Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Front Neuroanat ; 16: 1030060, 2022.
Article in English | MEDLINE | ID: mdl-36387996

ABSTRACT

Adult oligodendrocyte progenitor cells (OPCs) give rise to myelinating oligodendrocytes through life and play crucial roles in brain homeostasis and plasticity during health and disease. Cannabinoid compounds acting through CB1 receptors promote the proliferation and differentiation of OPCs in vitro and facilitate developmental myelination and myelin repair in vivo. However, CB1 receptor expression in adult OPCs in situ has not been corroborated by anatomical studies and the contribution of this receptor population to the (re)myelination effects of cannabinoids remains a matter of debate. Using electron microscopy methods applied to NG2-EYFP reporter mice we assessed the localization of CB1 receptors in OPCs of the adult mouse hippocampus. To control for the specificity of CB1 receptor immunostaining we generated transgenic mice bearing EYFP expression in NG2 glia and wild-type (NG2-EYFP-CB1 +/+) and knockout (NG2-EYFP-CB1 -/-) for CB1 receptors. Double immunogold and immunoperoxidase labeling for CB1 and EYFP, respectively, revealed that CB1 receptors are present in a low proportion of NG2 positive profiles within hippocampal stratum radiatum of NG2-EYFP-CB1 +/+ mice. Quantitative analysis of immunogold particles in synaptic structures and NG2 profiles showed that CB1 receptors are expressed at lower density in adult OPCs than in glutamatergic cells of the rodent hippocampus. These results highlight the presence of CB1 receptors in adult OPCs thus providing an anatomical substrate for the remyelination promoting effects of cannabinoids and open a novel perspective on the roles of the endocannabinoid system in brain physiology through the modulation of NG2 glia.

2.
Cell Death Dis ; 13(7): 585, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35798697

ABSTRACT

Cannabinoids are known to modulate oligodendrogenesis and developmental CNS myelination. However, the cell-autonomous action of these compounds on oligodendroglial cells in vivo, and the molecular mechanisms underlying these effects have not yet been studied. Here, by using oligodendroglial precursor cell (OPC)-targeted genetic mouse models, we show that cannabinoid CB1 receptors exert an essential role in modulating OPC differentiation at the critical periods of postnatal myelination. We found that selective genetic inactivation of CB1 receptors in OPCs in vivo perturbs oligodendrogenesis and postnatal myelination by altering the RhoA/ROCK signaling pathway, leading to hypomyelination, and motor and cognitive alterations in young adult mice. Conversely, pharmacological CB1 receptor activation, by inducing E3 ubiquitin ligase-dependent RhoA proteasomal degradation, promotes oligodendrocyte development and CNS myelination in OPCs, an effect that was not evident in OPC-specific CB1 receptor-deficient mice. Moreover, pharmacological inactivation of ROCK in vivo overcomes the defects in oligodendrogenesis and CNS myelination, and behavioral alterations found in OPC-specific CB1 receptor-deficient mice. Overall, this study supports a cell-autonomous role for CB1 receptors in modulating oligodendrogenesis in vivo, which may have a profound impact on the scientific knowledge and therapeutic manipulation of CNS myelination by cannabinoids.


Subject(s)
Cannabinoids , Oligodendrocyte Precursor Cells , Receptor, Cannabinoid, CB1 , Animals , Cannabinoids/pharmacology , Cell Differentiation/physiology , Gene Silencing , Mice , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Receptor, Cannabinoid, CB1/metabolism
3.
Br J Pharmacol ; 178(20): 4176-4192, 2021 10.
Article in English | MEDLINE | ID: mdl-34216154

ABSTRACT

BACKGROUND AND PURPOSE: Research on demyelinating disorders aims to find novel molecules that are able to induce oligodendrocyte precursor cell differentiation to promote central nervous system remyelination and functional recovery. Δ9 -Tetrahydrocannabinol (THC), the most prominent active constituent of the hemp plant Cannabis sativa, confers neuroprotection in animal models of demyelination. However, the possible effect of THC on myelin repair has never been studied. EXPERIMENTAL APPROACH: By using oligodendroglia-specific reporter mouse lines in combination with two models of toxin-induced demyelination, we analysed the effect of THC on the processes of oligodendrocyte regeneration and functional remyelination. KEY RESULTS: We show that THC administration enhanced oligodendrocyte regeneration, white matter remyelination and motor function recovery. THC also promoted axonal remyelination in organotypic cerebellar cultures. THC remyelinating action relied on the induction of oligodendrocyte precursor differentiation upon cell cycle exit and via CB1 cannabinoid receptor activation. CONCLUSIONS AND IMPLICATIONS: Overall, our study identifies THC administration as a promising pharmacological strategy aimed to promote functional CNS remyelination in demyelinating disorders.


Subject(s)
Demyelinating Diseases , Remyelination , White Matter , Animals , Cell Differentiation , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Dronabinol/pharmacology , Mice , Oligodendroglia
4.
Glia ; 69(3): 532-545, 2021 03.
Article in English | MEDLINE | ID: mdl-32956517

ABSTRACT

Δ9 -Tetrahydrocannabinol (THC), the main bioactive compound found in the plant Cannabis sativa, exerts its effects by activating cannabinoid receptors present in many neural cells. Cannabinoid receptors are also physiologically engaged by endogenous cannabinoid compounds, the so-called endocannabinoids. Specifically, the endocannabinoid 2-arachidonoylglycerol has been highlighted as an important modulator of oligodendrocyte (OL) development at embryonic stages and in animal models of demyelination. However, the potential impact of THC exposure on OL lineage progression during the critical periods of postnatal myelination has never been explored. Here, we show that acute THC administration at early postnatal ages in mice enhanced OL development and CNS myelination in the subcortical white matter by promoting oligodendrocyte precursor cell cycle exit and differentiation. Mechanistically, THC-induced-myelination was mediated by CB1 and CB2 cannabinoid receptors, as demonstrated by the blockade of THC actions by selective receptor antagonists. Moreover, the THC-mediated modulation of oligodendroglial differentiation relied on the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, as mTORC1 pharmacological inhibition prevented the THC effects. Our study identifies THC as an effective pharmacological strategy to enhance oligodendrogenesis and CNS myelination in vivo.


Subject(s)
Dronabinol , Endocannabinoids , Animals , Dronabinol/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Oligodendroglia , Receptors, Cannabinoid
5.
Biochem Pharmacol ; 157: 85-96, 2018 11.
Article in English | MEDLINE | ID: mdl-30118663

ABSTRACT

The endocannabinoid system exerts a crucial neuromodulatory role in many brain areas that is essential for proper regulation of neuronal activity. The role of cannabinoid signalling controlling neuronal activity in the adult brain is also evident when considering its contribution to adult brain insults or neurodegenerative diseases. In the context of brain genetic or acquired encephalopathies administration of cannabinoid-based molecules has demonstrated to exert symptomatic relief and hence, they are proposed as new potential therapeutic compounds. This review article summarizes the main evidences indicating the beneficial action of cannabinoid-derived molecules in preclinical models of neonatal hypoxia/ischemic damage. In a second part, we discuss the available evidences of therapeutic actions of cannabidiol in children with refractory epilepsy syndromes. Finally, we discuss the current view of cannabinoid signalling mechanisms active in the immature brain that affect in neural cell fate and can contribute to long-term neural cell plasticity.


Subject(s)
Brain/metabolism , Cannabinoids/therapeutic use , Endocannabinoids/metabolism , Epilepsy/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Animals , Brain/drug effects , Brain/growth & development , Brain/physiology , Humans , Hypoxia-Ischemia, Brain/etiology , Infant, Newborn , Receptors, Cannabinoid/metabolism , Signal Transduction
6.
J Neurosci ; 35(35): 12241-7, 2015 09 02.
Article in English | MEDLINE | ID: mdl-26338334

ABSTRACT

UNLABELLED: The identification of the molecular network that supports oligodendrocyte (OL) regeneration under demyelinating conditions has been a primary goal for regenerative medicine in demyelinating disorders. We recently described an essential function for TACE/ADAM17 in regulating oligodendrogenesis during postnatal myelination, but it is unknown whether this protein also plays a role in OL regeneration and remyelination under demyelinating conditions. By using genetic mouse models to achieve selective gain- or loss-of-function of TACE or EGFR in OL lineage cells in vivo, we found that TACE is critical for EGFR activation in OLs following demyelination, and therefore, for sustaining OL regeneration and CNS remyelination. TACE deficiency in oligodendrocyte progenitor cells following demyelination disturbs OL lineage cell expansion and survival, leading to a delay in the remyelination process. EGFR overexpression in TACE deficient OLs in vivo restores OL development and postnatal CNS myelination, but also OL regeneration and CNS remyelination following demyelination. Our study reveals an essential function of TACE in supporting OL regeneration and CNS remyelination that may contribute to the design of new strategies for therapeutic intervention in demyelinating disorders by promoting oligodendrocyte regeneration and myelin repair. SIGNIFICANCE STATEMENT: Oligodendrocyte (OL) regeneration has emerged as a promising new approach for the treatment of demyelinating disorders. By using genetic mouse models to selectively delete TACE expression in oligodendrocyte progenitors cells (OPs), we found that TACE/ADAM17 is required for supporting OL regeneration following demyelination. TACE genetic depletion in OPs abrogates EGFR activation in OL lineage cells, and perturbs cell expansion and survival, blunting the process of CNS remyelination. Moreover, EGFR overexpression in TACE-deficient OPs in vivo overcomes the defects in OL development during postnatal development but also OL regeneration during CNS remyelination. Our study identifies TACE as an essential player in OL regeneration that may provide new insights in the development of new strategies for promoting myelin repair in demyelinating disorders.


Subject(s)
ADAM Proteins/metabolism , Central Nervous System/pathology , Demyelinating Diseases/pathology , Gene Expression Regulation/physiology , Multiple Sclerosis/pathology , Oligodendroglia/physiology , Regeneration/physiology , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , ADAM17 Protein , Animals , Antigens/genetics , Antigens/metabolism , Cell Lineage/drug effects , Cell Lineage/genetics , Demyelinating Diseases/chemically induced , Disease Models, Animal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Oligodendroglia/drug effects , Oligodendroglia/ultrastructure , Proteoglycans/genetics , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Regeneration/drug effects
7.
J Neurosci ; 34(36): 11884-96, 2014 09 03.
Article in English | MEDLINE | ID: mdl-25186737

ABSTRACT

Several studies have elucidated the significance of a disintegrin and metalloproteinase proteins (ADAMs) in PNS myelination, but there is no evidence if they also play a role in oligodendrogenesis and CNS myelination. Our study identifies ADAM17, also called tumor necrosis factor-α converting enzyme (TACE), as a novel key modulator of oligodendrocyte (OL) development and CNS myelination. Genetic deletion of TACE in oligodendrocyte progenitor cells (OPs) induces premature cell cycle exit and reduces OL cell survival during postnatal myelination of the subcortical white matter (SCWM). These cellular and molecular changes lead to deficits in SCWM myelination and motor behavior. Mechanistically, TACE regulates oligodendrogenesis by modulating the shedding of EGFR ligands TGFα and HB-EGF and, consequently, EGFR signaling activation in OL lineage cells. Constitutive TACE depletion in OPs in vivo leads to similar alterations in CNS myelination and motor behavior as to what is observed in the EGFR hypofunctional mouse line EgfrWa2. EGFR overexpression in TACE-deficient OPs restores OL survival and development. Our study reveals an essential function of TACE in oligodendrogenesis, and demonstrates how this molecule modulates EGFR signaling activation to regulate postnatal CNS myelination.


Subject(s)
ADAM Proteins/metabolism , Brain/metabolism , Myelin Sheath/metabolism , Neurogenesis , Oligodendroglia/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Animals , Brain/growth & development , Brain/physiology , Cell Line , Cell Lineage , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Locomotion , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Transforming Growth Factor alpha/metabolism
8.
J Neurosci ; 34(23): 7917-30, 2014 06 04.
Article in English | MEDLINE | ID: mdl-24899714

ABSTRACT

Research on myelination has focused on identifying molecules capable of inducing oligodendrocyte (OL) differentiation in an effort to develop strategies that promote functional myelin regeneration in demyelinating disorders. Here, we show that transforming growth factor ß (TGFß) signaling is crucial for allowing oligodendrocyte progenitor (OP) cell cycle withdrawal, and therefore, for oligodendrogenesis and postnatal CNS myelination. Enhanced oligodendrogenesis and subcortical white matter (SCWM) myelination was detected after TGFß gain of function, while TGFß receptor II (TGFß-RII) deletion in OPs prevents their development into mature myelinating OLs, leading to SCWM hypomyelination in mice. TGFß signaling modulates OP cell cycle withdrawal and differentiation through the transcriptional modulation of c-myc and p21 gene expression, mediated by the interaction of SMAD3/4 with Sp1 and FoxO1 transcription factors. Our study is the first to demonstrate an autonomous and crucial role of TGFß signaling in OL development and CNS myelination, and may provide new avenues in the treatment of demyelinating diseases.


Subject(s)
Cell Cycle/physiology , Central Nervous System/metabolism , Myelin Sheath/metabolism , Oligodendroglia/physiology , Signal Transduction/physiology , Stem Cells/physiology , Transforming Growth Factor beta/metabolism , Age Factors , Animals , Animals, Newborn , Benzamides/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Central Nervous System/cytology , Dioxoles/pharmacology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/drug effects , Signal Transduction/drug effects , Smad3 Protein/genetics , Smad3 Protein/metabolism , Stem Cells/drug effects
9.
Int J Neuropsychopharmacol ; 16(6): 1407-19, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23298518

ABSTRACT

Cannabidiol (CBD), the main non-psychotomimetic component of the plant Cannabis sativa, exerts therapeutically promising effects on human mental health such as inhibition of psychosis, anxiety and depression. However, the mechanistic bases of CBD action are unclear. Here we investigate the potential involvement of hippocampal neurogenesis in the anxiolytic effect of CBD in mice subjected to 14 d chronic unpredictable stress (CUS). Repeated administration of CBD (30 mg/kg i.p., 2 h after each daily stressor) increased hippocampal progenitor proliferation and neurogenesis in wild-type mice. Ganciclovir administration to GFAP-thymidine kinase (GFAP-TK) transgenic mice, which express thymidine kinase in adult neural progenitor cells, abrogated CBD-induced hippocampal neurogenesis. CBD administration prevented the anxiogenic effect of CUS in wild type but not in GFAP-TK mice as evidenced in the novelty suppressed feeding test and the elevated plus maze. This anxiolytic effect of CBD involved the participation of the CB1 cannabinoid receptor, as CBD administration increased hippocampal anandamide levels and administration of the CB1-selective antagonist AM251 prevented CBD actions. Studies conducted with hippocampal progenitor cells in culture showed that CBD promotes progenitor proliferation and cell cycle progression and mimics the proliferative effect of CB1 and CB2 cannabinoid receptor activation. Moreover, antagonists of these two receptors or endocannabinoid depletion by fatty acid amide hydrolase overexpression prevented CBD-induced cell proliferation. These findings support that the anxiolytic effect of chronic CBD administration in stressed mice depends on its proneurogenic action in the adult hippocampus by facilitating endocannabinoid-mediated signalling.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Cannabidiol/therapeutic use , Hippocampus/drug effects , Neurogenesis/drug effects , Stress, Psychological/drug therapy , Animals , Anti-Anxiety Agents/pharmacology , Bromodeoxyuridine/metabolism , Camphanes/pharmacology , Cannabidiol/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cell Cycle/drug effects , Cell Line, Transformed , Cell Proliferation/drug effects , Disease Models, Animal , Feeding Behavior/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rimonabant , Stress, Psychological/pathology , Thymidine Kinase/metabolism
10.
Curr Med Res Opin ; 29(4): 291-303, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23252877

ABSTRACT

OBJECTIVE: A systematic review was conducted to analyze the tolerability of several oral iron supplements based on data obtained in available publications and to report the incidence of adverse effects (AEs) for each supplement both overall and gastrointestinal. METHODS: Electronic databases - Medline, the Cochrane Library, and Embase were searched for studies published up to January 2009. Clinical or observational studies reporting data on the tolerability of oral iron supplements were included. Results were described statistically and a quasi-binomial logistic regression model was developed to evaluate and compare the tolerability of the supplements studied. RESULTS: For this review 111 studies were included, with data on 10,695 patients. Ferrous sulfate with mucoproteose had the lowest incidence of AEs (4.1% for overall AEs, 3.7% for gastrointestinal AEs [GAEs]) and was used as the reference supplement in the regression model. Incidence rates of overall AEs for the other supplements were 7.3% for iron protein succinylate [GAEs: 7%; OR for AE compared to the reference supplement, 1.96], 23.5% for ferrous glycine sulfate [GAEs: 18.5%; OR: 5.90], 30.9% for ferrous gluconate [GAEs: 29.9%; OR: 11.06], 32.3% for ferrous sulfate without mucoproteose [GAEs: 30.2%; OR: 11.21], and 47.0% for ferrous fumarate [GAEs: 43.4%; OR: 19.87]. The differences in incidence of AEs between extended-release ferrous sulfate with mucoproteose and all other supplements except iron protein succinylate were statistically significant at p < 0.001. These findings are subject to some limitations as the designs and methodologies of the studies included show heterogeneity among them that has partially been counteracted by the large sample size provided by the substantial number of trials, which is considered a strength in tolerability studies. CONCLUSION: Extended-release ferrous sulfate with mucoproteose appears to be the best tolerated of the different oral iron supplements evaluated.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Dietary Supplements/adverse effects , Ferric Compounds/adverse effects , Ferrous Compounds/adverse effects , Ferric Compounds/administration & dosage , Ferrous Compounds/administration & dosage , Glycine/adverse effects , Glycine/analogs & derivatives , Humans , Metalloproteins/adverse effects , Succinates/adverse effects
11.
J Neurosci ; 32(47): 16651-65, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175820

ABSTRACT

The generation and specification of pyramidal neuron subpopulations during development relies on a complex network of transcription factors. The CB(1) cannabinoid receptor is the major molecular target of endocannabinoids and marijuana active compounds. This receptor has been shown to influence neural progenitor proliferation and axonal growth, but its involvement in neuronal differentiation and the functional impact in the adulthood caused by altering its signaling during brain development are not known. Here we show that the CB(1) receptor, by preventing Satb2 (special AT-rich binding protein 2)-mediated repression, increased Ctip2 (COUP-TF interacting protein 2) promoter activity, and Ctip2-positive neuron generation. Unbalanced neurogenic fate determination found in complete CB(1)(-/-) mice and in glutamatergic neuron-specific Nex-CB(1)(-/-) mice induced overt alterations in corticospinal motor neuron generation and subcerebral connectivity, thereby resulting in an impairment of skilled motor function in adult mice. Likewise, genetic deletion of CB(1) receptors in Thy1-YFP-H mice elicited alterations in corticospinal tract development. Altogether, these data demonstrate that the CB(1) receptor contributes to the generation of deep-layer cortical neurons by coupling endocannabinoid signals from the neurogenic niche to the intrinsic proneurogenic Ctip2/Satb2 axis, thus influencing appropriate subcerebral projection neuron specification and corticospinal motor function in the adulthood.


Subject(s)
Cell Differentiation/physiology , Matrix Attachment Region Binding Proteins/physiology , Motor Neurons/physiology , Pyramidal Cells/physiology , Pyramidal Tracts/physiology , Receptor, Cannabinoid, CB1/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Behavior, Animal/physiology , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Microscopy, Confocal , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Protein Kinase C/metabolism , Pyramidal Tracts/cytology , Real-Time Polymerase Chain Reaction
12.
J Biol Chem ; 287(2): 1198-209, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22102284

ABSTRACT

The endocannabinoid system is known to regulate neural progenitor (NP) cell proliferation and neurogenesis. In particular, CB(2) cannabinoid receptors have been shown to promote NP proliferation. As CB(2) receptors are not expressed in differentiated neurons, CB(2)-selective agonists are promising candidates to manipulate NP proliferation and indirectly neurogenesis by overcoming the undesired psychoactive effects of neuronal CB(1) cannabinoid receptor activation. Here, by using NP cells, brain organotypic cultures, and in vivo animal models, we investigated the signal transduction mechanism involved in CB(2) receptor-induced NP cell proliferation and neurogenesis. Exposure of hippocampal HiB5 NP cells to the CB(2) receptor-selective agonist HU-308 led to the activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin complex 1 (mTORC1) pathway, which, by inhibiting its downstream target p27Kip1, induced NP proliferation. Experiments conducted with the CB(2) receptor-selective antagonist SR144528, inhibitors of the PI3K/Akt/mTORC1 axis, and CB(2) receptor transient-transfection vector further supported that CB(2) receptors control NP cell proliferation via activation of mTORC1 signaling. Likewise, CB(2) receptor engagement induced cell proliferation in an mTORC1-dependent manner both in embryonic cortical slices and in adult hippocampal NPs. Thus, HU-308 increased ribosomal protein S6 phosphorylation and 5-bromo-2'-deoxyuridine incorporation in wild-type but not CB(2) receptor-deficient NPs of the mouse subgranular zone. Moreover, adult hippocampal NP proliferation induced by HU-308 and excitotoxicity was blocked by the mTORC1 inhibitor rapamycin. Altogether, these findings provide a mechanism of action and a rationale for the use of nonpsychotomimetic CB(2) receptor-selective ligands as a novel strategy for the control of NP cell proliferation and neurogenesis.


Subject(s)
Cell Proliferation , Hippocampus/metabolism , Neural Stem Cells/metabolism , Proteins/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/physiology , Animals , Camphanes/pharmacology , Cannabinoids/pharmacology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hippocampus/cytology , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Neural Stem Cells/cytology , Neurogenesis/drug effects , Neurogenesis/physiology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/genetics , Ribosomal Protein S6/genetics , Ribosomal Protein S6/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
13.
Brain ; 134(Pt 1): 119-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20929960

ABSTRACT

Endocannabinoids act as neuromodulatory and neuroprotective cues by engaging type 1 cannabinoid receptors. These receptors are highly abundant in the basal ganglia and play a pivotal role in the control of motor behaviour. An early downregulation of type 1 cannabinoid receptors has been documented in the basal ganglia of patients with Huntington's disease and animal models. However, the pathophysiological impact of this loss of receptors in Huntington's disease is as yet unknown. Here, we generated a double-mutant mouse model that expresses human mutant huntingtin exon 1 in a type 1 cannabinoid receptor-null background, and found that receptor deletion aggravates the symptoms, neuropathology and molecular pathology of the disease. Moreover, pharmacological administration of the cannabinoid Δ(9)-tetrahydrocannabinol to mice expressing human mutant huntingtin exon 1 exerted a therapeutic effect and ameliorated those parameters. Experiments conducted in striatal cells show that the mutant huntingtin-dependent downregulation of the receptors involves the control of the type 1 cannabinoid receptor gene promoter by repressor element 1 silencing transcription factor and sensitizes cells to excitotoxic damage. We also provide in vitro and in vivo evidence that supports type 1 cannabinoid receptor control of striatal brain-derived neurotrophic factor expression and the decrease in brain-derived neurotrophic factor levels concomitant with type 1 cannabinoid receptor loss, which may contribute significantly to striatal damage in Huntington's disease. Altogether, these results support the notion that downregulation of type 1 cannabinoid receptors is a key pathogenic event in Huntington's disease, and suggest that activation of these receptors in patients with Huntington's disease may attenuate disease progression.


Subject(s)
Corpus Striatum/metabolism , Huntington Disease/genetics , Neurons/metabolism , Receptor, Cannabinoid, CB1/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Survival , Dronabinol/pharmacology , Growth Hormone-Releasing Hormone/analogs & derivatives , Huntington Disease/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Receptor, Cannabinoid, CB1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rotarod Performance Test
14.
Brain ; 132(Pt 11): 3152-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19805493

ABSTRACT

Cannabinoid-derived drugs are promising agents for the development of novel neuroprotective strategies. Activation of neuronal CB(1) cannabinoid receptors attenuates excitotoxic glutamatergic neurotransmission, triggers prosurvival signalling pathways and palliates motor symptoms in animal models of neurodegenerative disorders. However, in Huntington's disease there is a very early downregulation of CB(1) receptors in striatal neurons that, together with the undesirable psychoactive effects triggered by CB(1) receptor activation, foster the search for alternative pharmacological treatments. Here, we show that CB(2) cannabinoid receptor expression increases in striatal microglia of Huntington's disease transgenic mouse models and patients. Genetic ablation of CB(2) receptors in R6/2 mice, that express human mutant huntingtin exon 1, enhanced microglial activation, aggravated disease symptomatology and reduced mice lifespan. Likewise, induction of striatal excitotoxicity in CB(2) receptor-deficient mice by quinolinic acid administration exacerbated brain oedema, microglial activation, proinflammatory-mediator state and medium-sized spiny neuron degeneration. Moreover, administration of CB(2) receptor-selective agonists to wild-type mice subjected to excitotoxicity reduced neuroinflammation, brain oedema, striatal neuronal loss and motor symptoms. Studies on ganciclovir-induced depletion of astroglial proliferation in transgenic mice expressing thymidine kinase under the control of the glial fibrillary acidic protein promoter excluded the participation of proliferating astroglia in CB(2) receptor-mediated actions. These findings support a pivotal role for CB(2) receptors in attenuating microglial activation and preventing neurodegeneration that may pave the way to new therapeutic strategies for neuroprotection in Huntington's disease as well as in other neurodegenerative disorders with a significant excitotoxic component.


Subject(s)
Huntington Disease , Microglia/metabolism , Neuroprotective Agents/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biomarkers/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Minocycline/pharmacology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Quinolinic Acid/pharmacology , Receptor, Cannabinoid, CB2/genetics , Rotarod Performance Test , Seizures/physiopathology
15.
Eur Arch Psychiatry Clin Neurosci ; 259(7): 371-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19588184

ABSTRACT

During brain development, functional neurogenesis is achieved by the concerted action of various steps that include the expansion of progenitor cells, neuronal specification, and establishment of appropriate synapses. Brain patterning and regionalization is regulated by a variety of extracellular signals and morphogens that, together with neuronal activity, orchestrate and regulate progenitor proliferation, differentiation, and neuronal maturation. In the adult brain, CB(1) cannabinoid receptors are expressed at very high levels in selective areas and are engaged by endocannabinoids, which act as retrograde messengers controlling neuronal function and preventing excessive synaptic activity. In addition, the endocannabinoid system is present at early developmental stages of nervous system formation. Recent studies have provided novel information on the role of this endogenous neuromodulatory system in the control of neuronal specification and maturation. Thus, cannabinoid receptors and locally produced endocannabinoids regulate neural progenitor proliferation and pyramidal specification of projecting neurons. CB(1) receptors also control axonal navigation, migration, and positioning of interneurons and excitatory neurons. Loss of function studies by genetic ablation or pharmacological blockade of CB(1) receptors interferes with long-range subcortical projections and, likewise, prenatal cannabinoid exposure induces different functional alterations in the adult brain. Potential implications of these new findings, such as the participation of the endocannabinoid system in the pathogenesis of neurodevelopmental disorders (e.g., schizophrenia) and the regulation of neurogenesis in brain depression, are discussed herein.


Subject(s)
Brain , Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Neurogenesis/physiology , Psychotic Disorders , Animals , Brain/cytology , Brain/growth & development , Brain/physiopathology , Cell Proliferation , Cognition/physiology , Humans , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Receptors, Cannabinoid/physiology , Signal Transduction/physiology , Stem Cells/physiology
16.
Menopause ; 16(5): 984-91, 2009.
Article in English | MEDLINE | ID: mdl-19407667

ABSTRACT

OBJECTIVE: There is increasing evidence to suggest that ossein-hydroxyapatite complex (OHC) is more effective than calcium supplements in maintaining bone mass. The aim of this meta-analysis was to determine whether OHC has a different clinical effect on bone mineral density (BMD) compared with calcium carbonate (CC). METHODS: A meta-analysis of randomized controlled clinical trials was carried out to evaluate the efficacy of OHC versus CC on trabecular BMD. We identified publications on clinical trials by a search of electronic databases, including MEDLINE (1966-November 2008), EMBASE (1974-November 2008), and the Cochrane Controlled Clinical Trials Register.The primary endpoint was percent change in BMD from baseline. Data were pooled in a random-effects model, and the weighted mean difference was calculated. A sensitivity analysis that excluded trials without full data was performed. RESULTS: Of the 18 controlled trials initially identified, 6 were included in the meta-analysis. There was no significant heterogeneity among the included trials. The percent change in BMD significantly favored the OHC group (1.02% [95% CI, 0.63-1.41], P < 0.00001). These results were confirmed in the sensitivity analysis. CONCLUSIONS: OHC is significantly more effective in preventing bone loss than CC.


Subject(s)
Bone Density/drug effects , Bone Diseases, Metabolic/prevention & control , Bone Resorption/prevention & control , Calcium Carbonate/therapeutic use , Durapatite/therapeutic use , Osteoporosis/prevention & control , Administration, Oral , Calcium Carbonate/pharmacology , Durapatite/pharmacology , Evidence-Based Practice , Humans , Randomized Controlled Trials as Topic , Research Design , Treatment Outcome
17.
Curr Pharm Des ; 14(23): 2279-88, 2008.
Article in English | MEDLINE | ID: mdl-18781978

ABSTRACT

Endocannabinoids act as retrograde messengers that, by inhibiting neurotransmitter release via presynaptic CB(1) cannabinoid receptors, regulate the functionality of many synapses. In addition, the endocannabinoid system participates in the control of neuron survival. Thus, CB(1) receptor activation has been shown to protect neurons from acute brain injury as well as in neuroinflammatory conditions and neurodegenerative diseases. Nonetheless, some studies have reported that cannabinoids can also exert neurotoxic actions. Cannabinoid neuroprotective activity relies on the inhibition of glutamatergic neurotransmission and on other various mechanisms, and is supported by the observation that the brain overproduces endocannabinoids upon damage. Coupling of neuronal CB(1) receptors to cell survival routes such as the phosphatidylinositol 3-kinase/Akt and extracellular signal-regulated kinase pathways may contribute to cannabinoid neuroprotective action. These pro-survival signals occur, at least in part, by the cross-talk between CB(1) receptors and growth factor tyrosine kinase receptors. Besides promoting neuroprotection, a role for the endocannabinoid system in the control of neurogenesis from neural progenitors has been put forward. In addition, activation of CB(2) cannabinoid receptors on glial cells may also participate in neuroprotection by limiting the extent of neuroinflammation. Altogether, these findings support that endocannabinoids constitute a new family of lipid mediators that act as instructive signals in the control of neuron survival.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction , Animals , Cell Survival/physiology , Humans , Neurodegenerative Diseases/physiopathology , Neuroglia/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
18.
J Biol Chem ; 283(19): 13320-9, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18334483

ABSTRACT

Cannabinoids are potential agents for the development of therapeutic strategies against multiple sclerosis. Here we analyzed the role of the peripheral CB(2) cannabinoid receptor in the control of myeloid progenitor cell trafficking toward the inflamed spinal cord and their contribution to microglial activation in an animal model of multiple sclerosis (experimental autoimmune encephalomyelitis, EAE). CB(2) receptor knock-out mice showed an exacerbated clinical score of the disease when compared with their wild-type littermates, and this occurred in concert with extended axonal loss, T-lymphocyte (CD4(+)) infiltration, and microglial (CD11b(+)) activation. Immature bone marrow-derived CD34(+) myeloid progenitor cells, which play a role in neuroinflammatory pathologies, were shown to express CB(2) receptors and to be abundantly recruited toward the spinal cords of CB(2) knock-out EAE mice. Bone marrow-derived cell transfer experiments further evidenced the increased contribution of these cells to microglial replenishment in the spinal cords of CB(2)-deficient animals. In line with these observations, selective pharmacological CB(2) activation markedly reduced EAE symptoms, axonal loss, and microglial activation. CB(2) receptor manipulation altered the expression pattern of different chemokines (CCL2, CCL3, CCL5) and their receptors (CCR1, CCR2), thus providing a mechanistic explanation for its role in myeloid progenitor recruitment during neuroinflammation. These findings demonstrate the protective role of CB(2) receptors in EAE pathology; provide evidence for a new site of CB(2) receptor action, namely the targeting of myeloid progenitor trafficking and its contribution to microglial activation; and support the potential use of non-psychoactive CB(2) agonists in therapeutic strategies for multiple sclerosis and other neuroinflammatory disorders.


Subject(s)
Cell Movement , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Receptor, Cannabinoid, CB2/deficiency , Receptor, Cannabinoid, CB2/genetics , Spinal Cord/cytology , Spinal Cord/metabolism
19.
J Biol Chem ; 282(33): 23892-8, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17556369

ABSTRACT

Endocannabinoids are lipid signaling mediators that exert an important neuromodulatory role and confer neuroprotection in several types of brain injury. Excitotoxicity and stroke can induce neural progenitor (NP) proliferation and differentiation as an attempt of neuroregeneration after damage. Here we investigated the mechanism of hippocampal progenitor cell engagement upon excitotoxicity induced by kainic acid administration and the putative involvement of the CB1 cannabinoid receptor in this process. Adult NPs express kainate receptors that mediate proliferation and neurosphere generation in vitro via CB1 cannabinoid receptors. Similarly, in vivo studies showed that excitotoxicity-induced hippocampal NPs proliferation and neurogenesis are abrogated in CB1-deficient mice and in wild-type mice administered with the selective CB1 antagonist rimonabant (N-piperidino-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-3-pyrazolecarboxamide; SR141716). Kainate stimulation increased basic fibroblast growth factor (bFGF) expression in cultured NPs in a CB1-dependent manner as this response was prevented by rimonabant and mimicked by endocannabinoids. Likewise, in vivo analyses showed that increased hippocampal expression of bFGF, as well as of brain-derived neurotrophic factor and epidermal growth factor, occurs upon excitotoxicity and that CB1 receptor ablation prevents this induction. Moreover, excitotoxicity increased the number of CB1+ bFGF+ cells, and this up-regulation preceded NP proliferation. In summary, our results show the involvement of the CB1 cannabinoid receptor in NP proliferation and neurogenesis induced by excitotoxic injury and support a role for bFGF signaling in this process.


Subject(s)
Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Nerve Regeneration/drug effects , Neurons/pathology , Neurotoxins/pharmacology , Receptor, Cannabinoid, CB1/physiology , Stem Cells/cytology , Animals , Cell Proliferation/drug effects , Hippocampus , Kainic Acid/pharmacology , Mice , Mice, Knockout , Neurons/drug effects , Receptor, Cannabinoid, CB1/deficiency , Stem Cells/drug effects
20.
Neuroscientist ; 13(2): 109-14, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404371

ABSTRACT

The endocannabinoid system exerts an important neuromodulatory function in different brain areas and is also known to be involved in the regulation of neural cell fate. Thus, CB(1) cannabinoid receptors are neuroprotective in different models of brain injury, and their expression is altered in various neurodegenerative diseases. Recent findings have demonstrated the presence of a functional endocannabinoid system in neural progenitor cells that participates in the regulation of cell proliferation and differentiation. In this Research Update, the authors address the experimental evidence regarding the regulatory role of cannabinoids in neurogenesis and analyze them in the context of those pathological disorders in which cannabinoid function and altered neuronal or glial generation is most relevant, for example, stroke and multiple sclerosis.


Subject(s)
Brain Diseases/metabolism , Brain/metabolism , Cannabinoid Receptor Modulators/metabolism , Cell Proliferation/drug effects , Endocannabinoids , Nerve Regeneration/physiology , Receptor, Cannabinoid, CB1/metabolism , Animals , Brain/physiopathology , Brain Diseases/drug therapy , Brain Diseases/physiopathology , Cannabinoid Receptor Modulators/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Humans , Nerve Regeneration/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB1/genetics , Stem Cells/drug effects , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...