Network


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

Hotspot


Dive into the research topics where Astrid Cannich is active.

Publication


Featured researches published by Astrid Cannich.


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 | 2010

Bimodal control of stimulated food intake by the endocannabinoid system

Luigi Bellocchio; Pauline Lafenetre; Astrid Cannich; Daniela Cota; Nagore Puente; Pedro Grandes; Francis Chaouloff; Pier Vincenzo Piazza; Giovanni Marsicano

Activation of cannabinoid type-1 receptors (CB1) is universally recognized as a powerful endogenous orexigenic signal, but the detailed underlying neuronal mechanisms are not fully understood. Using combined genetic and pharmacological approaches in mice, we found that ventral striatal CB1 receptors exerted a hypophagic action through inhibition of GABAergic transmission. Conversely, brain CB1 receptors modulating excitatory transmission mediated the well-known orexigenic effects of cannabinoids.


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.


Endocrinology | 2012

Hypothalamic CB1 Cannabinoid Receptors Regulate Energy Balance in Mice

Pierre Cardinal; Luigi Bellocchio; Samantha Clark; Astrid Cannich; Matthias Klugmann; Beat Lutz; Giovanni Marsicano; Daniela Cota

Cannabinoid type 1 (CB(1)) receptor activation is generally considered a powerful orexigenic signal and inhibition of the endocannabinoid system is beneficial for the treatment of obesity and related metabolic diseases. The hypothalamus plays a critical role in regulating energy balance by modulating both food intake and energy expenditure. Although CB(1) receptor signaling has been implicated in the modulation of both these mechanisms, a complete understanding of its role in the hypothalamus is still lacking. Here we combined a genetic approach with the use of adeno-associated viral vectors to delete the CB(1) receptor gene in the adult mouse hypothalamus and assessed the impact of such manipulation on the regulation of energy balance. Viral-mediated deletion of the CB(1) receptor gene in the hypothalamus led to the generation of Hyp-CB(1)-KO mice, which displayed an approximately 60% decrease in hypothalamic CB(1) receptor mRNA levels. Hyp-CB(1)-KO mice maintained on a normocaloric, standard diet showed decreased body weight gain over time, which was associated with increased energy expenditure and elevated β(3)-adrenergic receptor and uncoupling protein-1 mRNA levels in the brown adipose tissue but, surprisingly, not to changes in food intake. Additionally, Hyp-CB(1)-KO mice were insensitive to the anorectic action of the hormone leptin (5 mg/kg) and displayed a time-dependent hypophagic response to the CB(1) inverse agonist rimonabant (3 mg/kg). Altogether these findings suggest that hypothalamic CB(1) receptor signaling is a key determinant of energy expenditure under basal conditions and reveal its specific role in conveying the effects of leptin and pharmacological CB1 receptor antagonism on food intake.


Nature Neuroscience | 2009

Self-modulation of neocortical pyramidal neurons by endocannabinoids

Silvia Marinelli; Simone Pacioni; Astrid Cannich; Giovanni Marsicano; Alberto Bacci

Control of pyramidal neuron excitability is vital for the functioning of the neocortex. Somatodendritic slow self-inhibition (SSI) allows inhibitory neurons to regulate their own activity, but the existence of similar mechanisms in excitatory cells has not been shown. We found that in rodents endocannabinoids mediated SSI and long-term modulation of inhibitory connections in layer 2/3 pyramidal neurons with a distinct dendritic morphology, suggesting that a glutamatergic network in cortical circuits is self-regulated.


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.


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.


Nature Neuroscience | 2010

Synaptic activation of kainate receptors gates presynaptic CB 1 signaling at GABAergic synapses

Joana Lourenço; Astrid Cannich; Mario Carta; Françoise Coussen; Christophe Mulle; Giovanni Marsicano

Glutamate can control inhibitory synaptic transmission through activation of presynaptic kainate receptors. We found that glutamate released by train stimulation of Schaffer collaterals could lead to either short-term depression or short-term facilitation of inhibitory synaptic transmission in mouse CA1 pyramidal neurons, depending on the presence of cannabinoid type 1 (CB1) receptors on GABAergic afferents. The train-induced depression of inhibition (t-Di) required the mobilization of 2-arachidonoylglycerol through postsynaptic activation of metabotropic glutamate receptors and [Ca2+] rise. GluK1 (GluR5)-dependent depolarization of GABAergic terminals enabled t-Di by facilitating presynaptic CB1 signaling. Thus, concerted activation of presynaptic CB1 receptors and kainate receptors mediates short-term depression of inhibitory synaptic transmission. In contrast, in inhibitory connections expressing GluK1, but not CB1, receptors, train stimulation of Schaffer collaterals led to short-term facilitation. Thus, activation of kainate receptors by synaptically released glutamate gates presynaptic CB1 signaling, which in turn controls the direction of short-term heterosynaptic plasticity.

Collaboration


Dive into the Astrid Cannich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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
Top Co-Authors

Avatar

Pedro Grandes

University of the Basque Country

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge