Moreover, brief applications of IL-1β to voltage-clamped oocytes yielded a delayed potentiation of the GABA-elicited chloride currents (IGABA); this effect was suppressed by IL-1ra, the natural IL-1 receptor (IL-1RI) antagonist. Western blot analysis combined with IGABA recording and confocal images of GABAA Rs in oocytes showed that IL-1β stimulates the IL-1RI-dependent phosphatidylinositol 3-kinase activation and the consequent facilitation of phospho-Akt-mediated insertion of GABAARs into the cell surface. The interruption of this signaling pathway by specific phosphatidylinositol 3-kinase or Akt inhibitors suppresses the cytokine-mediated effects on GABAAR, whereas activation of the conditionally active form of Akt1 (myr-Akt1.ER*) with 4-hydroxytamoxifen reproduces the effects. These findings point to a previously unrecognized signaling pathway that connects IL-1β with increased “GABAergic tone.” We propose that through this mechanism IL-1β might alter synaptic strength at central GABAergic synapses and so contribute to the cognitive dysfunction observed in SAE.


Journal of Biological Chemistry | 2011

Function of partially duplicated human α77 nicotinic receptor subunit CHRFAM7A gene: potential implications for the cholinergic anti-inflammatory response.

Ana M. de Lucas-Cerrillo; M. Constanza Maldifassi; Francisco Arnalich; Jaime Renart; Gema Atienza; Rocío Serantes; Jesús Cruces; Aurora Sánchez-Pacheco; Eva Andrés-Mateos; Carmen Montiel

The neuronal α7 nicotinic receptor subunit gene (CHRNA7) is partially duplicated in the human genome forming a hybrid gene (CHRFAM7A) with the novel FAM7A gene. The hybrid gene transcript, dupα7, has been identified in brain, immune cells, and the HL-60 cell line, although its translation and function are still unknown. In this study, dupα7 cDNA has been cloned and expressed in GH4C1 cells and Xenopus oocytes to study the pattern and functional role of the expressed protein. Our results reveal that dupα7 transcript was natively translated in HL-60 cells and heterologously expressed in GH4C1 cells and oocytes. Injection of dupα7 mRNA into oocytes failed to generate functional receptors, but when co-injected with α7 mRNA at α7/dupα7 ratios of 5:1, 2:1, 1:1, 1:5, and 1:10, it reduced the nicotine-elicited α7 current generated in control oocytes (α7 alone) by 26, 53, 75, 93, and 94%, respectively. This effect is mainly due to a reduction in the number of functional α7 receptors reaching the oocyte membrane, as deduced from α-bungarotoxin binding and fluorescent confocal assays. Two additional findings open the possibility that the dominant negative effect of dupα7 on α7 receptor activity observed in vitro could be extrapolated to in vivo situations. (i) Compared with α7 mRNA, basal dupα7 mRNA levels are substantial in human cerebral cortex and higher in macrophages. (ii) dupα7 mRNA levels in macrophages are down-regulated by IL-1β, LPS, and nicotine. Thus, dupα7 could modulate α7 receptor-mediated synaptic transmission and cholinergic anti-inflammatory response.The neuronal α7 nicotinic receptor subunit gene (CHRNA7) is partially duplicated in the human genome forming a hybrid gene (CHRFAM7A) with the novel FAM7A gene. The hybrid gene transcript, dupα7, has been identified in brain, immune cells, and the HL-60 cell line, although its translation and function are still unknown. In this study, dupα7 cDNA has been cloned and expressed in GH4C1 cells and Xenopus oocytes to study the pattern and functional role of the expressed protein. Our results reveal that dupα7 transcript was natively translated in HL-60 cells and heterologously expressed in GH4C1 cells and oocytes. Injection of dupα7 mRNA into oocytes failed to generate functional receptors, but when co-injected with α7 mRNA at α7/dupα7 ratios of 5:1, 2:1, 1:1, 1:5, and 1:10, it reduced the nicotine-elicited α7 current generated in control oocytes (α7 alone) by 26, 53, 75, 93, and 94%, respectively. This effect is mainly due to a reduction in the number of functional α7 receptors reaching the oocyte membrane, as deduced from α-bungarotoxin binding and fluorescent confocal assays. Two additional findings open the possibility that the dominant negative effect of dupα7 on α7 receptor activity observed in vitro could be extrapolated to in vivo situations. (i) Compared with α7 mRNA, basal dupα7 mRNA levels are substantial in human cerebral cortex and higher in macrophages. (ii) dupα7 mRNA levels in macrophages are down-regulated by IL-1β, LPS, and nicotine. Thus, dupα7 could modulate α7 receptor-mediated synaptic transmission and cholinergic anti-inflammatory response.


FEBS Letters | 2000

Greater diversity than previously thought of chromaffin cell Ca2+ channels, derived from mRNA identification studies

Esther García-Palomero; Inmaculada Cuchillo-Ibáñez; Antonio G. García; Jaime Renart; Almudena Albillos; Carmen Montiel

Using reverse transcription followed by PCR amplification (RT‐PCR), we have identified multiple messenger RNAs encoding for the neuronal pore‐forming Ca2+ channel subunits α1A (P/Q channel), α1B (N channel), α1D (neuronal/endocrine L channel), α1E (R channel), α1G‐H (T channel) and α1S (skeletal muscle L channel) in bovine chromaffin cells. mRNAs for the auxiliary β2, β3, β4, α2/δ and γ2 subunits were also identified. In agreement with these molecular data, perforated patch‐clamp recordings of whole‐cell Ca2+ currents reveal the existence of functional R‐type Ca2+ channels in these cells that were previously undetected with other techniques. Our results provide a molecular frame for a much wider functional diversity of Ca2+ channels in chromaffin cells than that previously established using pharmacological and electrophysiological approaches.


FEBS Letters | 2001

Opposite effects of the Hsp90 inhibitor Geldanamycin: induction of apoptosis in PC12, and differentiation in N2A cells

M.Dolores López-Maderuelo; Margarita Fernández-Renart; Carmen Moratilla; Jaime Renart

The inhibitor of the Hsp90 chaperone Geldanamycin has been reported to have several cellular effects, such as inhibition of v‐src activity or destabilization of Raf‐1 among others. We show now that Geldanamycin treatment induces different phenotypes in different cell lines. In PC12 cells, it triggers apoptosis, whereas in the murine neuroblastoma N2A, it induces differentiation with neurite outgrowth. Geldanamycin effects cannot be mimicked by inhibition of the c‐src protein tyrosine kinases, and nerve growth factor does not protect PC12 cells from apoptosis. Mitogen‐activated protein kinase activities ERK and JNK are activated differently according to cell type: in PC12 cells JNK is activated, and its inhibition abolishes apoptosis, but not ERK; in N2A cells, both ERK and JNK are activated, but with peak activities at different times.


Nucleic Acids Research | 2011

Regulation of SNAIL1 and E-cadherin function by DNMT1 in a DNA methylation-independent context

Jesús Espada; Héctor Peinado; Lidia Lopez-Serra; Fernando Setién; Paula Lopez-Serra; Anna Portela; Jaime Renart; Elisa Carrasco; María Isabel Calvo; Angeles Juarranz; Amparo Cano; Manel Esteller

Mammalian DNA methyltransferase 1 (DNMT1) is essential for maintaining DNA methylation patterns after cell division. Disruption of DNMT1 catalytic activity results in whole genome cytosine demethylation of CpG dinucleotides, promoting severe dysfunctions in somatic cells and during embryonic development. While these observations indicate that DNMT1-dependent DNA methylation is required for proper cell function, the possibility that DNMT1 has a role independent of its catalytic activity is a matter of controversy. Here, we provide evidence that DNMT1 can support cell functions that do not require the C-terminal catalytic domain. We report that PCNA and DMAP1 domains in the N-terminal region of DNMT1 are sufficient to modulate E-cadherin expression in the absence of noticeable changes in DNA methylation patterns in the gene promoters involved. Changes in E-cadherin expression are directly associated with regulation of β-catenin-dependent transcription. Present evidence suggests that the DNMT1 acts on E-cadherin expression through its direct interaction with the E-cadherin transcriptional repressor SNAIL1.


The International Journal of Biochemistry & Cell Biology | 2011

The transmembrane domain of podoplanin is required for its association with lipid rafts and the induction of epithelial-mesenchymal transition

Beatriz Fernández-Muñoz; María M. Yurrita; Ester Martín-Villar; Patricia Carrasco-Ramírez; Diego Megías; Jaime Renart; Miguel Quintanilla

Podoplanin is a transmembrane glycoprotein that is upregulated in cancer and was reported to induce an epithelial-mesenchymal transition (EMT) in MDCK cells. The promotion of EMT was dependent on podoplanin binding to ERM (ezrin, radixin, moesin) proteins through its cytoplasmic (CT) domain, which led to RhoA-associated kinase (ROCK)-dependent ERM phosphorylation. Using detergent-resistant membrane (DRM) assays, as well as transmembrane (TM) interactions and ganglioside GM1 binding, we present evidence supporting the localization of podoplanin in raft platforms important for cell signalling. Podoplanin mutant constructs harbouring a heterologous TM region or lacking the CT tail were unable to associate with DRMs, stimulate ERM phosphorylation and promote EMT or cell migration. Similar effects were observed upon disruption of a GXXXG motif within the TM domain, which is involved in podoplanin self-assembly. In contrast, deletion of the extracellular (EC) domain did not affect podoplanin DRM association. Together, these data suggest that both the CT and TM domains are required for podoplanin localization in raft platforms, and that this association appears to be necessary for podoplanin-mediated EMT and cell migration.


Mechanisms of Development | 1976

Characterization and levels of the RNA polymerases during the embryogenesis of Artemia salina

Jaime Renart; Jesús Sebastián

Dormant embryos at the gastrula stage of Artemia salina contain three DNA-dependent RNA polymerases: I, II, and III. The enzymes are solubilized from whole embryos and they are separated by chromatography on DEAE Sephadex. The ratio of activities with native and denatured DNA at the optimal salt concentrations is 3.5 for RNA polymerase I, 0.1 for RNA polymerase II and 1.0 for RNA polymerase III.Mn(i2+) is more efficient than Mg(2+) for the three enzymes. RNA polymerase II is 50% inhibited by 5 ng/ml of alpha-amanitin while RNA polymerases I and III are 10% and 30% inhibited by 1 mg/ml. During the embryonic development there is am independent variation of the levels of the RNA polymerases. RNA polymerase I increases its specific activity 4-5-fold, RNA polymerase III increases 2-fold, and RNA polymerase II less than 2-fold. The increase in RNA polymerase activity may represent a mechanism to control the rate of synthesis of RNA during the embryogenesis of A. salina.


Oncotarget | 2016

Podoplanin is a component of extracellular vesicles that reprograms cell-derived exosomal proteins and modulates lymphatic vessel formation

Patricia Carrasco-Ramírez; David W. Greening; Germán Andrés; Shashi K. Gopal; Ester Martín-Villar; Jaime Renart; Richard J. Simpson; Miguel Quintanilla

Podoplanin (PDPN) is a transmembrane glycoprotein that plays crucial roles in embryonic development, the immune response, and malignant progression. Here, we report that cells ectopically or endogenously expressing PDPN release extracellular vesicles (EVs) that contain PDPN mRNA and protein. PDPN incorporates into membrane shed microvesicles (MVs) and endosomal-derived exosomes (EXOs), where it was found to colocalize with the canonical EV marker CD63 by immunoelectron microscopy. We have previously found that expression of PDPN in MDCK cells induces an epithelial-mesenchymal transition (EMT). Proteomic profiling of MDCK-PDPN cells compared to control cells shows that PDPN-induced EMT is associated with upregulation of oncogenic proteins and diminished expression of tumor suppressors. Proteomic analysis of exosomes reveals that MDCK-PDPN EXOs were enriched in protein cargos involved in cell adhesion, cytoskeletal remodeling, signal transduction and, importantly, intracellular trafficking and EV biogenesis. Indeed, expression of PDPN in MDCK cells stimulated both EXO and MV production, while knockdown of endogenous PDPN in human HN5 squamous carcinoma cells reduced EXO production and inhibited tumorigenesis. EXOs released from MDCK-PDPN and control cells both stimulated in vitro angiogenesis, but only EXOs containing PDPN were shown to promote lymphatic vessel formation. This effect was mediated by PDPN on the surface of EXOs, as demonstrated by a neutralizing specific monoclonal antibody. These results contribute to our understanding of PDPN-induced EMT in association to tumor progression, and suggest an important role for PDPN in EV biogenesis and/or release and for PDPN-EXOs in modulating lymphangiogenesis.


Oncogene | 2015

Podoplanin mediates ECM degradation by squamous carcinoma cells through control of invadopodia stability.

Ester Martín-Villar; B. Borda-d'Agua; Patricia Carrasco-Ramírez; Jaime Renart; Madeline Parsons; Miguel Quintanilla; Gareth E. Jones

Invadopodia are actin-rich cell membrane projections used by invasive cells to penetrate the basement membrane. Control of invadopodia stability is critical for efficient degradation of the extracellular matrix (ECM); however, the underlying molecular mechanisms remain poorly understood. Here, we uncover a new role for podoplanin, a transmembrane glycoprotein closely associated with malignant progression of squamous cell carcinomas (SCCs), in the regulation of invadopodia-mediated matrix degradation. Podoplanin downregulation in SCC cells impairs invadopodia stability, thereby reducing the efficiency of ECM degradation. We report podoplanin as a novel component of invadopodia-associated adhesion rings, where it clusters prior to matrix degradation. Early podoplanin recruitment to invadopodia is dependent on lipid rafts, whereas ezrin/moesin proteins mediate podoplanin ring assembly. Finally, we demonstrate that podoplanin regulates invadopodia maturation by acting upstream of the ROCK-LIMK-Cofilin pathway through the control of RhoC GTPase activity. Thus, podoplanin has a key role in the regulation of invadopodia function in SCC cells, controlling the initial steps of cancer cell invasion.

Collaboration


Dive into the Jaime Renart's collaboration.

Top Co-Authors

Avatar

Miguel Quintanilla

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Carmen Montiel

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Ester Martín-Villar

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Jesús Cruces

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Francisco Arnalich

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Margarita Díaz-Guerra

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Antonio G. García

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Esther García-Palomero

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Eva Andrés-Mateos

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Gema Atienza

Autonomous University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge