Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1386230, 2024.
Article in English | MEDLINE | ID: mdl-38962676

ABSTRACT

Background: Despite the evidence that energy balance is regulated differently in females and that the endocannabinoid system is sexually dimorphic, previous studies on the endocannabinoid system and energy balance predominantly used male models. Here, we characterize the effects of cannabinoid receptor deletion on body weight gain and glucose metabolism in female C57BL mice. Methods: Female mice lacking the cannabinoid-1 receptor (CB1R-/-), cannabinoid-2 receptor (CB2R-/-), or both receptors (CB1R-/-/CB2R-/-) and wild-type (WT) mice were fed with a low (LFD; 10% of calories from fat) or high-fat diet (HFD; 45% of calories from fat) for six weeks. Results: Female WT mice fed with HFD gained significantly more weight than WT mice fed with LFD (p < 0.001). Similar pattern was observed for CB2/- mice fed with HFD compared to CB2R-/- mice fed with LFD (p < 0.001), but not for CB1R-/- fed with HFD vs. LFD (p = 0.22) or CB1R-/-/CB2R-/- fed with HFD vs. LFD (p = 0.96). Comparing the 4 groups on LFD, weight gain of CB1R-/- mice was greater than all other genotypes (p < 0.05). When fed with HFD, the deletion of CB1R alone in females did not attenuate weight gain compared to WT mice (p = 0.72). Female CB1R-/-/CB2R-/- mice gained less weight than WT mice when fed with HFD (p = 0.007) despite similar food intake and locomotor activity, potentially owing to enhanced thermogenesis in the white adipose tissue. No significant difference in weight gain was observed for female CB2R-/- and WT mice on LFD or HFD. Fasting glucose, however, was higher in CB2R-/- mice fed with LFD than all other groups (p < 0.05). Conclusion: The effects of cannabinoid receptor deletion on glucose metabolism in female mice were similar to previously published findings on male mice, yet the effects on body weight gain and thermogenesis were attenuated in CB1R-/- mice.


Subject(s)
Diet, High-Fat , Energy Metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptor, Cannabinoid, CB1 , Receptor, Cannabinoid, CB2 , Weight Gain , Animals , Female , Mice , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/deficiency , Diet, High-Fat/adverse effects , Weight Gain/genetics , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/deficiency , Body Weight
2.
Microvasc Res ; 139: 104263, 2022 01.
Article in English | MEDLINE | ID: mdl-34655603

ABSTRACT

Cannabinoids are reported to regulate cardiovascular functions. Cannabinoid receptors 1 (CB1Rs) are widely expressed in both the neuronal system and vascular system, but the contribution of CB1Rs in vascular smooth muscle (CB1RSM) to cardiovascular functions is not clear yet. In this research, we analyzed the effects of CB1RSM on blood pressure, vasoconstriction, and vasodilation abilities by using conditionally CB1R knockout mice (CB1RSMKO). The results show no significant difference in basal blood pressure between the conscious CB1RSMKO and control mice, indicating that CB1RSM is not essential for basal blood pressure maintenance. The constriction of the CB1RSMKO mesenteric artery in vitro was not significantly altered compared with that of the control mice. In contrast, the relaxation to CB1R agonist 2-AG or WIN55212-2 was decreased in CB1RSMKO vessels, suggesting that activation of CB1RSM mediates the vasodilation effect of cannabinoids. Ischemia stroke mouse model was used to further identify the potential function of CB1RSM in pathological conditions, and the results showed that the infarct volume in CB1RSMKO mice is significantly increased compared with the control littermates. These results suggest that vascular CB1R may not play a central role in basal vascular health maintenance but is protective in ischemia states, such as stroke. The protection function may be mediated, at least partly, by the relaxation effect of CB1RSM-dependent activities of endocannabinoids.


Subject(s)
Infarction, Middle Cerebral Artery/metabolism , Ischemic Stroke/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Cannabinoid, CB1/deficiency , Vasodilation , Animals , Blood Pressure , Disease Models, Animal , Endocannabinoids/metabolism , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Middle Cerebral Artery/metabolism , Middle Cerebral Artery/physiopathology , Muscle, Smooth, Vascular/physiopathology , Receptor, Cannabinoid, CB1/genetics , Signal Transduction , Vasoconstriction
3.
Am J Physiol Endocrinol Metab ; 321(1): E146-E155, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34097543

ABSTRACT

Cannabinoid 1 receptor (CB1R) inverse agonists reduce body weight and improve several parameters of glucose homeostasis. However, these drugs have also been associated with deleterious side effects. CB1R expression is widespread in the brain and in peripheral tissues, but whether specific sites of expression can mediate the beneficial metabolic effects of CB1R drugs, while avoiding the untoward side effects, remains unclear. Evidence suggests inverse agonists may act on key sites within the central nervous system to improve metabolism. The ventromedial hypothalamus (VMH) is a critical node regulating energy balance and glucose homeostasis. To determine the contributions of CB1Rs expressed in VMH neurons in regulating metabolic homeostasis, we generated mice lacking CB1Rs in the VMH. We found that the deletion of CB1Rs in the VMH did not affect body weight in chow- and high-fat diet-fed male and female mice. We also found that deletion of CB1Rs in the VMH did not alter weight loss responses induced by the CB1R inverse agonist SR141716. However, we did find that CB1Rs of the VMH regulate parameters of glucose homeostasis independent of body weight in diet-induced obese male mice.NEW & NOTEWORTHY Cannabinoid 1 receptors (CB1Rs) regulate metabolic homeostasis, and CB1R inverse agonists reduce body weight and improve parameters of glucose metabolism. However, the cell populations expressing CB1Rs that regulate metabolic homeostasis remain unclear. CB1Rs are highly expressed in the ventromedial hypothalamic nucleus (VMH), which is a crucial node that regulates metabolism. With CRISPR/Cas9, we generated mice lacking CB1Rs specifically in VMH neurons and found that CB1Rs in VMH neurons are essential for the regulation of glucose metabolism independent of body weight regulation.


Subject(s)
Body Weight/physiology , Glucose/metabolism , Homeostasis/physiology , Neurons/metabolism , Receptor, Cannabinoid, CB1/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Body Composition/physiology , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , Diet, High-Fat , Energy Metabolism/physiology , Female , Gene Editing , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics
4.
Cell Death Dis ; 11(12): 1044, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33298885

ABSTRACT

Cannabinoid 1 receptor (CB1R) expression is upregulated in the liver with viral hepatitis, cirrhosis, and both alcoholic and non-alcoholic fatty liver disease (FLD), whereas its expression is muted under usual physiological conditions. Inhibiting CB1R has been shown to be beneficial in preserving hepatic function in FLD but it is unclear if inhibiting CB1R during an inflammatory response to an acute hepatic injury, such as toxin-induced injury, would also be beneficial. We found that intrinsic CB1R in hepatocytes regulated liver inflammation-related gene transcription. We tested if nullification of hepatocyte-specific CB1R (hCNR1-/-) in mice protects against concanavalin A (Con A)-induced liver injury. We looked for evidence of liver damage and markers of inflammation in response to Con A by measuring liver enzyme levels and proinflammatory cytokines (e.g., TNF-α, IL-1ß, IL-6, IL-17) in serum collected from hCNR1-/- and control mice. We observed a shift to the right in the dose-response curve for liver injury and inflammation in hCNR1-/- mice. We also found less inflammatory cell infiltration and focal necrosis in livers of hCNR1-/- mice compared to controls, resulting from downregulated apoptotic markers. This anti-apoptotic mechanism results from increased activation of nuclear factor kappa B (NF-κB), especially cAMP-dependent cannabinoid signaling and membrane-bound TNF-α, via downregulated TNF-α receptor 2 (TNFR2) transcription levels. Collectively, these findings provide insight into involvement of CB1R in the pathogenesis of acute liver injury.


Subject(s)
Concanavalin A/toxicity , Hepatocytes/metabolism , Liver/metabolism , Liver/pathology , NF-kappa B/metabolism , Receptor, Cannabinoid, CB1/deficiency , Signal Transduction , Animals , Apoptosis/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Hepatocytes/drug effects , Inflammation/pathology , Liver/drug effects , Male , Mice , Models, Biological , Protein Binding , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
5.
Brain Res Bull ; 160: 40-49, 2020 07.
Article in English | MEDLINE | ID: mdl-32294520

ABSTRACT

Endocannabinoid system activity contributes to the homeostatic defense against aging and thus may counteract the progression of brain aging. The cannabinoid type 1 (CB1) receptor activity declines with aging in the brain, which impairs neuronal network integrity and cognitive functions. However, the underlying mechanisms that link CB1 activity and memory decline remain unknown. Mitochondrial activity profoundly influences neuronal function, and age-dependent mitochondrial activity change is one of the known hallmarks of brain aging. As CB1 receptor is expressed on mitochondria and may regulate neuronal energy metabolism in hippocampus, we hypothesized that CB1 receptors might influence mitochondria in hippocampal neurons. Here, we found that CB1 receptor significantly affected mitochondrial autophagy (mitophagy) and morphology in an age-dependent manner. Serine 65-phosphorylated ubiquitin, a key marker for mitophagy, was reduced in adult CB1-deficient mice (CB1-KO) compared to those in wild type controls, particularly in CA1 pyramidal cell layer. Transmission electron microscopy (TEM) analysis showed reduced mitophagy-like events in hippocampus of adult CB1-KO. TEM analysis also showed that mitochondrial morphology in adult CB1-KO mice was altered shown by an increase in thin and elongated mitochondria in hippocampal neurons. 3D reconstruction of mitochondrial morphology after scanning electron microscopy additionally revealed an enhanced density of interconnected mitochondria. Altogether, these findings suggest that reduced CB1 signaling in CB1-KO mice leads to reduced mitophagy and abnormal mitochondrial morphology in hippocampal neurons during aging. These mitochondrial changes might be due to the impairments in mitochondrial quality control system, which links age-related decline in CB1 activity and impaired memory.


Subject(s)
Aging/metabolism , Autophagy/physiology , Hippocampus/metabolism , Mitochondrial Dynamics/physiology , Neurons/metabolism , Receptor, Cannabinoid, CB1/deficiency , Aging/pathology , Animals , Female , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology
6.
J Comp Neurol ; 528(1): 3-13, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31226222

ABSTRACT

Endocannabinoids and their receptors are highly abundant in the developing cerebral cortex and play major roles in early developmental processes, for example, neuronal proliferation, migration, and axonal guidance as well as postnatal plasticity. To investigate the role of the cannabinoid type 1 receptor (CB1) in the formation of sensory maps in the cerebral cortex, the topographic representation of the whiskers in the primary somatosensory cortex (barrel field) of adult mice with different cell type specific genetic deletion of CB1 was studied. A constitutive absence of CB1 (CB1-KO) significantly decreased the total area of the somatosensory cortical map, affecting barrel, and septal areas. Cell specific CB1 deletion in dorsal telencephalic glutamatergic neurons only (Glu-CB1-KO) or in both glutamatergic and forebrain GABAergic neurons (Glu/GABA-CB1-KO) resulted in an increased septa area in the barrel field map. No significant modifications in area parameters could be observed in GABA-CB1-KO mice. These data demonstrate that CB1 signaling especially in cortical glutamatergic neurons is essential for the development of topographic maps in the cerebral cortex.


Subject(s)
Brain Mapping/methods , Receptor, Cannabinoid, CB1/deficiency , Signal Transduction/physiology , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Receptor, Cannabinoid, CB1/analysis
7.
Neuroscience ; 421: 1-16, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31682822

ABSTRACT

The endocannabinoid system modulates synaptic transmission, controls neuronal excitability, and is involved in various brain functions including learning and memory. 2-arachidonoylglycerol, a major endocannabinoid produced by diacylglycerol lipase-α (DGLα), is released from postsynaptic neurons, retrogradely activates presynaptic CB1 cannabinoid receptors, and induces short-term or long-term synaptic plasticity. To examine whether and how the endocannabinoid system contributes to reward-based learning of a motor sequence, we subjected male CB1-knockout (KO) and DGLα-KO mice to three types of operant lever-press tasks. First, we trained mice to press one of three levers labeled A, B, and C for a food reward (one-lever task). Second, we trained mice to press the three levers in the order of A, B, and C (three-lever task). Third, the order of the levers was reversed to C, B, and A (reverse three-lever task). We found that CB1-KO mice and DGLα-KO mice exhibited essentially the same deficits in the operant lever-press tasks. In the one-lever task, both strains of knockout mice showed a slower rate of learning to press a lever for food. In the three-lever task, both strains of knockout mice showed a slower rate of learning of the motor sequence. In the reverse three-lever task, both strains of knockout mice needed more lever presses for reversal learning. These results suggest that the endocannabinoid system facilitates reward-based learning of a motor sequence by conferring the flexibility with which animals can switch between strategies.


Subject(s)
Arachidonic Acids/deficiency , Endocannabinoids/physiology , Glycerides/deficiency , Learning/physiology , Receptor, Cannabinoid, CB1/deficiency , Reward , Animals , Endocannabinoids/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Psychoneuroendocrinology ; 110: 104418, 2019 12.
Article in English | MEDLINE | ID: mdl-31491589

ABSTRACT

BACKGROUND: Obesity is a low-grade inflammation condition that facilitates the development of numerous comorbidities and the dysregulation of brain homeostasis. Additionally, obesity also causes distinct behavioral alterations both in humans and rodents. Here, we investigated the effect of inducible genetic deletion of the cannabinoid type 1 receptor (CB1) in adipocytes (Ati-CB1-KO mice) on obesity-induced memory deficits, depressive-like behavior, neuroinflammation and adult neurogenesis. METHODS: Behavioral, mRNA expression and immunohistochemical studies were performed in Ati-CB1-KO mice and corresponding wild-type controls under standard and high-fat diet. RESULTS: Adipocyte-specific CB1 deletion reversed metabolic disturbances associated with an obese condition confirming previous studies. As compared to obese mice, the metabolic amelioration in Ati-CB1-KO mice was associated with an improvement of mood-related behavior and recognition memory, concomitantly with an increase in cell proliferation in metabolic relevant neurogenic niches in hippocampus and hypothalamus. In mutant mice, these changes were related to an increased neuronal maturation/survival in the hippocampus. Furthermore, CB1 deletion in adipocytes was sufficient to reduce obesity-induced inflammation, gliosis and apoptosis in a brain region-specific manner. CONCLUSIONS: Overall our data provide compelling evidence of the physiological relevance of the adipocyte-brain crosstalk where adipocyte-specific CB1 influences obesity-related cognitive deficits and depression-like behavior, concomitantly with brain remodeling, such as adult neurogenesis and neuroinflammation in the hippocampus and hypothalamus.


Subject(s)
Adipocytes/metabolism , Depression/genetics , Diet, High-Fat/adverse effects , Memory Disorders/etiology , Neuritis/genetics , Neurogenesis/genetics , Receptor, Cannabinoid, CB1/genetics , Adult Stem Cells/physiology , Animals , Behavior, Animal/physiology , Brain/cytology , Brain/physiology , Depression/metabolism , Gene Deletion , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/psychology , Mice , Mice, Knockout , Neural Stem Cells/physiology , Neuritis/metabolism , Neuritis/pathology , Organ Specificity/genetics , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/metabolism
9.
Histochem Cell Biol ; 151(1): 5-20, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30196316

ABSTRACT

Surveys suggest that Cannabis provides benefit for people with inflammatory bowel disease. However, mechanisms underlying beneficial effects are not clear. We performed in situ hybridization RNAscope® combined with immunohistochemistry to show cell-specific distribution and regulation of cannabinoid receptor 1 and 2 (CB1, CB2), G protein-coupled receptor 55 (GPR55), and monoacylglycerol lipase (MGL) mRNA in immune cells using murine models of intestinal and systemic inflammation. In healthy animals, the presence in enteric ganglia is high for CB1 mRNA, but low for CB2 and GPR55 mRNAs. MGL mRNA is predominant throughout the intestinal wall including myenteric neurons, epithelium, circular and longitudinal muscular layers, and the lamina propria. Within the immune system, B220+ cells exhibit high gene expression for CB2 while the expression of CB2 in F4/80+ and CD3+ cells is less prominent. In contrast, GPR55 mRNA is highly present in F4/80+ and CD3+ cells. qRT-PCR of total colonic segments shows that the expression of GPR55 and MGL genes drops during intestinal inflammation. Also at cellular levels, GPR55 and MGL gene expression is reduced in F4/80+, but not CD3+ cells. As to systemic inflammation, reduced gene expression of MGL is observed in ileum by qRT-PCR, while at cellular levels, altered gene expression is also seen for CB1 and GPR55 in CD3+ but not F4/80+ cells. In summary, our study reveals changes in gene expression of members of the endocannabinoid system in situ attesting particularly GPR55 and MGL a distinct cellular role in the regulation of the immune response to intestinal and systemic inflammation.


Subject(s)
Asialoglycoproteins/metabolism , Endocannabinoids/metabolism , Inflammation/metabolism , Intestines/pathology , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Cannabinoid/metabolism , Animals , Asialoglycoproteins/analysis , Asialoglycoproteins/deficiency , Dextran Sulfate , Immunohistochemistry , In Situ Hybridization , Inflammation/chemically induced , Inflammation/pathology , Intestines/chemistry , Lectins, C-Type/analysis , Lectins, C-Type/deficiency , Lipopolysaccharides , Membrane Proteins/analysis , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/analysis , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB2/analysis , Receptor, Cannabinoid, CB2/deficiency , Receptors, Cannabinoid/analysis
10.
Eur J Neurosci ; 48(5): 2118-2138, 2018 09.
Article in English | MEDLINE | ID: mdl-30103253

ABSTRACT

Cannabinoids are capable of modulating mood, arousal, cognition and behavior, in part via their effects on the noradrenergic nucleus locus coeruleus (LC). Dysregulation of LC signaling and norepinephrine (NE) efflux in the medial prefrontal cortex (mPFC) can lead to the development of psychiatric disorders, and CB1r deletion results in alterations of α2- and ß1-adrenoceptors in the mPFC, suggestive of increased LC activity. To determine how CB1r deletion alters LC signaling, whole-cell patch-clamp electrophysiology was conducted in LC-NE neurons of male and female wild type (WT) and CB1r-knock out (KO) mice. CB1r deletion caused a significant increase in LC-NE excitability and input resistance in male but not female mice when compared to WT. CB1r deletion also caused adaptations in several indices of noradrenergic function. CB1r/CB2r-KO male mice had a significant increase in cortical NE levels and tyrosine hydroxylase and CRF levels in the LC compared to WT males. CB1r/CB2r-KO female mice showed a significant increase in LC α2-AR levels compared to WT females. To further probe actions of the endocannabinoid system as an anti-stress neuromediator, the effect of CB1r deletion on CRF-induced responses in the LC was investigated. The increase in LC-NE excitability observed in male and female WT mice following CRF (300 nM) bath application was not observed in CB1r-KO mice. These results indicate that cellular adaptations following CB1r deletion cause a disruption in LC-NE signaling in males but not females, suggesting underlying sex differences in compensatory mechanisms in KO mice as well as basal endocannabinoid regulation of LC-NE activity.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Norepinephrine/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Animals , Cannabinoids/metabolism , Female , Locus Coeruleus/drug effects , Male , Mice, Knockout , Neurons/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/deficiency , Sex Characteristics , Tyrosine 3-Monooxygenase/metabolism
11.
Neuropharmacology ; 139: 217-225, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30025920

ABSTRACT

Endogenous cannabinoids are diffusible lipid ligands of the main cannabinoid receptors type 1 and 2 (CB1R and CB2R). In the central nervous system endocannabinoids are produced in an activity-dependent manner and have been identified as retrograde modulators of synaptic transmission. Additionally, some neurons display a cell-autonomous slow self-inhibition (SSI) mediated by endocannabinoids. In these neurons, repetitive action potential firing triggers the production of endocannabinoids, which induce a long-lasting hyperpolarization of the membrane potential, rendering the cells less excitable. Different endocannabinoid receptors and effector mechanisms have been described underlying SSI in different cell types and brain areas. Here, we investigate SSI in neurons of layer 2/3 in the somatosensory cortex. High-frequency bursts of action potentials induced SSI in pyramidal cells (PC) and regular spiking non-pyramidal cells (RSNPC), but not in fast-spiking interneurons (FS). In RSNPCs the hyperpolarization was accompanied by a change in input resistance due to the activation of G protein-coupled inward-rectifying K+ (GIRK) channels. A CB2R-specific agonist induced the long-lasting hyperpolarization, whereas preincubation with a CB2R-specific inverse agonist suppressed SSI. Additionally, using cannabinoid receptor knockout mice, we found that SSI was still intact in CB1R-deficient but abolished in CB2R-deficient mice. Taken together, we describe an additional SSI mechanism in which the activity-induced release of endocannabinoids activates GIRK channels via CB2Rs. These findings expand our knowledge about cell type-specific differential neuronal cannabinoid receptor signaling and suggest CB2R-selective compounds as potential therapeutic approaches.


Subject(s)
Neural Inhibition/physiology , Neurons/metabolism , Receptor, Cannabinoid, CB2/metabolism , Somatosensory Cortex/metabolism , Animals , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neurons/drug effects , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/deficiency , Receptor, Cannabinoid, CB2/genetics , Somatosensory Cortex/drug effects , Tissue Culture Techniques
12.
Epilepsia ; 59(1): 79-91, 2018 01.
Article in English | MEDLINE | ID: mdl-29171003

ABSTRACT

OBJECTIVE: Status epilepticus (SE) is a life-threatening and commonly drug-refractory condition. Novel therapies are needed to rapidly terminate seizures to prevent mortality and morbidity. Monoacylglycerol lipase (MAGL) is the key enzyme responsible for the hydrolysis of the endocannabinoid 2-arachidonoylglycerol (2-AG) and a major contributor to the brain pool of arachidonic acid (AA). Inhibiting of monoacylglycerol lipase modulates synaptic activity and neuroinflammation, 2 mediators of excessive neuronal activation underlying seizures. We studied the effect of a potent and selective irreversible MAGL inhibitor, CPD-4645, on SE that was refractory to diazepam, its neuropathologic sequelae, and the mechanism underlying the drug's effects. METHODS: Diazepam-resistant SE was induced in adult mice fed with standard or ketogenic diet or in cannabinoid receptor type 1 (CB1) receptor knock-out mice. CPD-4645 (10 mg/kg, subcutaneously) or vehicle was dosed 1 and 7 h after status epilepticus onset in video-electroencephalography (EEG) recorded mice. At the end of SE, mice were examined in the novel object recognition test followed by neuronal cellloss analysis. RESULTS: CPD-4645 maximal plasma and brain concentrations were attained 0.5 h postinjection (half-life = 3.7 h) and elevated brain 2-AG levels by approximately 4-fold. CPD-4645 administered to standard diet-fed mice progressively reduced spike frequency during 3 h postinjection, thereby shortening SE duration by 47%. The drug immediately abrogated SE in ketogenic diet-fed mice. CPD-4645 rescued neuronal cell loss and cognitive deficit and reduced interleukin (IL)-1ß and cyclooxygenase 2 (COX-2) brain expression resulting from SE. The CPD-4645 effect on SE was similar in mice lacking CB1 receptors. SIGNIFICANCE: MAGL represents a novel therapeutic target for treating status epilepticus and improving its sequelae. CPD-4645 therapeutic effects appear to be predominantly mediated by modulation of neuroinflammation.


Subject(s)
Carbamates/therapeutic use , Monoacylglycerol Lipases/antagonists & inhibitors , Piperidines/therapeutic use , Status Epilepticus , Sulfonamides/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Brain Waves/drug effects , Brain Waves/physiology , Carbamates/chemistry , Carbamates/pharmacokinetics , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Diazepam/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistant Epilepsy/chemically induced , Drug Resistant Epilepsy/enzymology , Drug Resistant Epilepsy/therapy , Electroencephalography , Excitatory Amino Acid Agonists/toxicity , Fluoresceins/metabolism , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoacylglycerol Lipases/metabolism , Neurons/drug effects , Neurons/pathology , Piperidines/chemistry , Piperidines/pharmacology , Random Allocation , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Recognition, Psychology/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/enzymology , Status Epilepticus/therapy , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Time Factors
13.
Diabetes Obes Metab ; 20(3): 698-708, 2018 03.
Article in English | MEDLINE | ID: mdl-29106063

ABSTRACT

AIMS: To determine the specific role of podocyte-expressed cannabinoid-1 receptor (CB1 R) in the development of diabetic nephropathy (DN), relative to CB1 R in other renal cell types. MATERIAL AND METHODS: We developed a mouse model with a podocyte-specific deletion of CB1 R (pCB1Rko) and challenged this model with streptozotocin (STZ)-induced type-1 DN. We also assessed the podocyte response to high glucose in vitro and its effects on CB1 R activation. RESULTS: High glucose exposure for 48 hours led to an increase in CB1 R gene expression (CNR1) and endocannabinoid production in cultured human podocytes. This was associated with podocyte injury, reflected by decreased podocin and nephrin expression. These changes could be prevented by Cnr1-silencing, thus identifying CB1R as a key player in podocyte injury. After 12 weeks of chronic hyperglycaemia, STZ-treated pCB1Rko mice showed elevated blood glucose similar to that of their wild-type littermates. However, they displayed less albuminuria and less podocyte loss than STZ-treated wild-type mice. Unexpectedly, pCB1Rko mice also have milder tubular dysfunction, fibrosis and reduction of cortical microcirculation compared to wild-type controls, which is mediated, in part, by podocyte-derived endocannabinoids acting via CB1 R on proximal tubular cells. CONCLUSIONS: Activation of CB1 R in podocytes contributes to both glomerular and tubular dysfunction in type-1 DN, which highlights the therapeutic potential of peripheral CB1 R blockade.


Subject(s)
Diabetic Nephropathies/physiopathology , Kidney Glomerulus/physiology , Kidney Tubules, Proximal/physiology , Podocytes/metabolism , Receptors, Cannabinoid/deficiency , Animals , Arginase/metabolism , Cell Hypoxia/physiology , Cells, Cultured , Diabetes Mellitus, Experimental/physiopathology , Glucose/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/blood supply , Membrane Proteins/metabolism , Mice , Microcirculation/physiology , Oxidative Stress/physiology , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/metabolism
14.
Aging Cell ; 16(5): 1051-1061, 2017 10.
Article in English | MEDLINE | ID: mdl-28752643

ABSTRACT

The endocannabinoid system plays a role in regulating bone mass and bone cell activity and inactivation of the type 1 (Cnr1) or type 2 (Cnr2) cannabinoid receptors influences peak bone mass and age-related bone loss. As the Cnr1 and Cnr2 receptors have limited homology and are activated by different ligands, we have evaluated the effects of combined deficiency of Cnr1 and 2 receptors (Cnr1/2-/- ) on bone development from birth to old age and studied ovariectomy induced bone loss in female mice. The Cnr1/2-/- mice had accelerated bone accrual at birth when compared with wild type littermates, and by 3 months of age, they had higher trabecular bone mass. They were also significantly protected against ovariectomy-induced bone loss due to a reduction in osteoclast number. The Cnr1/2-/- mice had reduced age-related bone loss when compared with wild-type due to a reduction in osteoclast number. Although bone formation was reduced and bone marrow adiposity increased in Cnr1/2-/- mice, the osteoclast defect outweighed the reduction in bone formation causing preservation of bone mass with aging. This contrasts with the situation previously reported in mice with inactivation of the Cnr1 or Cnr2 receptors individually where aged-related bone loss was greater than in wild-type. We conclude that the Cnr1 and Cnr2 receptors have overlapping but nonredundant roles in regulating osteoclast and osteoblast activities. These observations indicate that combined inhibition of Cnr1 and Cnr2 receptors may be beneficial in preventing age-related bone loss, whereas blockade of individual receptors may be detrimental.


Subject(s)
Bone Density/genetics , Osteoclasts/metabolism , Osteoporosis/genetics , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Animals , Cell Count , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Femur/metabolism , Femur/pathology , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/pathology , Osteogenesis/genetics , Osteoporosis/etiology , Osteoporosis/pathology , Osteoporosis/prevention & control , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Ovariectomy/adverse effects , Primary Cell Culture , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB2/deficiency , Signal Transduction , Tibia/metabolism , Tibia/pathology
15.
J Pharmacol Exp Ther ; 362(2): 296-305, 2017 08.
Article in English | MEDLINE | ID: mdl-28592614

ABSTRACT

GW405833, widely accepted as a cannabinoid receptor 2 (CB2) agonist, suppresses pathologic pain in preclinical models without the unwanted central side effects of cannabinoid receptor 1 (CB1) agonists; however, recent in vitro studies have suggested that GW405833 may also behave as a noncompetitive CB1 antagonist, suggesting that its pharmacology is more complex than initially appreciated. Here, we further investigated the pharmacologic specificity of in vivo antinociceptive actions of GW405833 in models of neuropathic (i.e., partial sciatic nerve ligation model) and inflammatory (i.e., complete Freund's adjuvant model) pain using CB2 and CB1 knockout (KO) mice, their respective wild-type (WT) mice, and both CB2 and CB1 antagonists. GW405833 (3, 10, and 30 mg/kg i.p.) dose dependently reversed established mechanical allodynia in both pain models in WT mice; however, the antiallodynic effects of GW405833 were fully preserved in CB2KO mice and absent in CB1KO mice. Furthermore, the antiallodynic efficacy of GW405833 (30 mg/kg i.p.) was completely blocked by the CB1 antagonist rimonabant (10 mg/kg i.p.) but not by the CB2 antagonist SR144528 (10 mg/kg i.p.). Thus, the antinociceptive properties of GW405833 are dependent on CB1 receptors. GW405833 (30 mg/kg i.p.) was also inactive in a tetrad of tests measuring cardinal signs of CB1 activation. Additionally, unlike rimonabant (10 mg/kg i.p.), GW405833 (10 mg/kg, i.p.) did not act as a CB1 antagonist in vivo to precipitate withdrawal in mice treated chronically with Δ9-tetrahydrocannabinol. The present results suggest that the antiallodynic efficacy of GW405833 is CB1-dependent but does not seem to involve engagement of the CB1 receptor's orthosteric site.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Indoles/pharmacology , Morpholines/pharmacology , Neuralgia/drug therapy , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Cannabinoid Receptor Agonists/therapeutic use , Dose-Response Relationship, Drug , Female , Indoles/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/therapeutic use , Neuralgia/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB2/deficiency
16.
Mol Hum Reprod ; 23(7): 500-508, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28460003

ABSTRACT

STUDY QUESTION: What is the role of the endocannabinoid system (eCS) on the lipopolysaccharide (LPS) effects on uterine explants from 7-day pregnant mice in a murine model of endotoxin-induced miscarriage? SUMMARY ANSWER: We found evidence for cannabinoid receptor type2 (CB2) involvement in LPS-induced increased prostaglandin-F2α (PGF2α) synthesis and diminished cyclic adenosine monophosphate (cAMP) intracellular content in uterine explants from early pregnant mice. WHAT IS KNOWN ALREADY: Genital tract infections by Gram-negative bacteria are a common complication of human pregnancy that results in an increased risk of pregnancy loss. LPS, the main component of the Gram-negative bacterial wall, elicits a strong maternal inflammatory response that results in embryotoxicity and embryo resorption in a murine model endotoxin-induced early pregnancy loss. We have previously shown that the eCS mediates the embryotoxic effects of LPS, mainly via CB1 receptor activation. STUDY DESIGN, SIZE, DURATION: An in vitro study of mice uterine explants was performed to investigate the eCS in mediating the effects of LPS on PGF2α production and cAMP intracellular content. PARTICIPANTS/MATERIALS, SETTING, METHODS: Eight to 12-week-old virgin female BALB/c or CD1 (wild-type [WT] or CB1-knockout [CB1-KO]) mice were paired with 8- to 12-week-old BALB/c or CD1 (WT or CB1-KO) males, respectively. On day 7 of pregnancy, BALB/c, CD1 WT or CD1 CB1-KO mice were euthanized, the uteri were excised, implantation sites were removed and the uterine tissues were separated from decidual and embryo tissues. Uterine explants were cultured and exposed for an appropriate amount of time to different pharmacological treatments. The tissues were then collected for cAMP assay and PGF2α content determination by radioimmunoassay. MAIN RESULTS AND THE ROLE OF CHANCE: In vitro treatment of uteri explants from 7-day pregnant BALB/c or CD1 (WT or CB1-KO) mice with LPS induced an increased production of PGF2α (P < 0.05) and a reduction of the tissue content of cAMP (P < 0.05). These effects were mediated by CB2 receptors since exposure to AM630 (a specific CB2 receptor antagonist) prevented these LPS-induced effects (P < 0.05). Collectively, our results suggest a role for the eCS mediating LPS-induced deleterious effects on reproductive tissues. LIMITATIONS, REASONS FOR CAUTION: Since our experimental design involves in vitro experiments of uterine explants, the extrapolation of the results presented here to humans is limited. WIDER IMPLICATIONS OF THE FINDINGS: Our findings provide evidence for the role of CB2 receptors in reproductive events as well as their participation as a mediator of LPS deleterious effects on reproductive tissues. LARGE SCALE DATA: None. STUDY FUNDING AND COMPETING INTEREST(S): Dr Ana María Franchi was funded by Agencia Nacional para la Promoción Científica y Tecnológica (PICT 2010/0813 and PICT 2013/0097) and by Consejo Nacional de Investigaciones Científicas y Técnicas (PIP 2012/0061). Dr Carlos Davio was funded by Agencia Nacional para la Promoción Científica y Tecnológica (PICT 2013/2050). The authors have no competing interests.


Subject(s)
Abortion, Spontaneous/metabolism , Cyclic AMP/metabolism , Lipopolysaccharides/pharmacology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Uterus/drug effects , Abortion, Spontaneous/chemically induced , Abortion, Spontaneous/genetics , Abortion, Spontaneous/pathology , Animals , Cannabinoid Receptor Agonists/pharmacology , Cyclic AMP/antagonists & inhibitors , Dinoprost/biosynthesis , Disease Models, Animal , Female , Gene Deletion , Gene Expression , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Culture Techniques , Pregnancy , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB2/metabolism , Uterus/metabolism , Uterus/pathology
17.
Respir Physiol Neurobiol ; 240: 48-52, 2017 06.
Article in English | MEDLINE | ID: mdl-28254562

ABSTRACT

Endocannabinoids (ECs) are bioactive lipid mediators acting on two distinct cannabinoid receptors (CB1 and CB2), which are ubiquitously expressed in many tissues including the respiratory system. Despite numerous experimental data showing that cannabinomimetics influence respiration, the role of endogenously produced ECs in respiratory control has not been verified yet. Pulse oximetry was used in the present study to directly measure changes in respiratory parameters during elevation of EC levels. The cannabinoid reuptake inhibitor AM-404 (10mgkg-1, i.v.), but not its vehicle, induced a transient reduction of respiratory rate with a concomitant depression of arterial oxygen saturation and increase in breath distension in wild-type mice. In contrast, CB1 knock-out mice showed no alteration in any of these parameters upon administration of AM-404. Our results imply that the EC system has an important role in the physiological control of respiration by modulating the respiratory rate and consequently influencing arterial oxygen saturation. Furthermore, this mechanism is entirely dependent on CB1 receptors.


Subject(s)
Endocannabinoids/metabolism , Receptor, Cannabinoid, CB1/deficiency , Respiratory Insufficiency , Animals , Arachidonic Acids/pharmacology , Heart Rate/drug effects , Heart Rate/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oximetry , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , Receptor, Cannabinoid, CB1/genetics , Respiration/drug effects , Respiration/genetics , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/genetics , Respiratory Insufficiency/metabolism , Time Factors
18.
Cereb Cortex ; 27(11): 5303-5317, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28334226

ABSTRACT

Neuronal migration is a fundamental process of brain development, and its disruption underlies devastating neurodevelopmental disorders. The transcriptional programs governing this process are relatively well characterized. However, how environmental cues instruct neuronal migration remains poorly understood. Here, we demonstrate that the cannabinoid CB1 receptor is strictly required for appropriate pyramidal neuron migration in the developing cortex. Acute silencing of the CB1 receptor alters neuronal morphology and impairs radial migration. Consequently, CB1 siRNA-electroporated mice display cortical malformations mimicking subcortical band heterotopias and increased seizure susceptibility in adulthood. Importantly, rescuing the CB1 deficiency-induced radial migration arrest by knockdown of the GTPase protein RhoA restored the hyperexcitable neuronal network and seizure susceptibility. Our findings show that CB1 receptor/RhoA signaling regulates pyramidal neuron migration, and that deficient CB1 receptor signaling may contribute to cortical development malformations leading to refractory epilepsy independently of its canonical neuromodulatory role in the adult brain.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Pyramidal Cells/metabolism , Receptor, Cannabinoid, CB1/deficiency , Seizures/metabolism , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Disease Models, Animal , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Electroporation , Fluorescent Antibody Technique , Gene Knockdown Techniques , In Situ Hybridization , Mice, Transgenic , Microscopy, Confocal , Pentylenetetrazole , Pyramidal Cells/pathology , RNA, Small Interfering , Receptor, Cannabinoid, CB1/genetics , Seizures/pathology , Tissue Culture Techniques , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
19.
J Psychiatr Res ; 90: 46-59, 2017 07.
Article in English | MEDLINE | ID: mdl-28222356

ABSTRACT

The development of exaggerated avoidance behavior is largely responsible for the decreased quality of life in patients suffering from anxiety disorders. Studies using animal models have contributed to the understanding of the neural mechanisms underlying the acquisition of avoidance responses. However, much less is known about its extinction. Here we provide evidence in mice that learning about the safety of an environment (i.e., safety learning) rather than repeated execution of the avoided response in absence of negative consequences (i.e., response extinction) allowed the animals to overcome their avoidance behavior in a step-down avoidance task. This process was context-dependent and could be blocked by pharmacological (3 mg/kg, s.c.; SR141716) or genetic (lack of cannabinoid CB1 receptors in neurons expressing dopamine D1 receptors) inactivation of CB1 receptors. In turn, the endocannabinoid reuptake inhibitor AM404 (3 mg/kg, i.p.) facilitated safety learning in a CB1-dependent manner and attenuated the relapse of avoidance behavior 28 days after conditioning. Safety learning crucially depended on endocannabinoid signaling at level of the hippocampus, since intrahippocampal SR141716 treatment impaired, whereas AM404 facilitated safety learning. Other than AM404, treatment with diazepam (1 mg/kg, i.p.) impaired safety learning. Drug effects on behavior were directly mirrored by drug effects on evoked activity propagation through the hippocampal trisynaptic circuit in brain slices: As revealed by voltage-sensitive dye imaging, diazepam impaired whereas AM404 facilitated activity propagation to CA1 in a CB1-dependent manner. In line with this, systemic AM404 enhanced safety learning-induced expression of Egr1 at level of CA1. Together, our data render it likely that AM404 promotes safety learning by enhancing information flow through the trisynaptic circuit to CA1.


Subject(s)
Avoidance Learning/physiology , Extinction, Psychological/physiology , Hippocampus/metabolism , Animals , Arachidonic Acids/pharmacology , Avoidance Learning/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Early Growth Response Protein 1/metabolism , Extinction, Psychological/drug effects , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Voltage-Sensitive Dye Imaging
20.
Nature ; 539(7630): 555-559, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27828947

ABSTRACT

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.


Subject(s)
Cannabinoids/adverse effects , Memory Disorders/chemically induced , Memory/drug effects , Memory/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Adenylyl Cyclases/metabolism , Animals , Cannabinoids/metabolism , Cell Respiration/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Electron Transport/drug effects , Energy Metabolism/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/enzymology , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , NADH Dehydrogenase/metabolism , Oxidative Phosphorylation/drug effects , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...