Network


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

Hotspot


Dive into the research topics where Nicolas Aubert is active.

Publication


Featured researches published by Nicolas Aubert.


Journal of Neurochemistry | 2004

Opposite regulation of the mitochondrial apoptotic pathway by C2-ceramide and PACAP through a MAP-kinase-dependent mechanism in cerebellar granule cells

Anthony Falluel-Morel; Nicolas Aubert; David Vaudry; Magali Basille; Marc Fontaine; Alain Fournier; Hubert Vaudry; Bruno J. Gonzalez

The sphingomyelin‐derived messenger ceramides provoke neuronal apoptosis through caspase‐3 activation, while the neuropeptide pituitary adenylate cyclase‐activating polypeptide (PACAP) promotes neuronal survival and inhibits caspase‐3 activity. However, the mechanisms leading to the opposite regulation of caspase‐3 by C2‐ceramide and PACAP are currently unknown. Here, we show that PACAP prevents C2‐ceramide‐induced inhibition of mitochondrial potential and C2‐ceramide‐evoked cytochrome c release. C2‐ceramide stimulated Bax expression, but had no effect on Bcl‐2, while PACAP abrogated the action of C2‐ceramide on Bax and stimulated Bcl‐2 expression. The effects of C2‐ceramide and PACAP on Bax and Bcl‐2 were blocked, respectively, by the JNK inhibitor L‐JNKI1 and the MEK inhibitor U0126. L‐JNKI1 prevented the alteration of mitochondria induced by C2‐ceramide while U0126 suppressed the protective effect of PACAP against the deleterious action of C2‐ceramide on mitochondrial potential. Moreover, L‐JNKI1 inhibited the stimulatory effect of C2‐ceramide on caspase‐9 and ‐3 and prevented C2‐ceramide‐induced cell death. U0126 blocked PACAP‐induced Bcl‐2 expression, abrogated the inhibitory effect of PACAP on ceramide‐induced caspase‐9 activity, and promoted granule cell death. The present study reveals that C2‐ceramide and PACAP exert opposite effects on Bax and Bcl‐2 through, respectively, JNK‐ and ERK‐dependent mechanisms. These data indicate that the mitochondrial pathway plays a pivotal role in the pro‐ and anti‐apoptotic effects of C2‐ceramide and PACAP.


Journal of Neurochemistry | 2006

PACAP and C2‐ceramide generate different AP‐1 complexes through a MAP‐kinase‐dependent pathway: involvement of c‐Fos in PACAP‐induced Bcl‐2 expression

Nicolas Aubert; Anthony Falluel-Morel; David Vaudry; Xavier Xifró; José Rodríguez-Álvarez; Cecile Fisch; Stéphane de Jouffrey; Jean‐François Lebigot; Alain Fournier; Hubert Vaudry; Bruno J. Gonzalez

The neuropeptide pituitary adenylate cyclase‐activating polypeptide (PACAP) inhibits C2‐ceramide‐induced cell death through blockade of the mitochondrial apoptotic pathway in rat cerebellar granule neurones. However, the gene induction processes and transcription factors involved in the anti‐apoptotic effect of PACAP remain unknown. Here, we show that PACAP and C2‐ceramide activate activator protein‐1 (AP‐1) DNA binding in a dose‐ and time‐dependent manner, but generate different AP‐1 dimers. Thus, PACAP increased the proportion of c‐Fos and Jun D while C2‐ceramide increased c‐Jun and reduced c‐Fos in AP‐1 complexes. In addition, PACAP strongly activated c‐Fos gene expression while C2‐ceramide markedly increased c‐Jun phosphorylation. The effect of PACAP on c‐Fos expression was blocked by the mitogen‐activated protein kinase/extracellular signal‐regulated kinase (MEK) inhibitor, U0126, while phosphorylation of c‐Jun induced by C2‐ceramide was abrogated by the protein phosphatase 2A (PP2A) inhibitor, okadaic acid. Transfection of immature granule cells with c‐Fos siRNA, which strongly reduced basal and PACAP‐stimulated levels of the protein, totally prevented the stimulatory effect of PACAP on Bcl‐2 expression. The present study demonstrates that AP‐1 complexes containing c‐Fos mediate the effect of PACAP on Bcl‐2 gene expression in cerebellar granule neurones. Our data also indicate that different AP‐1 dimers are associated with the pro‐apoptotic effect of C2‐ceramide and the anti‐apoptotic effect of PACAP.


Journal of Neuroendocrinology | 2007

The Neuropeptide Pituitary Adenylate Cyclase-Activating Polypeptide Exerts Anti-Apoptotic and Differentiating Effects during Neurogenesis: Focus on Cerebellar Granule Neurones and Embryonic Stem Cells

Anthony Falluel-Morel; M. Chafai; David Vaudry; M. Basille; Michèle Cazillis; Nicolas Aubert; Estelle Louiset; S. DeJouffrey; J.-F. Le Bigot; Alain Fournier; Pierre Gressens; William Rostène; Hubert Vaudry; Bruno J. Gonzalez

Pituitary adenylate cyclase‐activating polypeptide (PACAP) was originally isolated from ovine hypothalamus on the basis of its hypophysiotrophic activity. It has subsequently been shown that PACAP and its receptors are widely distributed in the central nervous system of adult mammals, indicating that PACAP may act as a neurotransmitter and/or neuromodulator. It has also been found that PACAP and its receptors are expressed in germinative neuroepithelia, suggesting that PACAP could be involved in neurogenesis. There is now compelling evidence that PACAP exerts neurotrophic activities in the developing cerebellum and in embryonic stem (ES) cells. In particular, the presence of PACAP receptors has been demonstrated in the granule layer of the immature cerebellar cortex, and PACAP has been shown to promote survival, inhibit migration and activate neurite outgrowth of granule cell precursors. In cerebellar neuroblasts, PACAP is a potent inhibitor of the mitochondrial apoptotic pathway through activation of the MAPkinase extracellular regulated kinase. ES cells and embryoid bodies (EB) also express PACAP receptors and PACAP facilitates neuronal orientation and induces the appearance of an electrophysiological activity. Taken together, the anti‐apoptotic and pro‐differentiating effects of PACAP characterised in cerebellar neuroblasts as well as ES and EB cells indicate that PACAP acts not only as a neurohormone and a neurotransmitter, but also as a growth factor.


Journal of Biological Chemistry | 2006

N-methyl-D-aspartate blocks activation of JNK and mitochondrial apoptotic pathway induced by potassium deprivation in cerebellar granule cells.

Xavier Xifró; Anthony Falluel-Morel; Alfredo Miñano; Nicolas Aubert; Rut Fadó; Cristina Malagelada; David Vaudry; Hubert Vaudry; Bruno J. Gonzalez; José Rodríguez-Álvarez

During the postnatal development of cerebellum, lack of excitatory innervation from the mossy fibers results in cerebellar granule cell (CGC) apoptosis during the migration of the cells toward the internal granule cell layer. Accordingly, CGCs die by apoptosis when cultured in physiological KCl concentrations (5 mm; K5), and they survive in the presence of depolarizing conditions such as high KCl concentration (25 mm; K25) or N-methyl-d-aspartate (NMDA). We have recently shown that NMDA is able to exert a long lasting neuroprotective effect when added to immature (2 days in vitro) CGC cultures by inhibition of caspase-3 activity. Here we show that NMDA- and K25-mediated neuroprotection is associated with an increase in the levels of Bcl-2, an inhibition of K5-mediated increase in Bax, and the inhibition of the release of apoptogenic factors from mitochondria such as Smac/DIABLO and cytochrome c. Moreover, we have shown that similar effects are observed when c-Jun N-terminal kinases (JNKs) are inhibited and that treatment of CGC cultures with NMDA blocks K5-mediated JNK activation. These results allow us to postulate that the inhibition of JNK-mediated release of apoptogenic factors from mitochondria is involved in the NMDA protection from K5-mediated apoptosis of CGCs.


Neuroscience | 2009

Distribution and functional characterization of pituitary adenylate cyclase–activating polypeptide receptors in the brain of non-human primates

Valérie Jolivel; M. Basille; Nicolas Aubert; S. de Jouffrey; Philippe Ancian; J.-F. Le Bigot; Pauline Noack; Marc Massonneau; Alain Fournier; Hubert Vaudry; Bruno J. Gonzalez; David Vaudry

The distribution and density of pituitary adenylate cyclase-activating polypeptide (PACAP) binding sites have been investigated in the brain of the primates Jacchus callithrix (marmoset) and Macaca fascicularis (macaque) using [(125)I]-PACAP27 as a radioligand. PACAP binding sites were widely expressed in the brain of these two species with particularly high densities in the septum, hypothalamus and habenula. A moderate density of recognition sites was seen in all subdivisions of the cerebral cortex with a heterogenous distribution, the highest concentrations occurring in layers I and VI while the underlying white matter was almost devoid of binding sites. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis revealed intense expression of the mRNAs encoding the short and hop-1 variants of pituitary adenylate cyclase-activating polypeptide-specific receptor (PAC1-R) in the cortex of both marmoset and macaque, whereas vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating polypeptide mutual receptor, subtype 1 (VPAC1-R) and vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating polypeptide mutual receptor, subtype 2 (VPAC2-R) mRNAs were expressed at a much lower level. In situ hybridization histochemistry showed intense expression of PAC1-R and weak expression of VPAC1-R mRNAs in layer IV of the cerebral cortex. Incubation of cortical tissue slices with PACAP induced a dose-dependent stimulation of cyclic AMP formation, indicating that PACAP binding sites correspond to functional receptors. Moreover, treatment of primate cortical slices with 100 nM PACAP significantly reduced the activity of caspase-3, a key enzyme of the apoptotic cascade. The present results indicate that PACAP should exert the same neuroprotective effect in the brain of primates as in rodents and suggest that PAC1-R agonists may have a therapeutic value to prevent neuronal cell death after stroke or in specific neurodegenerative diseases.


Neurobiology of Disease | 2008

PACAP prevents toxicity induced by cisplatin in rat and primate neurons but not in proliferating ovary cells: Involvement of the mitochondrial apoptotic pathway

Nicolas Aubert; David Vaudry; Anthony Falluel-Morel; Arnaud Desfeux; Cecile Fisch; Philippe Ancian; Stéphane de Jouffrey; Jean-François Le Bigot; Alain Couvineau; Marc Laburthe; Alain Fournier; Vincent Laudenbach; Hubert Vaudry; Bruno J. Gonzalez

Cisplatin is a chemotherapeutic agent whose use is limited by side effects including neuropathies. In proliferating cells, toxic action of cisplatin is based on DNA interactions, while, in quiescent cells, it can induce apoptosis by interacting with proteins. In the present study, we compared cytotoxic mechanisms activated by cisplatin in primate and rodent neurons and in ovary cells in order to determine whether the anti-apoptotic peptide PACAP could selectively reduce neurotoxicity. In quiescent neurons, JNK and sphingomyelinase inhibitors blocked cisplatin-induced cell death. Toxicity was associated with DNA laddering, caspase-3 and -9 activations and Bax induction. These effects were prevented by PACAP. In proliferating cells, cisplatin activated caspase-8 but had no effect on caspase-9. PACAP exerted no protective effect. These data indicate that cisplatin activates distinct apoptotic pathways in quiescent neurons and proliferating cells and that PACAP may reduce neurotoxicity of cisplatin without affecting its chemotherapeutic efficacy.


Journal of Molecular Neuroscience | 2008

Interactions of PACAP and Ceramides in the Control of Granule Cell Apoptosis During Cerebellar Development

Anthony Falluel-Morel; Nicolas Aubert; David Vaudry; Arnaud Desfeux; Aurélie Allais; Delphine Burel; M. Basille; Hubert Vaudry; Vincent Laudenbach; Bruno J. Gonzalez

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that belongs to the secretin/glucagon/vasoactive intestinal polypeptide superfamily. The PACAPergic system is actively expressed in the developing cerebellum of mammals. In particular, PACAP receptors are expressed by granule cell precursors suggesting a role of the peptide in neurogenesis of this cell type. Consistent with this hypothesis, several studies reported antiapoptotic effects of PACAP in the developing cerebellum. On the other hand, the sphingomyelin metabolites ceramides are recognized as important signaling molecules that play pivotal roles during neuronal development. Ceramides, which production can be induced by death factors such as FasL or TNFalpha, are involved in the control of cell survival during brain development through activation of caspase-dependent mechanisms. The present review focuses on the interactions between PACAP and ceramides in the control of granule cell survival and on the transduction mechanisms associated with the anti- and proapoptotic effects of PACAP and ceramides, respectively.


Cell Death & Differentiation | 2008

Bax siRNA promotes survival of cultured and allografted granule cell precursors through blockade of caspase-3 cleavage.

S. S. Zhokhov; Arnaud Desfeux; Nicolas Aubert; Anthony Falluel-Morel; Alain Fournier; Vincent Laudenbach; H. Vaudry; Bruno J. Gonzalez

Transplantation of neuronal precursor cells (NPCs) into the central nervous system could represent a powerful therapeutical tool against neurodegenerative diseases. Unfortunately, numerous NPCs die shortly after transplantation, predominantly due to caspase-dependent apoptosis. Using a culture of cerebellar neuronal precursors, we have previously demonstrated protective effect of the neuropeptide PACAP, which suppresses ceramide-induced apoptosis by blockade of the mitochondrial apoptotic pathway. The main objective of this study was to determine whether Bax repression can promote survival of NPCs allotransplanted into a host animal. In vivo and ex vivo experiments revealed that C2-ceramide increases Bax expression, while PACAP reverses this effect. In vitro tests using cerebellar NPCs demonstrated that the Bax-specific small interfering RNA (siRNA) could reduce their death and caspase-3 cleavage within the first 24 h. BrdU-labelled NPCs were subjected to transfection procedure with or without siRNA introduction before using for in vivo transplantation. Twenty-four hours after, the allografted NPCs containing siRNA showed significantly reduced level of caspase-3 cleavage, and the volume of their implants was almost twofold higher than in the case of empty-transfected precursors. These data evidence an important role of Bax in life/death decision of grafted NPCs and suggest that RNA interference strategy may be applicable for maintaining NPCs survival within the critical first hours after their transplantation.


Regulatory Peptides | 2006

Ontogeny of PACAP receptors in the human cerebellum: Perspectives of therapeutic applications

Magali Basille; Anthony Falluel-Morel; David Vaudry; Nicolas Aubert; Alain Fournier; Pierre Fréger; Nicole Gallo-Payet; Hubert Vaudry; Bruno J. Gonzalez

It is now well established that pituitary adenylate cyclase-activating polypeptide (PACAP) exerts anti-apoptotic and pro-differentiating actions during development of the rodent cerebellum. Cell signaling involved in the neurotrophic effects of PACAP has been precisely investigated. In particular, PACAP is a potent inhibitor of the mitochondrial apoptotic pathway through an ERK- and PKA-dependent mechanism. However, transposition of the neurodevelopmental activities of PACAP to the human cerebellum remains speculative, essentially because of the lack of data concerning the PACAP-ergic system. The present review is based on recent results that provide the first molecular, pharmacological and anatomical characterizations of PACAP receptors in the developing human cerebellum. It is now clearly established that the distribution pattern of PAC1-R and VPAC1-R mRNA in the human cerebellum is very similar to that already described in rodents. [(125)I]PACAP27 binding sites are closely associated with germinative neuroepithelia in fetal stages and with mature granule cells in infants and adults. Pharmacological characterization revealed that, in fetuses, PACAP binding sites exhibit a PAC1-R profile while, in adult patients, they correspond to a heterogeneous population of PAC1-R and VPAC(1/2)-R. Altogether, these data provide the first evidence that PACAP may exert neurodevelopmental functions in the human cerebellum.


The Journal of Comparative Neurology | 2007

Molecular, cellular, and functional characterizations of pituitary adenylate cyclase-activating polypeptide and its receptors in the cerebellum of New and Old World monkeys

Nicolas Aubert; Magali Basille; Anthony Falluel-Morel; David Vaudry; Christine Bucharles; Valérie Jolivel; Cecile Fisch; Stéphane de Jouffrey; Jean-François Le Bigot; Alain Fournier; Hubert Vaudry; Bruno J. Gonzalez

The neuropeptide pituitary adenylate cyclase‐activating polypeptide (PACAP) exerts trophic activities during cerebellar development, and a neuroprotective effect of PACAP has been demonstrated in pathological conditions such as stroke. However, all these data have been obtained in rodents, and neuroprotective effects of PACAP in primates remain unknown. Because of their evolutionary relationships with humans, monkeys represent powerful models for validating the therapeutic interest in PACAP. The objective of the present study was to characterize PACAP and its receptors in the cerebellum of two nonhuman primates. RT‐PCR and in situ hybridization experiments revealed that PACAP is expressed in the cerebellum by Purkinje cells. Via immunohistochemistry, PACAP was detected in Purkinje cells and radial glial fibers. With regard to PACAP receptors, PAC1‐R and VPAC1‐R were detected by RT‐PCR. In situ hybridization revealed a strong expression of PAC1‐R and VPAC1‐R in the granule cell layer (GCL), and VPAC1‐R was also expressed in the Purkinje cell layer. A high density of PACAP binding sites was visualized in the GCL and the Purkinje cell layer. Competition studies indicated that, in the GCL, PACAP induced complete displacement of [125I]PACAP27 binding, whereas vasoactive intestinal polypeptide (VIP) was a weak competitor. In contrast, in the Purkinje cell layer, both PACAP and VIP displaced [125I]PACAP27 binding. Measurement of cAMP levels showed that PACAP is a powerful activator of adenylyl cyclase, whereas VIP is about 100‐fold less potent. Altogether, these observations constitute the first demonstration of a functional PACAPergic system in monkey cerebellum. They strongly suggest that neuroprotective effects of PACAP can be transposed to primates, including human. J. Comp. Neurol. 504:427–439, 2007.

Collaboration


Dive into the Nicolas Aubert's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alain Fournier

Institut national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ludovic Galas

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge