Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
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
2.
Autophagy ; 16(12): 2289-2291, 2020 12.
Article in English | MEDLINE | ID: mdl-32981464

ABSTRACT

The recreational and medical use of cannabis is largely increasing worldwide. Cannabis use, however, can cause adverse side effects, so conducting innovative studies aimed to understand and potentially reduce cannabis-evoked harms is important. Previous research conducted on cultured neural cells had supported that CNR1/CB1R (cannabinoid receptor 1), the main molecular target of cannabis, affects macroautophagy/autophagy. However, it was not known whether CNR1 controls autophagy in the brain in vivo, and, eventually, what the functional consequences of a potential CNR1-autophagy connection could be. We have now found that Δ9-tetrahydrocannabinol (THC), the major intoxicating constituent of cannabis, impairs autophagy in the mouse striatum. Administration of autophagy activators (specifically, the rapalog temsirolimus and the disaccharide trehalose) rescues THC-induced autophagy inhibition and motor dyscoordination. The combination of various genetic strategies in vivo supports the idea that CNR1 molecules located on neurons belonging to the direct (striatonigral) pathway are required for the autophagy- and motor-impairing activity of THC. By identifying autophagy as a mechanistic link between THC and motor performance, our findings may open a new conceptual view on how cannabis acts in the brain.


Subject(s)
Cannabinoids , Animals , Autophagy , Brain , Dronabinol/pharmacology , Mice
3.
Elife ; 92020 08 10.
Article in English | MEDLINE | ID: mdl-32773031

ABSTRACT

The use of cannabis is rapidly expanding worldwide. Thus, innovative studies aimed to identify, understand and potentially reduce cannabis-evoked harms are warranted. Here, we found that Δ9-tetrahydrocannabinol, the psychoactive ingredient of cannabis, disrupts autophagy selectively in the striatum, a brain area that controls motor behavior, both in vitro and in vivo. Boosting autophagy, either pharmacologically (with temsirolimus) or by dietary intervention (with trehalose), rescued the Δ9-tetrahydrocannabinol-induced impairment of motor coordination in mice. The combination of conditional knockout mouse models and viral vector-mediated autophagy-modulating strategies in vivo showed that cannabinoid CB1 receptors located on neurons belonging to the direct (striatonigral) pathway are required for the motor-impairing activity of Δ9-tetrahydrocannabinol by inhibiting local autophagy. Taken together, these findings identify inhibition of autophagy as an unprecedented mechanistic link between cannabinoids and motor performance, and suggest that activators of autophagy might be considered as potential therapeutic tools to treat specific cannabinoid-evoked behavioral alterations.


Subject(s)
Autophagy/drug effects , Cannabinoids/pharmacology , Psychomotor Performance/drug effects , Putamen/physiology , Substantia Nigra/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Putamen/drug effects , Substantia Nigra/drug effects
4.
Nature ; 583(7817): 603-608, 2020 07.
Article in English | MEDLINE | ID: mdl-32641832

ABSTRACT

Astrocytes take up glucose from the bloodstream to provide energy to the brain, thereby allowing neuronal activity and behavioural responses1-5. By contrast, astrocytes are under neuronal control through specific neurotransmitter receptors5-7. However, whether the activation of astroglial receptors can directly regulate cellular glucose metabolism to eventually modulate behavioural responses is unclear. Here we show that activation of mouse astroglial type-1 cannabinoid receptors associated with mitochondrial membranes (mtCB1) hampers the metabolism of glucose and the production of lactate in the brain, resulting in altered neuronal functions and, in turn, impaired behavioural responses in social interaction assays. Specifically, activation of astroglial mtCB1 receptors reduces the phosphorylation of the mitochondrial complex I subunit NDUFS4, which decreases the stability and activity of complex I. This leads to a reduction in the generation of reactive oxygen species by astrocytes and affects the glycolytic production of lactate through the hypoxia-inducible factor 1 pathway, eventually resulting in neuronal redox stress and impairment of behavioural responses in social interaction assays. Genetic and pharmacological correction of each of these effects abolishes the effect of cannabinoid treatment on the observed behaviour. These findings suggest that mtCB1 receptor signalling can directly regulate astroglial glucose metabolism to fine-tune neuronal activity and behaviour in mice.


Subject(s)
Astrocytes/metabolism , Energy Metabolism , Glucose/metabolism , Mitochondria/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cannabinoid Receptor Agonists/pharmacology , Cells, Cultured , Dronabinol/pharmacology , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species/metabolism , Receptor, Cannabinoid, CB1/agonists , Social Behavior
5.
Neuropharmacology ; 150: 134-144, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30914306

ABSTRACT

Cannabinoids exert neuroprotection in a wide array of preclinical models. A number of these studies has focused on cannabinoid CB1 receptors in striatal medium spiny neurons (MSNs) and the most characteristic MSN-degenerative disease, Huntington's disease (HD). Accruing evidence supports that astrocytes contribute to drive HD progression, and that they express CB1 receptors, degrade endocannabinoids, and modulate endocannabinergic transmission. However, the possible role of the astroglial endocannabinoid system in controlling MSN integrity remains unknown. Here, we show that JZL-184, a selective inhibitor of monoacylglycerol lipase (MGL), the key enzyme that deactivates the endocannabinoid 2-arachidonoylglycerol, prevented the mutant huntingtin-induced up-regulation of the pro-inflammatory cytokine tumor necrosis factor-α in primary mouse striatal astrocytes via CB1 receptors. To study the role of astroglial MGL in vivo, we injected stereotactically into the mouse dorsal striatum viral vectors that encode mutant or normal huntingtin under the control of the glial fibrillary acidic protein promoter. We observed that, in wild-type mice, pharmacological blockade of MGL with JZL-184 (8 mg/kg/day, i.p.) conferred neuroprotection against mutant huntingtin-induced striatal damage, as evidenced by the prevention of MSN loss, astrogliosis, and motor coordination impairment. We next found that conditional mutant mice bearing a genetic deletion of MGL selectively in astroglial cells (MGLfloxed/floxed;GFAP-Cre/+ mice) were resistant to mutant huntingtin-induced MSN loss, astrogliosis, and motor coordination impairment. Taken together, these data support that astroglial MGL controls the availability of a 2-arachidonoylglycerol pool that ensues protection of MSNs in the mouse striatum in vivo, thus providing a potential druggable target for reducing striatal neurodegeneration.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Monoacylglycerol Lipases/metabolism , Neurons/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Benzodioxoles/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Glial Fibrillary Acidic Protein/metabolism , Huntington Disease/pathology , Mice , Monoacylglycerol Lipases/antagonists & inhibitors , Neurons/drug effects , Neurons/pathology , Piperidines/pharmacology
6.
Neuropsychopharmacology ; 43(5): 964-977, 2018 04.
Article in English | MEDLINE | ID: mdl-28102227

ABSTRACT

The dorsal striatum is a key node for many neurobiological processes such as motor activity, cognitive functions, and affective processes. The proper functioning of striatal neurons relies critically on metabotropic receptors. Specifically, the main adenosine and endocannabinoid receptors present in the striatum, ie, adenosine A2A receptor (A2AR) and cannabinoid CB1 receptor (CB1R), are of pivotal importance in the control of neuronal excitability. Facilitatory and inhibitory functional interactions between striatal A2AR and CB1R have been reported, and evidence supports that this cross-talk may rely, at least in part, on the formation of A2AR-CB1R heteromeric complexes. However, the specific location and properties of these heteromers have remained largely unknown. Here, by using techniques that allowed a precise visualization of the heteromers in situ in combination with sophisticated genetically modified animal models, together with biochemical and pharmacological approaches, we provide a high-resolution expression map and a detailed functional characterization of A2AR-CB1R heteromers in the dorsal striatum. Specifically, our data unveil that the A2AR-CB1R heteromer (i) is essentially absent from corticostriatal projections and striatonigral neurons, and, instead, is largely present in striatopallidal neurons, (ii) displays a striking G protein-coupled signaling profile, where co-stimulation of both receptors leads to strongly reduced downstream signaling, and (iii) undergoes an unprecedented dysfunction in Huntington's disease, an archetypal disease that affects striatal neurons. Altogether, our findings may open a new conceptual framework to understand the role of coordinated adenosine-endocannabinoid signaling in the indirect striatal pathway, which may be relevant in motor function and neurodegenerative diseases.


Subject(s)
Corpus Striatum/metabolism , Protein Structure, Quaternary , Receptor, Adenosine A2A/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction , Animals , Humans , Huntington Disease/metabolism , Mice , Neural Pathways/metabolism , Protein Subunits/biosynthesis
7.
Cereb Cortex ; 28(1): 307-322, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29121220

ABSTRACT

The vast majority of neurons within the striatum are GABAergic medium spiny neurons (MSNs), which receive glutamatergic input from the cortex and thalamus, and form two major efferent pathways: the direct pathway, expressing dopamine D1 receptor (D1R-MSNs), and the indirect pathway, expressing dopamine D2 receptor (D2R-MSNs). While molecular mechanisms of MSN degeneration have been identified in animal models of striatal damage, the molecular factors that dictate a selective vulnerability of D1R-MSNs or D2R-MSNs remain unknown. Here, we combined genetic, chemogenetic, and pharmacological strategies with behavioral and neurochemical analyses, and show that the pool of cannabinoid CB1 receptor (CB1R) located on corticostriatal terminals efficiently safeguards D1R-MSNs, but not D2R-MSNs, from different insults. This cell-specific response relies on the regulation of glutamatergic signaling, and is independent from the CB1R-dependent control of astroglial activity in the striatum. These findings define cortical CB1R as a pivotal synaptic player in dictating a differential vulnerability of D1R-MSNs versus D2R-MSNs, and increase our understanding of the role of coordinated cannabinergic-glutamatergic signaling in establishing corticostriatal circuits and its dysregulation in neurodegenerative diseases.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Genetic Vectors , Glutamic Acid/metabolism , Humans , Huntingtin Protein/administration & dosage , Huntingtin Protein/genetics , Huntingtin Protein/toxicity , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Receptor, Cannabinoid, CB1/genetics , Synaptic Transmission/physiology
8.
J Neurosci ; 36(41): 10611-10624, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27733612

ABSTRACT

The dorsal striatum is a major input structure of the basal ganglia and plays a key role in the control of vital processes such as motor behavior, cognition, and motivation. The functionality of striatal neurons is tightly controlled by various metabotropic receptors. Whereas the Gs/Gi-protein-dependent tuning of striatal neurons is fairly well known, the precise impact and underlying mechanism of Gq-protein-dependent signals remain poorly understood. Here, using different experimental approaches, especially designer receptor exclusively activated by designer drug (DREADD) chemogenetic technology, we found that sustained activation of Gq-protein signaling impairs the functionality of striatal neurons and we unveil the precise molecular mechanism underlying this process: a phospholipase C/Ca2+/proline-rich tyrosine kinase 2/cJun N-terminal kinase pathway. Moreover, engagement of this intracellular signaling route was functionally active in the mouse dorsal striatum in vivo, as proven by the disruption of neuronal integrity and behavioral tasks. To analyze this effect anatomically, we manipulated Gq-protein-dependent signaling selectively in neurons belonging to the direct or indirect striatal pathway. Acute Gq-protein activation in direct-pathway or indirect-pathway neurons produced an enhancement or a decrease, respectively, of activity-dependent parameters. In contrast, sustained Gq-protein activation impaired the functionality of direct-pathway and indirect-pathway neurons and disrupted the behavioral performance and electroencephalography-related activity tasks controlled by either anatomical framework. Collectively, these findings define the molecular mechanism and functional relevance of Gq-protein-driven signals in striatal circuits under normal and overactivated states. SIGNIFICANCE STATEMENT: The dorsal striatum is a major input structure of the basal ganglia and plays a key role in the control of vital processes such as motor behavior, cognition, and motivation. Whereas the Gs/Gi-protein-dependent tuning of striatal neurons is fairly well known, the precise impact and underlying mechanism of Gq-protein-dependent signals remain unclear. Here, we show that striatal circuits can be "turned on" by acute Gq-protein signaling or "turned off" by sustained Gq-protein signaling. Specifically, sustained Gq-protein signaling inactivates striatal neurons by an intracellular pathway that relies on cJun N-terminal kinase. Overall, this study sheds new light onto the molecular mechanism and functional relevance of Gq-protein-driven signals in striatal circuits under normal and overactivated states.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Neostriatum/physiology , Neural Pathways/physiology , Signal Transduction/physiology , Animals , Behavior, Animal/physiology , Calcium Signaling/physiology , Electroencephalography , Male , Mice , Mice, Inbred C57BL , Psychomotor Performance/physiology , Space Perception/physiology , Type C Phospholipases/physiology
9.
J Neurol ; 263(7): 1390-400, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27159993

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disease for which there is no curative treatment available. Given that the endocannabinoid system is involved in the pathogenesis of HD mouse models, stimulation of specific targets within this signaling system has been investigated as a promising therapeutic agent in HD. We conducted a double-blind, randomized, placebo-controlled, cross-over pilot clinical trial with Sativex(®), a botanical extract with an equimolecular combination of delta-9-tetrahydrocannabinol and cannabidiol. Both Sativex(®) and placebo were dispensed as an oral spray, to be administered up to 12 sprays/day for 12 weeks. The primary objective was safety, assessed by the absence of more severe adverse events (SAE) and no greater deterioration of motor, cognitive, behavioral and functional scales during the phase of active treatment. Secondary objectives were clinical improvement of Unified Huntington Disease Rating Scale scores. Twenty-six patients were randomized and 24 completed the trial. After ruling-out period and sequence effects, safety and tolerability were confirmed. No differences on motor (p = 0.286), cognitive (p = 0.824), behavioral (p = 1.0) and functional (p = 0.581) scores were detected during treatment with Sativex(®) as compared to placebo. No significant molecular effects were detected on the biomarker analysis. Sativex(®) is safe and well tolerated in patients with HD, with no SAE or clinical worsening. No significant symptomatic effects were detected at the prescribed dosage and for a 12-week period. Also, no significant molecular changes were observed on the biomarkers. Future study designs should consider higher doses, longer treatment periods and/or alternative cannabinoid combinations.Clincaltrals.gov identifier: NCT01502046.


Subject(s)
Huntington Disease/drug therapy , Plant Extracts/therapeutic use , Plant Structures , Adult , Amino Acids/pharmacology , Amyloid beta-Peptides/cerebrospinal fluid , Biogenic Monoamines/cerebrospinal fluid , Cannabidiol , Cross-Over Studies , Dronabinol , Drug Combinations , Endocannabinoids/genetics , Endocannabinoids/metabolism , Female , Fibroblasts/drug effects , Follow-Up Studies , Gene Expression Regulation/drug effects , Humans , Huntington Disease/blood , Huntington Disease/cerebrospinal fluid , Male , Mental Status Schedule , MicroRNAs/blood , Middle Aged , Outcome Assessment, Health Care , Peptide Fragments/cerebrospinal fluid , Pilot Projects , Severity of Illness Index , tau Proteins/cerebrospinal fluid
10.
Proc Natl Acad Sci U S A ; 111(22): 8257-62, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843137

ABSTRACT

The CB1 cannabinoid receptor, the main molecular target of endocannabinoids and cannabis active components, is the most abundant G protein-coupled receptor in the mammalian brain. Of note, CB1 receptors are expressed at the synapses of two opposing (i.e., GABAergic/inhibitory and glutamatergic/excitatory) neuronal populations, so the activation of one and/or another receptor population may conceivably evoke different effects. Despite the widely reported neuroprotective activity of the CB1 receptor in animal models, the precise pathophysiological relevance of those two CB1 receptor pools in neurodegenerative processes is unknown. Here, we first induced excitotoxic damage in the mouse brain by (i) administering quinolinic acid to conditional mutant animals lacking CB1 receptors selectively in GABAergic or glutamatergic neurons, and (ii) manipulating corticostriatal glutamatergic projections remotely with a designer receptor exclusively activated by designer drug pharmacogenetic approach. We next examined the alterations that occur in the R6/2 mouse, a well-established model of Huntington disease, upon (i) fully knocking out CB1 receptors, and (ii) deleting CB1 receptors selectively in corticostriatal glutamatergic or striatal GABAergic neurons. The data unequivocally identify the restricted population of CB1 receptors located on glutamatergic terminals as an indispensable player in the neuroprotective activity of (endo)cannabinoids, therefore suggesting that this precise receptor pool constitutes a promising target for neuroprotective therapeutic strategies.


Subject(s)
Cerebral Cortex/physiology , Corpus Striatum/physiology , Neurons/physiology , Receptor, Cannabinoid, CB1/physiology , Aged , Animals , Caenorhabditis elegans Proteins/metabolism , Cerebral Cortex/cytology , Corpus Striatum/cytology , Endocannabinoids/metabolism , Endocannabinoids/physiology , Endocannabinoids/therapeutic use , Female , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Glutamic Acid/metabolism , Humans , Integrases/genetics , Male , Mice , Mice, Knockout , Middle Aged , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Neurotoxins/metabolism , Organ Culture Techniques , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptors, GABA-A/metabolism , Synaptosomes/physiology
11.
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
12.
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
13.
J Surg Res ; 138(1): 111-20, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17173932

ABSTRACT

BACKGROUND: Palatoplasty has the undesired side effect of impaired mid-facial growth. To avoid this problem, we propose an alternative to palatoplasty. We hypothesize that if BMP-2 is injected together with a carrier into the periosteum of the cleft palate borders, border volume will increase and connective tissue cells will be activated to produce extra bone. Once these borders supported by bone reach the midline, extraction of their covering epithelia with trypsin will permit adhesion of the underlying tissues. We investigated in vitro the ability of cleft palate connective tissue cells to produce extra bone in the presence of BMP-2 and the possibility of using trypsin to remove the epithelium covering the cleft palate borders without impairing the underlying tissues' ability to adhere. MATERIALS AND METHODS: We used the cleft palate presented by tgf-beta(3) null mice and small fragments of human cleft palate mucoperiosteum as models. Immunolabeling BMP-2-treated or untreated cultures with TUNEL and anti-osteocalcin or PCNA antibodies was performed. The epithelium of the cleft palate borders was removed with a trypsin solution, and the de-epithelialized tissues were cultured in apposition. RESULTS: BMP-2 induces differentiation toward bone on cleft palate connective tissue cells without producing cell death or proliferation. Trypsin removal of the cleft palate margins' epithelium does not impair the underlying tissues' adhesion. CONCLUSION: It is possible to generate extra bone at the cleft palate margins and to chemically eliminate their covering epithelia without damaging the underlying tissues, which allows further investigation in vivo of this new approach for cleft palate closure.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Cleft Palate/drug therapy , Palate/drug effects , Palate/pathology , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/physiology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cleft Palate/pathology , Connective Tissue/drug effects , Connective Tissue/physiology , Female , Male , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Palate/physiology , Pregnancy , Tissue Culture Techniques , Transforming Growth Factor beta/physiology , Trypsin/pharmacology
14.
Dev Biol ; 265(1): 207-18, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14697364

ABSTRACT

Palate fusion is a complex process that involves the coordination of a series of cellular changes including cell death and epithelial to mesenchymal transition (EMT). Since members of the Snail family of zinc-finger regulators are involved in both triggering of the EMT and cell survival, we decided to study their putative role in palatal fusion. Furthermore, Snail genes are induced by transforming growth factor beta gene (TGF-beta) superfamily members, and TGF-beta(3) null mutant mice (TGF-beta(3)-/-) show a cleft palate phenotype. Here we show that in the wild-type mouse at the time of fusion, Snail is expressed in a few cells of the midline epithelial seam (MES), compatible with a role in triggering of the EMT in a small subpopulation of the MES. We also find an intriguing relationship between the expression of Snail family members and cell survival associated to the cleft palate condition. Indeed, Snail is expressed in the medial edge epithelial (MEE) cells in TGF-beta(3)-/-mouse embryo palates, where it is activated by the aberrant expression of its inducer, TGF-beta(1), in the underlying mesenchyme. In contrast to Snail-deficient wild-type pre-adhesion MEE cells, Snail-expressing TGF-beta(3) mutant MEE cells survive as they do their counterparts in the chick embryo. Interestingly, Slug is the Snail family member expressed in the chick MEE, providing another example of interchange of Snail and Slug expression between avian and mammalian embryos. We propose that in the absence of TGF-beta(3), TGF-beta(1) is upregulated in the mesenchyme, and that in both physiological (avian) and pathological (TGF-beta(3)-/-mammalian) cleft palates, it induces the expression of Snail genes promoting the survival of the MEE cells and permitting their subsequent differentiation into keratinized stratified epithelium.


Subject(s)
Cleft Palate/veterinary , DNA-Binding Proteins/physiology , Palate/embryology , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Zinc Fingers/physiology , Animals , Apoptosis/physiology , Cleft Palate/embryology , DNA-Binding Proteins/genetics , Epithelium/embryology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Snail Family Transcription Factors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...