Network


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

Hotspot


Dive into the research topics where Miguel F. Segura is active.

Publication


Featured researches published by Miguel F. Segura.


Molecular Pharmacology | 2006

Reactive oxygen species and p38 mitogen-activated protein kinase activate bax to induce mitochondrial cytochrome c release and apoptosis in response to malonate

M. Gomez-Lazaro; Maria F. Galindo; R. M. Melero-Fernandez de Mera; F.J. Fernandez-Gomez; Caoimhín G. Concannon; Miguel F. Segura; Joan X. Comella; Jochen H. M. Prehn; Joaquín Jordán

Malonate, an inhibitor of mitochondrial complex II, is a widely used toxin to study neurodegeneration in Huntingtons disease and ischemic stroke. We have shown previously that malonate increased reactive oxygen species (ROS) production in human SH-SY5Y neuroblastoma cells, leading to oxidative stress, cytochrome c release, and apoptotic cell death. Expression of a green fluorescent protein-Bax fusion protein in SH-SY5Y neuroblastoma cells demonstrated a Bax redistribution from the cytosol to mitochondria after 12 to 24 h of malonate treatment that coincided with mitochondrial potential collapse and chromatin condensation. Inhibition of Bax translocation using furosemide, as well as Bax gene deletion, afforded significant protection against malonate-induced apoptosis. Further experiments revealed that malonate induced a prominent increase in the level of activated p38 mitogen-activated protein (MAP) kinase and that treatment with the p38 MAP kinase inhibitor SKF86002 potently blocked malonate-induced Bax translocation and apoptosis. Treatment with vitamin E diminished ROS production, reduced the activation status of p38 MAP kinase, inhibited Bax translocation, and protected against malonate-induced apoptosis. Our data suggest that malonate-induced ROS production and subsequent p38 MAP kinase activation mediates the activation of the pro-apoptotic Bax protein to induce mitochondrial membrane permeabilization and neuronal apoptosis.


Journal of Neurochemistry | 2008

6‐Hydroxydopamine activates the mitochondrial apoptosis pathway through p38 MAPK‐mediated, p53‐independent activation of Bax and PUMA

M. Gomez-Lazaro; Maria F. Galindo; Caoimhín G. Concannon; Miguel F. Segura; F.J. Fernandez-Gomez; Nuria Llecha; Joan X. Comella; Jochen H. M. Prehn; Joaquín Jordán

J. Neurochem. (2008) 104, 1599–1612.


Cancer Research | 2013

BRD4 sustains melanoma proliferation and represents a new target for epigenetic therapy.

Miguel F. Segura; Barbara Fontanals-Cirera; Avital Gaziel-Sovran; Maria V. Guijarro; Doug Hanniford; Guangtao Zhang; Pilar González-Gómez; Marta Morante; Luz Jubierre; Weijia Zhang; Farbod Darvishian; Michael Ohlmeyer; Iman Osman; Ming-Ming Zhou; Eva Hernando

Metastatic melanoma remains a mostly incurable disease. Although newly approved targeted therapies are efficacious in a subset of patients, resistance and relapse rapidly ensue. Alternative therapeutic strategies to manipulate epigenetic regulators and disrupt the transcriptional program that maintains tumor cell identity are emerging. Bromodomain and extraterminal domain (BET) proteins are epigenome readers known to exert key roles at the interface between chromatin remodeling and transcriptional regulation. Here, we report that BRD4, a BET family member, is significantly upregulated in primary and metastatic melanoma tissues compared with melanocytes and nevi. Treatment with BET inhibitors impaired melanoma cell proliferation in vitro and tumor growth and metastatic behavior in vivo, effects that were mostly recapitulated by individual silencing of BRD4. RNA sequencing of BET inhibitor-treated cells followed by Gene Ontology analysis showed a striking impact on transcriptional programs controlling cell growth, proliferation, cell-cycle regulation, and differentiation. In particular, we found that, rapidly after BET displacement, key cell-cycle genes (SKP2, ERK1, and c-MYC) were downregulated concomitantly with the accumulation of cyclin-dependent kinase (CDK) inhibitors (p21 and p27), followed by cell-cycle arrest. Importantly, BET inhibitor efficacy was not influenced by BRAF or NRAS mutational status, opening the possibility of using these small-molecule compounds to treat patients for whom no effective targeted therapy exists. Collectively, our study reveals a critical role for BRD4 in melanoma tumor maintenance and renders it a legitimate and novel target for epigenetic therapy directed against the core transcriptional program of melanoma.


Journal of Cell Biology | 2004

The death receptor antagonist FAIM promotes neurite outgrowth by a mechanism that depends on ERK and NF-κB signaling

Carme Solé; Xavier Dolcet; Miguel F. Segura; Humberto Gutierrez; Maria-Teresa Diaz-Meco; Raffaella Gozzelino; Daniel Sanchis; Jose R. Bayascas; Carme Gallego; Jorge Moscat; Alun M. Davies; Joan X. Comella

Fas apoptosis inhibitory molecule (FAIM) is a protein identified as an antagonist of Fas-induced cell death. We show that FAIM overexpression fails to rescue neurons from trophic factor deprivation, but exerts a marked neurite growth–promoting action in different neuronal systems. Whereas FAIM overexpression greatly enhanced neurite outgrowth from PC12 cells and sympathetic neurons grown with nerve growth factor (NGF), reduction of endogenous FAIM levels by RNAi decreased neurite outgrowth in these cells. FAIM overexpression promoted NF-κB activation, and blocking this activation by using a super-repressor IκBα or by carrying out experiments using cortical neurons from mice that lack the p65 NF-κB subunit prevented FAIM-induced neurite outgrowth. The effect of FAIM on neurite outgrowth was also blocked by inhibition of the Ras–ERK pathway. Finally, we show that FAIM interacts with both Trk and p75 neurotrophin receptor NGF receptors in a ligand-dependent manner. These results reveal a new function of FAIM in promoting neurite outgrowth by a mechanism involving activation of the Ras–ERK pathway and NF-κB.


Journal of Neurochemistry | 2007

Lifeguard/neuronal membrane protein 35 regulates Fas ligand-mediated apoptosis in neurons via microdomain recruitment

Miriam Fernández; Miguel F. Segura; Carme Solé; Alicia Colino; Joan X. Comella; Valentín Ceña

Fas ligand (FasL)‐receptor system plays an essential role in regulating cell death in the developing nervous system, and it has been implicated in neurodegenerative and inflammatory responses in the CNS. Lifeguard (LFG) is a protein highly expressed in the hippocampus and the cerebellum, and it shows a particularly interesting regulation by being up‐regulated during postnatal development and in the adult. We show that over‐expression of LFG protected cortical neurons from FasL‐induced apoptosis and decreased caspase‐activation. Reduction of endogenous LFG expression by small interfering RNA sensitized cerebellar granular neurons to FasL‐induced cell death and caspase‐8 activation, and also increased sensitivity of cortical neurons. In differentiated cerebellar granular neurons, protection from FasL‐induced cell death could be attributed exclusively to LFG and appears to be independent of FLICE inhibitor protein. Thus, LFG is an endogenous inhibitor of FasL‐mediated neuronal death and it mediates the FasL resistance of CNS differentiated neurons. Finally, we also demonstrate that LFG is detected in lipid rafts microdomains, where it may interact with Fas receptor and regulate FasL‐activated signaling pathways.


The Journal of Neuroscience | 2007

The long form of fas apoptotic inhibitory molecule is expressed specifically in neurons and protects them against death receptor-triggered apoptosis

Miguel F. Segura; Carme Solé; Marta Pascual; Rana S. Moubarak; M. José Pérez-García; Raffaella Gozzelino; Victoria Iglesias; Nahuai Badiola; Jose R. Bayascas; Nuria Llecha; José Rodríguez-Álvarez; Eduardo Soriano; Victor J. Yuste; Joan X. Comella

Death receptors (DRs) and their ligands are expressed in developing nervous system. However, neurons are generally resistant to death induction through DRs and rather their activation promotes neuronal outgrowth and branching. These results suppose the existence of DRs antagonists expressed in the nervous system. Fas apoptosis inhibitory molecule (FAIMS) was first identified as a Fas antagonist in B-cells. Soon after, a longer alternative spliced isoform with unknown function was identified and named FAIML. FAIMS is widely expressed, including the nervous system, and we have shown previously that it promotes neuronal differentiation but it is not an anti-apoptotic molecule in this system. Here, we demonstrate that FAIML is expressed specifically in neurons, and its expression is regulated during the development. Expression could be induced by NGF through the extracellular regulated kinase pathway in PC12 (pheochromocytoma cell line) cells. Contrary to FAIMS, FAIML does not increase the neurite outgrowth induced by neurotrophins and does not interfere with nuclear factor κB pathway activation as FAIMS does. Cells overexpressing FAIML are resistant to apoptotic cell death induced by DRs such as Fas or tumor necrosis factor R1. Reduction of endogenous expression by small interfering RNA shows that endogenous FAIML protects primary neurons from DR-induced cell death. The detailed analysis of this antagonism shows that FAIML can bind to Fas receptor and prevent the activation of the initiator caspase-8 induced by Fas. In conclusion, our results indicate that FAIML could be responsible for maintaining initiator caspases inactive after receptor engagement protecting neurons from the cytotoxic action of death ligands.


Cell Research | 2008

BCL-XL regulates TNF-α-mediated cell death independently of NF-κB, FLIP and IAPs

Raffaella Gozzelino; Carme Solé; Nuria Llecha; Miguel F. Segura; Rana S. Moubarak; Victoria Iglesias-Guimarais; M. José Pérez-García; Stéphanie Reix; Jisheng Zhang; Nahuai Badiola; Daniel Sanchis; José Rodríguez-Álvarez; Ramon Trullas; Victor J. Yuste; Joan X. Comella

Upon activation, tumor necrosis factor alpha (TNF-α) receptor can engage apoptotic or survival pathways. Inhibition of macromolecular synthesis is known to sensitize cells to TNF-α-induced cell death. It is believed that this sensitization is due to the transcriptional blockade of genes regulated by NF-κB. Nevertheless, such evidence has remained elusive in the nervous system. Here, we show that TNF-α cannot normally induce apoptosis in PC12 cells or cortical neurons. However, cells treated with Actinomycin D (ActD) become susceptible to TNF-α-induced cell death through the activation of caspase-8, generation of tBid and activation of caspase-9 and -3. Analysis of several proteins involved in TNF-α receptor signaling showed no significant downregulation of NF-κB target genes, such as IAPs or FLIP, under such conditions. However, Bcl-xL protein levels, but not those of Bcl-2, Bax and Bak, are reduced by ActD or TNF-α/ActD treatments. Moreover, Bcl-xL overexpression fully protects cells against TNF-α/ActD-induced cell death. When endogenous levels of Bcl-xL are specifically downregulated by lentiviral-based RNAi, cells no longer require ActD to be sensitive to TNF-α-triggered apoptosis. Furthermore, Bcl-xL downregulation does not affect TNF-α-mediated NF-κB activation. Altogether, our results demonstrate that Bcl-xL, and not Bcl-2, FLIP or IAPs, acts as the endogenous regulator of neuronal resistance/sensitivity to TNF-α-induced apoptosis in an NF-κB-independent manner.


Oncogene | 2016

BRG1/SMARCA4 is essential for neuroblastoma cell viability through modulation of cell death and survival pathways

Luz Jubierre; Aroa Soriano; Laura Planells-Ferrer; Laia París-Coderch; S P Tenbaum; O A Romero; Rana S. Moubarak; A Almazán-Moga; Carla Molist; Josep Roma; Samuel Navarro; R Noguera; M Sánchez-Céspedes; Joan X. Comella; H G Palmer; J. Sánchez de Toledo; Soledad Gallego; Miguel F. Segura

Neuroblastoma (NB) is a neoplasm of the sympathetic nervous system, and is the most common solid tumor of infancy. NBs are very heterogeneous, with a clinical course ranging from spontaneous regression to resistance to all current forms of treatment. High-risk patients need intense chemotherapy, and only 30–40% will be cured. Relapsed or metastatic tumors acquire multi-drug resistance, raising the need for alternative treatments. Owing to the diverse mechanisms that are responsible of NB chemoresistance, we aimed to target epigenetic factors that control multiple pathways to bypass therapy resistance. We found that the SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4/BRG1) was consistently upregulated in advanced stages of NB, with high BRG1 levels being indicative of poor outcome. Loss-of-function experiments in vitro and in vivo showed that BRG1 is essential for the proliferation of NB cells. Furthermore, whole-genome transcriptome analysis revealed that BRG1 controls the expression of key elements of oncogenic pathways such as PI3K/AKT and BCL2, which offers a promising new combination therapy for high-risk NB.


The Journal of Neuroscience | 2010

The Death Receptor Antagonist FLIP-L Interacts with Trk and Is Necessary for Neurite Outgrowth Induced by Neurotrophins

Rana S. Moubarak; Carme Solé; Marta Pascual; Humberto Gutierrez; M. Llovera; M. J. Perez-Garcia; Raffaella Gozzelino; Miguel F. Segura; V. Iglesias-Guimarais; Stéphanie Reix; R. M. Soler; Alun M. Davies; Eduardo Soriano; Victor J. Yuste; Joan X. Comella

FLICE-inhibitory protein (FLIP) is an endogenous inhibitor of the signaling pathway triggered by the activation of death receptors. Here, we reveal a novel biological function for the long form of FLIP (FLIP-L) in neuronal differentiation, which can be dissociated from its antiapoptotic role. We show that FLIP-L is expressed in different regions of the mouse embryonic nervous system. Immunohistochemistry of mouse brain sections at different stages reveals that, in neurons, FLIP is expressed early during the embryonic neuronal development (embryonic day 16) and decreases at later stages (postnatal days 5–15), when its expression is essentially detected in glial cells. FLIP-L overexpression significantly enhances neurotrophin-induced neurite outgrowth in motoneurons, superior cervical ganglion neurons, and PC12 cells. Conversely, the downregulation of FLIP-L protein levels by specific RNA interference significantly reduces neurite outgrowth, even in the presence of the appropriate neurotrophin stimulus. Moreover, NGF-dependent activation of two main intracellular pathways involved in the regulation of neurite outgrowth, extracellular signal-regulated kinases (ERKs) and nuclear factor κB (NF-κB), is impaired when endogenous FLIP-L is downregulated, although TrkA remains activated. Finally, we demonstrate that FLIP-L interacts with TrkA, and not with p75NTR, in an NGF-dependent manner, and endogenous FLIP-L interacts with TrkB in whole-brain lysates from embryonic day 15 mice embryos. Altogether, we uncover a new role for FLIP-L as an unexpected critical player in neurotrophin-induced mitogen-activated protein kinase/ERK- and NF-κB-mediated control of neurite growth in developing neurons.


The Journal of Neuroscience | 2013

FAIM-L Is an IAP-Binding Protein That Inhibits XIAP Ubiquitinylation and Protects from Fas-Induced Apoptosis

Rana S. Moubarak; Laura Planells-Ferrer; Jorge Urresti; Stéphanie Reix; Miguel F. Segura; Paulina Carriba; Fernando Marqués-Fernández; Carme Solé; Núria Llecha-Cano; Joaquín López-Soriano; Daniel Sanchis; Victor J. Yuste; Joan X. Comella

The neuronal long isoform of Fas Apoptotic Inhibitory Molecule (FAIM-L) protects from death receptor (DR)-induced apoptosis, yet its mechanism of protection remains unknown. Here, we show that FAIM-L protects rat neuronal Type II cells from Fas-induced apoptosis. XIAP has previously emerged as a molecular discriminator that is upregulated in Type II and downregulated in Type I apoptotic signaling. We demonstrate that FAIM-L requires sustained endogenous levels of XIAP to protect Type II cells as well as murine cortical neurons from Fas-induced apoptosis. FAIM-L interacts with the BIR2 domain of XIAP through an IAP-binding motif, the mutation of which impairs the antiapoptotic function of FAIM-L. Finally, we report that FAIM-L inhibits XIAP auto-ubiquitinylation and maintains its stability, thus conferring protection from apoptosis. Our results bring new understanding of the regulation of endogenous XIAP by a DR antagonist, pointing out at FAIM-L as a promising therapeutic tool for protection from apoptosis in pathological situations where XIAP levels are decreased.

Collaboration


Dive into the Miguel F. Segura's collaboration.

Top Co-Authors

Avatar

Soledad Gallego

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Aroa Soriano

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Josep Roma

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Joan X. Comella

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Carla Molist

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Luz Jubierre

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Rana S. Moubarak

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Ana Almazán-Moga

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Carme Solé

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Isaac Vidal

Autonomous University of Barcelona

View shared research outputs
Researchain Logo
Decentralizing Knowledge