Network


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

Hotspot


Dive into the research topics where Edgar Soria-Gómez is active.

Publication


Featured researches published by Edgar Soria-Gómez.


Nature Neuroscience | 2012

Mitochondrial CB1 receptors regulate neuronal energy metabolism

Giovanni Benard; Federico Massa; Nagore Puente; Joana Lourenço; Luigi Bellocchio; Edgar Soria-Gómez; Isabel Matias; Anna Delamarre; Mathilde Metna-Laurent; Astrid Cannich; Etienne Hebert-Chatelain; Christophe Mulle; Silvia Ortega-Gutiérrez; Mar Martín-Fontecha; Matthias Klugmann; Stephan Guggenhuber; Beat Lutz; Jürg Gertsch; Francis Chaouloff; María L. López-Rodríguez; Pedro Grandes; Rodrigue Rossignol; Giovanni Marsicano

The mammalian brain is one of the organs with the highest energy demands, and mitochondria are key determinants of its functions. Here we show that the type-1 cannabinoid receptor (CB1) is present at the membranes of mouse neuronal mitochondria (mtCB1), where it directly controls cellular respiration and energy production. Through activation of mtCB1 receptors, exogenous cannabinoids and in situ endocannabinoids decreased cyclic AMP concentration, protein kinase A activity, complex I enzymatic activity and respiration in neuronal mitochondria. In addition, intracellular CB1 receptors and mitochondrial mechanisms contributed to endocannabinoid-dependent depolarization-induced suppression of inhibition in the hippocampus. Thus, mtCB1 receptors directly modulate neuronal energy metabolism, revealing a new mechanism of action of G protein–coupled receptor signaling in the brain.


Nature Neuroscience | 2014

The endocannabinoid system controls food intake via olfactory processes

Edgar Soria-Gómez; Luigi Bellocchio; Leire Reguero; Gabriel Lepousez; Claire Martin; Mounir Bendahmane; Sabine Ruehle; Floor Remmers; Tiffany Desprez; Isabelle Matias; Theresa Wiesner; Astrid Cannich; Antoine Nissant; Aya Wadleigh; Hans-Christian Pape; Anna Chiarlone; Carmelo Quarta; Danièle Verrier; Peggy Vincent; Federico Massa; Beat Lutz; Manuel Guzmán; Hirac Gurden; Guillaume Ferreira; Pierre-Marie Lledo; Pedro Grandes; Giovanni Marsicano

Hunger arouses sensory perception, eventually leading to an increase in food intake, but the underlying mechanisms remain poorly understood. We found that cannabinoid type-1 (CB1) receptors promote food intake in fasted mice by increasing odor detection. CB1 receptors were abundantly expressed on axon terminals of centrifugal cortical glutamatergic neurons that project to inhibitory granule cells of the main olfactory bulb (MOB). Local pharmacological and genetic manipulations revealed that endocannabinoids and exogenous cannabinoids increased odor detection and food intake in fasted mice by decreasing excitatory drive from olfactory cortex areas to the MOB. Consistently, cannabinoid agonists dampened in vivo optogenetically stimulated excitatory transmission in the same circuit. Our data indicate that cortical feedback projections to the MOB crucially regulate food intake via CB1 receptor signaling, linking the feeling of hunger to stronger odor processing. Thus, CB1 receptor–dependent control of cortical feedback projections in olfactory circuits couples internal states to perception and behavior.


Proceedings of the National Academy of Sciences of the United States of America | 2014

A restricted population of CB1 cannabinoid receptors with neuroprotective activity

Anna Chiarlone; Luigi Bellocchio; Cristina Blázquez; Eva Resel; Edgar Soria-Gómez; Astrid Cannich; José Javier Ferrero; Onintza Sagredo; Cristina Benito; Julián Romero; José Sánchez-Prieto; Beat Lutz; Javier Fernández-Ruiz; Ismael Galve-Roperh; Manuel Guzmán

Significance Cannabinoids and their endogenous counterparts, the so-called endocannabinoids, promote neuroprotection in laboratory animals by engaging CB1 cannabinoid receptors, one of the most abundant types of receptors in the brain. However, the assessment of the physiological relevance and therapeutic potential of the CB1 receptor in neurological diseases is hampered, at least in part, by the lack of knowledge of the neuron-population specificity of CB1 receptor action. This study shows that a unique and well-defined population of CB1 receptors, namely that located on glutamatergic terminals, plays a key neuroprotective role in the mouse brain. This finding opens a new conceptual view on how the CB1 receptor evokes neuroprotection, and provides preclinical support for improving the development of cannabinoid-based neuroprotective therapies. The CB1 cannabinoid receptor, the main molecular target of endocannabinoids and cannabis active components, is the most abundant G protein-coupled receptor in the mammalian brain. Of note, CB1 receptors are expressed at the synapses of two opposing (i.e., GABAergic/inhibitory and glutamatergic/excitatory) neuronal populations, so the activation of one and/or another receptor population may conceivably evoke different effects. Despite the widely reported neuroprotective activity of the CB1 receptor in animal models, the precise pathophysiological relevance of those two CB1 receptor pools in neurodegenerative processes is unknown. Here, we first induced excitotoxic damage in the mouse brain by (i) administering quinolinic acid to conditional mutant animals lacking CB1 receptors selectively in GABAergic or glutamatergic neurons, and (ii) manipulating corticostriatal glutamatergic projections remotely with a designer receptor exclusively activated by designer drug pharmacogenetic approach. We next examined the alterations that occur in the R6/2 mouse, a well-established model of Huntington disease, upon (i) fully knocking out CB1 receptors, and (ii) deleting CB1 receptors selectively in corticostriatal glutamatergic or striatal GABAergic neurons. The data unequivocally identify the restricted population of CB1 receptors located on glutamatergic terminals as an indispensable player in the neuroprotective activity of (endo)cannabinoids, therefore suggesting that this precise receptor pool constitutes a promising target for neuroprotective therapeutic strategies.


Nature | 2016

A cannabinoid link between mitochondria and memory.

Etienne Hebert-Chatelain; Tifany Desprez; Román Serrat; Luigi Bellocchio; Edgar Soria-Gómez; Arnau Busquets-Garcia; Antonio C. Pagano Zottola; Anna Delamarre; Astrid Cannich; Peggy Vincent; Marjorie Varilh; Laurie M. Robin; Geoffrey Terral; M. Dolores García-Fernández; Michelangelo Colavita; Wilfrid Mazier; Filippo Drago; Nagore Puente; Leire Reguero; Izaskun Elezgarai; Jean-William Dupuy; Daniela Cota; Maria-Luz Lopez-Rodriguez; Gabriel Barreda-Gómez; Federico Massa; Pedro Grandes; Giovanni Benard; Giovanni Marsicano

Cellular activity in the brain depends on the high energetic support provided by mitochondria, the cell organelles which use energy sources to generate ATP. Acute cannabinoid intoxication induces amnesia in humans and animals, and the activation of type-1 cannabinoid receptors present at brain mitochondria membranes (mtCB1) can directly alter mitochondrial energetic activity. Although the pathological impact of chronic mitochondrial dysfunctions in the brain is well established, the involvement of acute modulation of mitochondrial activity in high brain functions, including learning and memory, is unknown. Here, we show that acute cannabinoid-induced memory impairment in mice requires activation of hippocampal mtCB1 receptors. Genetic exclusion of CB1 receptors from hippocampal mitochondria prevents cannabinoid-induced reduction of mitochondrial mobility, synaptic transmission and memory formation. mtCB1 receptors signal through intra-mitochondrial Gαi protein activation and consequent inhibition of soluble-adenylyl cyclase (sAC). The resulting inhibition of protein kinase A (PKA)-dependent phosphorylation of specific subunits of the mitochondrial electron transport system eventually leads to decreased cellular respiration. Hippocampal inhibition of sAC activity or manipulation of intra-mitochondrial PKA signalling or phosphorylation of the Complex I subunit NDUFS2 inhibit bioenergetic and amnesic effects of cannabinoids. Thus, the G protein-coupled mtCB1 receptors regulate memory processes via modulation of mitochondrial energy metabolism. By directly linking mitochondrial activity to memory formation, these data reveal that bioenergetic processes are primary acute regulators of cognitive functions.


The Journal of Neuroscience | 2012

Bimodal Control of Fear-Coping Strategies by CB1 Cannabinoid Receptors

Mathilde Metna-Laurent; Edgar Soria-Gómez; Danièle Verrier; Martina Conforzi; Pierrick Jégo; Pauline Lafenetre; Giovanni Marsicano

To maximize their chances of survival, animals need to rapidly and efficiently respond to aversive situations. These responses can be classified as active or passive and depend on the specific nature of threats, but also on individual fear coping styles. In this study, we show that the control of excitatory and inhibitory brain neurons by type-1 cannabinoid (CB1) receptors is a key determinant of fear coping strategies in mice. In classical fear conditioning, a switch between initially predominant passive fear responses (freezing) and active behaviors (escape attempts and risk assessment) develops over time. Constitutive genetic deletion of CB1 receptors in CB1−/− mice disrupted this pattern by favoring passive responses. This phenotype can be ascribed to endocannabinoid control of excitatory neurons, because it was reproduced in conditional mutant mice lacking CB1 receptors from cortical glutamatergic neurons. CB1 receptor deletion from GABAergic brain neurons led to the opposite phenotype, characterized by the predominance of active coping. The CB1 receptor agonist Δ9-tetrahydrocannabinol exerted a biphasic control of fear coping strategies, with lower and higher doses favoring active and passive responses, respectively. Finally, viral re-expression of CB1 receptors in the amygdala of CB1−/− mice restored the normal switch between the two coping strategies. These data strongly suggest that CB1 receptor signaling bimodally controls the spontaneous adoption of active or passive coping strategies in individuals. This primary function of the endocannabinoid system in shaping individual behavioral traits should be considered when studying the mechanisms of physiological and pathological fear.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Activation of the sympathetic nervous system mediates hypophagic and anxiety-like effects of CB₁ receptor blockade.

Luigi Bellocchio; Edgar Soria-Gómez; Carmelo Quarta; Mathilde Metna-Laurent; Pierre Cardinal; Elke Binder; Astrid Cannich; Anna Delamarre; Martin Häring; Mar Martín-Fontecha; David Vega; Thierry Leste-Lasserre; Dusan Bartsch; Krisztina Monory; Beat Lutz; Francis Chaouloff; Uberto Pagotto; Manuel Guzmán; Daniela Cota; Giovanni Marsicano

Complex interactions between periphery and the brain regulate food intake in mammals. Cannabinoid type-1 (CB1) receptor antagonists are potent hypophagic agents, but the sites where this acute action is exerted and the underlying mechanisms are not fully elucidated. To dissect the mechanisms underlying the hypophagic effect of CB1 receptor blockade, we combined the acute injection of the CB1 receptor antagonist rimonabant with the use of conditional CB1-knockout mice, as well as with pharmacological modulation of different central and peripheral circuits. Fasting/refeeding experiments revealed that CB1 receptor signaling in many specific brain neurons is dispensable for the acute hypophagic effects of rimonabant. CB1 receptor antagonist-induced hypophagia was fully abolished by peripheral blockade of β-adrenergic transmission, suggesting that this effect is mediated by increased activity of the sympathetic nervous system. Consistently, we found that rimonabant increases gastrointestinal metabolism via increased peripheral β-adrenergic receptor signaling in peripheral organs, including the gastrointestinal tract. Blockade of both visceral afferents and glutamatergic transmission in the nucleus tractus solitarii abolished rimonabant-induced hypophagia. Importantly, these mechanisms were specifically triggered by lipid-deprivation, revealing a nutrient-specific component acutely regulated by CB1 receptor blockade. Finally, peripheral blockade of sympathetic neurotransmission also blunted central effects of CB1 receptor blockade, such as fear responses and anxiety-like behaviors. These data demonstrate that, independently of their site of origin, important effects of CB1 receptor blockade are expressed via activation of peripheral sympathetic activity. Thus, CB1 receptors modulate bidirectional circuits between the periphery and the brain to regulate feeding and other behaviors.


Biological Psychiatry | 2013

Ventral Tegmental Area Cannabinoid Type-1 Receptors Control Voluntary Exercise Performance

Sarah Dubreucq; Audrey Durand; Isabelle Matias; Giovanni Benard; Elodie Richard; Edgar Soria-Gómez; Christelle Glangetas; Laurent Groc; Aya Wadleigh; Federico Massa; Dusan Bartsch; Giovanni Marsicano; François Georges; Francis Chaouloff

BACKGROUND We have shown that the endogenous stimulation of cannabinoid type-1 (CB₁) receptors is a prerequisite for voluntary running in mice, but the precise mechanisms through which the endocannabinoid system exerts a tonic control on running performance remain unknown. METHODS We analyzed the respective impacts of constitutive/conditional CB₁ receptor mutations and of CB₁ receptor blockade on wheel-running performance. We then assessed the consequences of ventral tegmental area (VTA) CB₁ receptor blockade on the wheel-running performances of wildtype (gamma-aminobutyric acid [GABA]-CB₁⁺/⁺) and mutant (GABA-CB₁⁻/⁻) mice for CB₁ receptors in brain GABA neurons. Using in vivo electrophysiology, the consequences of wheel running on VTA dopamine (DA) neuronal activity were examined in GABA-CB₁⁺/⁺ and GABA-CB₁⁻/⁻ mice. RESULTS Conditional deletion of CB₁ receptors from brain GABA neurons, but not from several other neuronal populations or from astrocytes, decreased wheel-running performance in mice. The inhibitory consequences of either the systemic or the intra-VTA administration of CB1 receptor antagonists on running behavior were abolished in GABA-CB₁⁻/⁻ mice. The absence of CB1 receptors from GABAergic neurons led to a depression of VTA DA neuronal activity after acute/repeated wheel running. CONCLUSIONS This study provides evidence that CB₁ receptors on VTA GABAergic terminals exert a permissive control on rodent voluntary running performance. Furthermore, it is shown that CB₁ receptors located on GABAergic neurons impede negative consequences of voluntary exercise on VTA DA neuronal activity. These results position the endocannabinoid control of inhibitory transmission as a prerequisite for wheel-running performance in mice.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Striatal GABAergic and cortical glutamatergic neurons mediate contrasting effects of cannabinoids on cortical network synchrony

Carola Sales-Carbonell; Pavel E Rueda-Orozco; Edgar Soria-Gómez; György Buzsáki; Giovanni Marsicano; David Robbe

Activation of type 1 cannabinoid receptors (CB1R) decreases GABA and glutamate release in cortical and subcortical regions, with complex outcomes on cortical network activity. To date there have been few attempts to disentangle the region- and cell-specific mechanisms underlying the effects of cannabinoids on cortical network activity in vivo. Here we addressed this issue by combining in vivo electrophysiological recordings with local and systemic pharmacological manipulations in conditional mutant mice lacking CB1R expression in different neuronal populations. First we report that cannabinoids induce hypersynchronous thalamocortical oscillations while decreasing the amplitude of faster cortical oscillations. Then we demonstrate that CB1R at striatonigral synapses (basal ganglia direct pathway) mediate the thalamocortical hypersynchrony, whereas activation of CB1R expressed in cortical glutamatergic neurons decreases cortical synchrony. Finally we show that activation of CB1 expressed in cortical glutamatergic neurons limits the cannabinoid-induced thalamocortical hypersynchrony. By reporting that CB1R activations in cortical and subcortical regions have contrasting effects on cortical synchrony, our study bridges the gap between cellular and in vivo network effects of cannabinoids. Incidentally, the thalamocortical hypersynchrony we report suggests a potential mechanism to explain the sensory “high” experienced during recreational consumption of marijuana.


Molecular metabolism | 2013

Astroglial CB1 cannabinoid receptors regulate leptin signaling in mouse brain astrocytes

Barbara Bosier; Luigi Bellocchio; Mathilde Metna-Laurent; Edgar Soria-Gómez; Isabelle Matias; Etienne Hebert-Chatelain; Astrid Cannich; Marlène Maitre; Thierry Leste-Lasserre; Pierre Cardinal; Juan Mendizabal-Zubiaga; Miren Josune Canduela; Leire Reguero; Emmanuel Hermans; Pedro Grandes; Daniela Cota; Giovanni Marsicano

Type-1 cannabinoid (CB1) and leptin (ObR) receptors regulate metabolic and astroglial functions, but the potential links between the two systems in astrocytes were not investigated so far. Genetic and pharmacological manipulations of CB1 receptor expression and activity in cultured cortical and hypothalamic astrocytes demonstrated that cannabinoid signaling controls the levels of ObR expression. Lack of CB1 receptors also markedly impaired leptin-mediated activation of signal transducers and activators of transcription 3 and 5 (STAT3 and STAT5) in astrocytes. In particular, CB1 deletion determined a basal overactivation of STAT5, thereby leading to the downregulation of ObR expression, and leptin failed to regulate STAT5-dependent glycogen storage in the absence of CB1 receptors. These results show that CB1 receptors directly interfere with leptin signaling and its ability to regulate glycogen storage, thereby representing a novel mechanism linking endocannabinoid and leptin signaling in the regulation of brain energy storage and neuronal functions.


Neuroscience | 2012

Cannabinoid type 1 receptors located on single-minded 1–expressing neurons control emotional behaviors

S. Dubreucq; S. Kambire; M. Conforzi; Mathilde Metna-Laurent; Astrid Cannich; Edgar Soria-Gómez; E. Richard; Giovanni Marsicano; Francis Chaouloff

This study has investigated the role of hypothalamic and amygdalar type-1 cannabinoid (CB1) receptors in the emotional and neuroendocrine responses to stress. To do so, we used the Cre/loxP system to generate conditional mutant mice lacking the CB1 gene in neurons expressing the transcription factor single-minded 1 (Sim1). This choice was dictated by former evidence for Sim1-Cre transgenic mice bearing Cre activity in all areas expressing Sim1, which chiefly includes the hypothalamus (especially the paraventricular nucleus, the supraoptic nucleus, and the posterior hypothalamus) and the mediobasal amygdala. Genomic DNA analyses in Sim1-CB1(-/-) mice indicated that the CB1 allele was excised from the hypothalamus and the amygdala, but not from the cortex, the striatum, the thalamus, the nucleus accumbens, the brainstem, the hippocampus, the pituitary gland, and the spinal cord. Double-fluorescent in situ hybridization experiments further indicated that Sim1-CB1(-/-) mice displayed a weaker CB1 receptor mRNA expression in the paraventricular nucleus of the hypothalamus and the mediobasal part of the amygdala, compared to wild-type animals. Individually housed Sim1-CB1(-/-) mice and their Sim1-CB1(+/+) littermates were exposed to anxiety and fear memory tests under basal conditions as well as after acute/repeated social stress. A principal component analysis of the behaviors of Sim1-CB1(-/-) and Sim1-CB1(+/+) mice in anxiety tests (open field, elevated plus-maze, and light/dark box) revealed that CB1 receptors from Sim1-expressing neurons exert tonic, albeit opposite, controls of locomotor and anxiety reactivity to novel environments. No difference between genotypes was observed during the recall of contextual fear conditioning or during active avoidance learning. Sim1-CB1(-/-), but not Sim1-CB1(+/+), mice proved sensitive to an acute social stress as this procedure reverted the increased ambulation in the center of the open field. The stimulatory influence of repeated social stress on body and adrenal weights, water intake, and sucrose preference was similar in the two genotypes. On the other hand, repeated social stress abolished the decrease in cued-fear conditioned expression that was observed in Sim1-CB1(-/-) mice, compared to Sim1-CB1(+/+) mice. This study suggests that CB1 receptors located on Sim1-expressing neurons exert a tonic control on locomotor reactivity, unconditioned anxiety, and cued-fear expression under basal conditions as well as after acute or repeated stress.

Collaboration


Dive into the Edgar Soria-Gómez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arnau Busquets-Garcia

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Daniela Cota

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge