Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Neuropharmacology ; 200: 108806, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34562441

ABSTRACT

Synapse pruning is essential not only for the developmental establishment of synaptic connections in the brain but also for the pathogenesis of neurodevelopmental and neurodegenerative disorders. However, there are no effective pharmacological means to regulate synaptic pruning during early development. Using the eye-specific segregation of the dorsal lateral geniculate nucleus (dLGN) as a model of synaptic pruning coupled with adenosine A2A receptor (A2AR) antagonism and knockout, we demonstrated while genetic deletion of the A2AR throughout the development attenuated eye-specific segregation with the attenuated microglial phagocytosis at postnatal day 5 (P5), selective treatment with the A2AR antagonist KW6002 at P2-P4 facilitated synaptic pruning of visual pathway with microglial activation, increased lysosomal activity in microglia and increased microglial engulfment of retinal ganglion cell (RGC) inputs in the dLGN at P5 (but not P10). Furthermore, KW6002-mediated facilitation of synaptic pruning was activity-dependent since tetrodotoxin (TTX) treatment abolished the KW6002 facilitation. Moreover, the A2AR antagonist also modulated postsynaptic proteins and synaptic density at early postnatal stages as revealed by the reduced immunoreactivity of postsynaptic proteins (Homer1 and metabotropic glutamate receptor 5) and colocalization of presynaptic VGlut2 and postsynaptic Homer1 puncta in the dLGN. These findings suggest that A2AR can control pruning by multiple actions involving the retinal wave, microglia engulfment, and postsynaptic stability. Thus, A2AR antagonists may represent a novel pharmacological strategy to modulate microglia-mediated synaptic pruning and treatment of neurodevelopmental disorders associated with dysfunctional pruning.


Subject(s)
Geniculate Bodies/drug effects , Microglia/drug effects , Neuronal Plasticity/drug effects , Receptor, Adenosine A2A/drug effects , Retinal Ganglion Cells/drug effects , Animals , Homer Scaffolding Proteins/drug effects , Mice , Mice, Knockout , Phagocytosis/drug effects , Purines , Receptor, Metabotropic Glutamate 5/drug effects , Tetrodotoxin/pharmacology
2.
Brain Res ; 1768: 147577, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34217728

ABSTRACT

The olfactory bulbectomized (OBX) rat model is a well-established model of depression in which antidepressant drugs reverse deficits in the passive avoidance test 14 days after administration. Recently, the olfactory bulbectomized rat model has been proposed to be a model of Alzheimer's disease (AD), and the available data indicate similarities between the changes that typically occur in AD and those observed in OBX animals. In the present study, the occurrence of neurochemical impairments related to AD were investigated 8 months after OB ablation. The expression of the nitric oxide synthases eNOS and nNOS, receptor for advanced glycation endproducts (RAGEs) and dimethylarginine dimethylaminohydrolase (DDAH1) in the prefrontal cortices (PFCs), hippocampi and striata of olfactory bulbectomized and sham-operated rats was evaluated. Subsequently, the impact of the administration of a positive allosteric modulator of the mGlu5 receptor, CDPPB (14 days, 2.5 or 5 mg/kg), on OBX-related changes was assessed. OB ablation induced typical deficits in passive avoidance. Significant aberrations in the expression of both isoforms of NOS were observed in the hippocampus and striatum, and the expression of DDAH1 was increased in the PFCs of OBX animals. CDPPB at a dose of 5 mg/kg ameliorated cognitive impairment in the passive avoidance test and partially reversed the changes in eNOS and nNOS expression induced by the lesion. The results of this study confirm that some of the neurochemical changes observed in OBX animals may resemble those associated with AD pathology and that activation of the mGlu5 receptor may partially counteract these pathological alterations.


Subject(s)
Cognitive Dysfunction/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Alzheimer Disease/physiopathology , Amidohydrolases/metabolism , Animals , Antidepressive Agents/metabolism , Benzamides/pharmacology , Brain/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Hippocampus/metabolism , Male , Nervous System/chemistry , Nitric Oxide Synthase/metabolism , Olfactory Bulb/physiology , Prefrontal Cortex/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/metabolism , Receptor, Metabotropic Glutamate 5/drug effects
3.
Pharmacol Ther ; 224: 107831, 2021 08.
Article in English | MEDLINE | ID: mdl-33705840

ABSTRACT

Methamphetamine abuse leads to devastating consequences, including addiction, crime, and death. Despite decades of research, no medication has been approved by the U.S. Food and Drug Administration for the treatment of Methamphetamine Use Disorder. Thus, there is a need for new therapeutic approaches. Animal studies demonstrate that methamphetamine exposure dysregulates forebrain function involving the Group-I metabotropic glutamate receptor subtype 5 (mGlu5), which is predominantly localized to postsynaptic sites. Allosteric modulators of mGlu5 offer a unique opportunity to modulate glutamatergic neurotransmission selectively, thereby potentially ameliorating methamphetamine-induced disruptions. Negative allosteric modulators of mGlu5 attenuate the effects of methamphetamine, including rewarding/reinforcing properties of the drug across animal models, and have shown promising effects in clinical trials for Anxiety Disorder and Major Depressive Disorder. Preclinical studies have also sparked great interest in mGlu5 positive allosteric modulators, which exhibit antipsychotic and anxiolytic properties, and facilitate extinction learning when access to methamphetamine is removed, possibly via the amelioration of methamphetamine-induced cognitive deficits. Clinical research is now needed to elucidate the mechanisms underlying the mGlu5 receptor-related effects of methamphetamine and the contributions of these effects to addictive behaviors. The growing array of mGlu5 allosteric modulators provides excellent tools for this purpose and may offer the prospect of developing tailored and effective medications for Methamphetamine Use Disorder.


Subject(s)
Amphetamine-Related Disorders , Methamphetamine , Receptor, Metabotropic Glutamate 5 , Amphetamine-Related Disorders/drug therapy , Animals , Clinical Trials as Topic , Humans , Receptor, Metabotropic Glutamate 5/drug effects
4.
J Neurochem ; 156(2): 225-248, 2021 01.
Article in English | MEDLINE | ID: mdl-31926033

ABSTRACT

We have previously shown that treatment with a mGluR5 positive allosteric modulator (PAM) is neuroprotective after experimental traumatic brain injury (TBI), limiting post-traumatic neuroinflammation by reducing pro-inflammatory microglial activation and promoting anti-inflammatory and neuroprotective responses. However, the specific molecular mechanisms governing this anti-inflammatory shift in microglia remain unknown. Here we show that the mGluR5 PAM, VU0360172 (VuPAM), regulates microglial inflammatory responses through activation of Akt, resulting in the inhibition of GSK-3ß. GSK-3ß regulates the phosphorylation of CREB, thereby controlling the expression of inflammation-related genes and microglial plasticity. The anti-inflammatory action of VuPAM in microglia is reversed by inhibiting Akt/GSK-3ß/CREB signaling. Using a well-characterized TBI model and CX3CR1gfp/+ mice to visualize microglia in vivo, we demonstrate that VuPAM enhances Akt/GSK-3ß/CREB signaling in the injured cortex, as well as anti-inflammatory microglial markers. Furthermore, in situ analysis revealed that GFP + microglia in the cortex of VuPAM-treated TBI mice co-express pCREB and the anti-inflammatory microglial phenotype marker YM1. Taken together, our data show that VuPAM decreases pro-inflammatory microglial activation by modulating Akt/GSK-3ß/CREB signaling. These findings serve to clarify the potential neuroprotective mechanisms of mGluR5 PAM treatment after TBI, and suggest novel therapeutic targets for post-traumatic neuroinflammation. Cover Image for this issue: https://doi.org/10.1111/jnc.15048.


Subject(s)
Brain Injuries, Traumatic/metabolism , Microglia/drug effects , Neuroprotective Agents/pharmacology , Niacinamide/analogs & derivatives , Receptor, Metabotropic Glutamate 5/drug effects , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Microglia/metabolism , Niacinamide/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/physiology
5.
Addict Biol ; 26(2): e12913, 2021 03.
Article in English | MEDLINE | ID: mdl-32339332

ABSTRACT

Intracellular interactions between protein kinases and metabotropic receptors in the striatum regulate behavioral changes in response to drug exposure. We investigated the difference in the degree of interaction between extracellular signal-regulated kinase (ERK) and metabotropic glutamate receptor subtype 5 (mGluR5) in the nucleus accumbens (NAc) after repeated exposure to nicotine in adult and adolescent rats. The results showed that repeated exposure to nicotine (0.5 mg/kg/day, s.c.) for seven consecutive days increased ERK phosphorylation more in adults than in adolescents. Furthermore, membrane expression of mGluR5 in gamma-aminobutyric acid (GABA) medium spiny neurons was higher in adults than adolescents as a result of repeated exposure to nicotine. Blockade of mGluR5 with MPEP (0.5 nmol/side) decreased the repeated nicotine-induced increase in ERK phosphorylation. Either blockade of mGluR5 or inhibition of ERK with SL327 (150 nmol/side) decreased the repeated nicotine-induced increase in the level of inositol-1,4,5-triphosphate (IP3 ), a key transducer associated with mGluR5-coupled signaling cascades. Similarly, interference of binding between activated ERK and mGluR5 by the blocking peptide, Tat-mGluR5-i (2 nmol/side), decreased the repeated nicotine-induced increases in IP3 and locomotor activity in adults. These findings suggest that the intracellular interaction between ERK and mGluR5 in the NAc is stronger in adult than in adolescent rats, which enhances the understanding of age-associated behavioral changes that occur after repeated exposure to nicotine.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Nicotine/pharmacology , Nucleus Accumbens/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Adolescent , Adult , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Humans , Male , Nicotine/administration & dosage , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
6.
J Psychiatry Neurosci ; 46(1): E1-E13, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32559027

ABSTRACT

Background: Glutamate transmission is implicated in drug-induced behavioural sensitization and the associated long-lasting increases in mesolimbic output. Metabotropic glutamate type 5 (mGlu5) receptors might be particularly important, but most details are poorly understood. Methods: We first assessed in mice (n = 51, all male) the effects of repeated dextroamphetamine administration (2.0 mg/kg, i.p.) on locomotor activity and binding of the mGlu5 ligand [3H]ABP688. In a parallel study, in 19 stimulant-drug-naïve healthy human volunteers (14 female) we administered 3 doses of dextroamphetamine (0.3 mg/kg, p.o.) or placebo, followed by a fourth dose 2 weeks later. We measured [11C]ABP688 binding using positron emission tomography before and after the induction phase. We assessed psychomotor and behavioural sensitization using speech rate, eye blink rate and self-report. We measured the localization of mGlu5 relative to synaptic markers in mouse striatum using immunofluorescence. Results: We observed amphetamine-induced psychomotor sensitization in mice and humans. We did not see group differences in mGlu5 availability following 3 pre-challenge amphetamine doses, but group differences did develop in mice administered 5 doses. In mice and humans, individual differences in mGlu5 binding after repeated amphetamine administration were negatively correlated with the extent of behavioural sensitization. In drug-naïve mice, mGlu5 was expressed at 67% of excitatory synapses on dendrites of striatal medium spiny neur. Limitations: Correlational results should be interpreted as suggestive because of the limited sample size. We did not assess sex differences. Conclusion: Together, these results suggest that changes in mGlu5 availability are not part of the earliest neural adaptations in stimulant-induced behavioural sensitization, but low mGlu5 binding might identify a higher propensity for sensitization.


Subject(s)
Central Nervous System Sensitization/drug effects , Central Nervous System Stimulants/pharmacology , Corpus Striatum , Dextroamphetamine/pharmacology , Locomotion/drug effects , Prefrontal Cortex , Psychomotor Performance/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Adult , Animals , Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dextroamphetamine/administration & dosage , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oximes/pharmacokinetics , Positron-Emission Tomography , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyridines/pharmacokinetics , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
7.
ACS Chem Neurosci ; 11(7): 1048-1057, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32167745

ABSTRACT

The metabotropic glutamate receptor subtype mGluR5 has been proposed as a potential drug target for CNS disorders such as anxiety, depression, Parkinson's disease, and epilepsy. The AstraZeneca compound AZD9272 has previously been labeled with carbon-11 and used as a PET radioligand for mGluR5 receptor binding. The molecular structure of AZD9272 allows one to label the molecule with fluorine-18 without altering the structure. The aim of this study was to develop a fluorine-18 analogue of AZD9272 and to examine its binding distribution in the nonhuman primate brain in vivo as well as to obtain whole body radiation dosimetry. 18F-AZD9272 was successfully synthesized from a nitro precursor. The radioligand was stable, with a radiochemical purity of >99% at 2 h after formulation in a sterile phosphate buffered solution (pH = 7.4). After injection of 18F-AZD9272 in two cynomolgus monkeys, the maximum whole brain radioactivity concentration was 4.9-6.7% of the injected dose (n = 2) and PET images showed a pattern of regional radioactivity consistent with that previously obtained for 11C-AZD9272. The percentage of parent radioligand in plasma was 59 and 64% (n = 2) at 120 min after injection of 18F-AZD9272, consistent with high metabolic stability. Two whole body PET scans were performed in nonhuman primates for a total of 231 min after injection of 18F-AZD9272. Highest uptakes were seen in liver and small intestine, followed by brain and kidney. The estimated effective dose was around 0.017 mSv/MBq. 18F-AZD9272 shows suitable properties as a PET radioligand for in vivo imaging of binding in the primate brain. 18F-labeled AZD9272 offers advantages over 11C-AZD9272 in terms of higher image resolution, combined with a longer half-life. Moreover, based on the distribution and the estimated radiation burden, imaging of 18F-AZD9272 could be used as an improved tool for quantitative assessment and characterization of AZD9272 binding sites in the human brain by using PET.


Subject(s)
Radiopharmaceuticals/metabolism , Receptor, Metabotropic Glutamate 5/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Tissue Distribution/physiology , Animals , Brain/metabolism , Brain/pathology , Carbon Radioisotopes/metabolism , Haplorhini , Macaca fascicularis , Oxadiazoles , Pyridines , Radiochemistry/methods , Radiometry/methods , Radiopharmaceuticals/pharmacology , Whole Body Imaging/methods
8.
Br J Pharmacol ; 177(2): 449-463, 2020 01.
Article in English | MEDLINE | ID: mdl-31658362

ABSTRACT

BACKGROUND AND PURPOSE: Marijuana is the illicit drug most commonly used among pregnant and breastfeeding women. Different studies reported long-term adverse effects induced by in utero exposure to the main component of marijuana, Δ9 -tetrahydrocannabinol (THC), both in rodents and in humans. However, little is known about any potential sex-dependent effects of marijuana consumption during pregnancy on newborns at early developmental ages. EXPERIMENTAL APPROACH: We studied the effects of prenatal exposure to the cannabinoid receptor agonist WIN55,212-2 (WIN; 0.5 mg·kg-1 from GD5 to GD20) on the emotional reactivity and cognitive performance of male and female rat offspring from infancy through adolescence and tested the role of mGlu5 receptor signalling in the observed effects. KEY RESULTS: Prenatally WIN-exposed male infant pups emitted less isolation-induced ultrasonic vocalizations compared with male control pups, when separated from the dam and siblings and showed increased locomotor activity while females were spared. These effects were normalized when male pups were treated with the positive allosteric modulator of mGlu5 receptor CDPPB. When tested at the prepubertal and pubertal periods, WIN-prenatally exposed rats of both sexes did not show any difference in social play behaviour, anxiety and temporal order memory. CONCLUSIONS AND IMPLICATIONS: We reveal a previously undisclosed sexual divergence in the consequences of fetal cannabinoids on newborns at early developmental ages, which is dependent on mGlu5 receptor signalling. These results provide new impetus for the urgent need to investigate the functional and behavioural substrates of prenatal cannabinoid exposure in both the male offspring and the female offspring.


Subject(s)
Behavior, Animal/drug effects , Benzoxazines/toxicity , Brain/drug effects , Cannabinoid Receptor Agonists/toxicity , Cognition/drug effects , Emotions/drug effects , Morpholines/toxicity , Naphthalenes/toxicity , Prenatal Exposure Delayed Effects , Receptor, Metabotropic Glutamate 5/drug effects , Age Factors , Animals , Benzamides/pharmacology , Brain/metabolism , Female , Locomotion/drug effects , Male , Memory/drug effects , Pregnancy , Pyrazoles/pharmacology , Rats, Wistar , Receptor, Metabotropic Glutamate 5/metabolism , Sex Factors , Social Behavior , Vocalization, Animal/drug effects
9.
Behav Brain Res ; 379: 112347, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31706797

ABSTRACT

Metaplastic effects of the NMDARs blocker ketamine at the neural and behavioural levels have been described as potential mechanisms underlying the beneficial effects in treatment-resistant depression. However, ketamine effects on addictive behaviours are still unexplored. In the present study, we investigated the effects of ketamine given under a "metaplasticity-inducing dose regimen" on sucrose-related renewal and contextual memory reconsolidation in rats. After a molecular analysis of ketamine modulation of GluN2B, GluA1 and mGluR5 receptors levels in nucleus accumbens, hippocampus and amygdala, two behavioural models were used to investigate ketamine effects: i) context-induced renewal of sucrose-seeking, and ii) sucrose memory reconsolidation. Ketamine was administrated 24 h before the renewal test or the retrieval. At the molecular level, ketamine i) decreased GluN2B, GluA1 and mGluR5 receptors in hippocampus, ii) decreased GluA1 and mGluR5 but increased GluN2B in nucleus accumbens and iii) increased GluN2B and mGluR5 in amygdala. At the behavioural level, ketamine given prior to renewal significantly inhibited responding compared to vehicle, while no significant effects were observed on reconsolidation of contextual memory. In conclusion, the molecular analysis of ketamine metaplastic effects in key brain areas suggest a possible involvement of glutamatergic receptors in the inhibition of sucrose renewal but not of contextual memory reconsolidation. The inhibition of renewal could be correlated to hippocampal and accumbal decreased levels of GluA1 and mGluR5, whereas, the lack of effect on contextual memory reconsolidation could be correlated to decreased GluN2B expression in hippocampus, landmark of destabilization-insensitive state.


Subject(s)
Amygdala/drug effects , Behavior, Animal/physiology , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/drug effects , Hippocampus/drug effects , Ketamine/pharmacology , Memory Consolidation/drug effects , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Receptors, AMPA/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Amygdala/metabolism , Animals , Excitatory Amino Acid Antagonists/administration & dosage , Hippocampus/metabolism , Ketamine/administration & dosage , Male , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
10.
Neuropharmacology ; 160: 107785, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31541651

ABSTRACT

Alzheimer's Disease (AD) is the most prevalent neurodegenerative disorder. Despite advances in the understanding of its pathophysiology, none of the available therapies prevents disease progression. Excess glutamate plays an important role in excitotoxicity by activating ionotropic receptors. However, the mechanisms modulating neuronal cell survival/death via metabotropic glutamate receptors (mGluRs) are not completely understood. Recent data indicates that CDPPB, a positive allosteric modulator of mGluR5, has neuroprotective effects. Thus, this work aimed to investigate CDPPB treatment effects on amyloid-ß (Aß) induced pathological alterations in vitro and in vivo and in a transgenic mouse model of AD (T41 mice). Aß induced cell death in primary cultures of hippocampal neurons, which was prevented by CDPPB. Male C57BL/6 mice underwent stereotaxic surgery for unilateral intra-hippocampal Aß injection, which induced memory deficits, neurodegeneration, neuronal viability reduction and decrease of doublecortin-positive cells, a marker of immature neurons and neuronal proliferation. Treatment with CDPPB for 8 days reversed neurodegeneration and doublecortin-positive cells loss and recovered memory function. Fourteen months old T41 mice presented cognitive deficits, neuronal viability reduction, gliosis and Aß accumulation. Treatment with CDPPB for 28 days increased neuronal viability (32.2% increase in NeuN+ cells) and reduced gliosis in CA1 region (Iba-1+ area by 31.3% and GFAP+ area by 37.5%) in transgenic animals, without inducing hepatotoxicity. However, it did not reverse cognitive deficit. Despite a four-week treatment did not prevent memory loss in aged transgenic mice, CDPPB is protective against Aß stimulus. Therefore, this drug represents a potential candidate for further investigations as AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Benzamides/pharmacology , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation , Amyloid beta-Peptides/adverse effects , Animals , Benzamides/administration & dosage , Disease Models, Animal , Hippocampus/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Peptide Fragments/adverse effects , Pyrazoles/administration & dosage , Receptor, Metabotropic Glutamate 5/metabolism
11.
Neuron ; 104(2): 338-352.e7, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31420117

ABSTRACT

Conventional antidepressants have limited efficacy and many side effects, highlighting the need for fast-acting and specific medications. Induction of the synaptic protein Homer1a mediates the effects of different antidepressant treatments, including the rapid action of ketamine and sleep deprivation (SD). We show here that mimicking Homer1a upregulation via intravenous injection of cell-membrane-permeable TAT-Homer1a elicits rapid antidepressant effects in various tests. Similar to ketamine and SD, in vitro and in vivo application of TAT-Homer1a enhances mGlu5 signaling, resulting in increased mTOR pathway phosphorylation, and upregulates synaptic AMPA receptor expression and activity. The antidepressant action of SD and Homer1a induction depends on mGlu5 activation specifically in excitatory CaMK2a neurons and requires enhanced AMPA receptor activity, translation, and trafficking. Moreover, our data demonstrate a pronounced therapeutic potential of different TAT-fused peptides that directly modulate mGlu5 and AMPA receptor activity and thus might provide a novel strategy for rapid and effective antidepressant treatment.


Subject(s)
Behavior, Animal/drug effects , Brain/metabolism , Depressive Disorder, Major/metabolism , Homer Scaffolding Proteins/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Receptors, AMPA/drug effects , Synapses/drug effects , Animals , Depressive Disorder, Major/genetics , Disease Models, Animal , Gene Products, tat , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Mice , Mice, Knockout , Peptide Fragments , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Sleep Deprivation/metabolism , Synapses/metabolism , TOR Serine-Threonine Kinases/drug effects , Up-Regulation
12.
J Neurochem ; 151(3): 301-315, 2019 11.
Article in English | MEDLINE | ID: mdl-31376155

ABSTRACT

Allosteric modulators of metabotropic glutamate receptor 5 (mGlu5 ) are a promising therapeutic strategy for a number of neurological disorders. Multiple mGlu5 -positive allosteric modulator (PAM) chemotypes have been discovered that act as either pure PAMs or as PAM-agonists in recombinant and native cells. While these compounds have been tested in paradigms of receptor activation, their effects on receptor regulatory processes are largely unknown. In this study, acute desensitization of mGlu5 mediated intracellular calcium mobilization by structurally diverse mGlu5 orthosteric and allosteric ligands was assessed in human embryonic kidney 293 cells and primary murine neuronal cultures from both striatum and cortex. We aimed to determine the intrinsic efficacy and modulatory capacity of diverse mGlu5 PAMs [(R)-5-((3-fluorophenyl)ethynyl)-N-(3-hydroxy-3-methylbutan-2-yl)picolinamide (VU0424465), N-cyclobutyl-6-((3-fluorophenyl)ethynyl)picolinamide (VU0360172), 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE), ((4-fluorophenyl) (2-(phenoxymethyl)-6,7-dihydrooxazolo[5,4-c]pyridin-5(4H)-yl)methanone) (VU0409551), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB)] on receptor desensitization and whether cellular context influences receptor regulatory processes. Only VU0424465 and VU0409551 induced desensitization alone in human embryonic kidney 293-mGlu5 cells, while all PAMs enhanced (S)-3,5-dihydroxyphenylglycine (DHPG)-induced desensitization. All mGlu5 PAMs induced receptor desensitization alone and enhanced DHPG-induced desensitization in striatal neurons. VU0424465 and VU0360172 were the only PAMs that induced desensitization alone in cortical neurons. With the exception of (CDPPB), PAMs enhanced DHPG-induced desensitization in cortical neurons. Moreover, differential apparent affinities, efficacies, and cooperativities with DHPG were observed for VU0360172, VU0409551, and VU0424465 when comparing receptor activation and desensitization in a cell type-dependent manner. These data indicate that biased mGlu5 allosteric modulator pharmacology extends to receptor regulatory processes in a tissue dependent manner, adding yet another layer of complexity to rational mGlu5 drug discovery.


Subject(s)
Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation/drug effects , Animals , Benzamides/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Oxazoles/pharmacology , Picolinic Acids/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism
13.
ACS Chem Neurosci ; 10(8): 3427-3436, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31132237

ABSTRACT

As part of the G-protein coupled receptor (GPCR) family, metabotropic glutamate (mGlu) receptors play an important role as drug targets of cognitive diseases. Selective allosteric modulators of mGlu subtype 5 (mGlu5) have the potential to alleviate symptoms of numerous central nervous system disorders such as schizophrenia in a more targeted fashion. Multiple mGlu5 positive allosteric modulators (PAMs), such as 1-(3-fluorophenyl)-N-((3-fluorophenyl)-methylideneamino)-methanimine (DFB), 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)-benzamide (CDPPB), and 4-nitro-N-(1,3-diphenyl-1H-pyrazol-5-yl)-benzamide (VU-29), exert their actions by binding to a defined allosteric site on mGlu5 located in the seven-transmembrane domain (7TM) and shared by mGlu5 negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)-pyridine (MPEP). Actions of the PAM N-{4-chloro-2-[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phenyl}-2-hydroxybenzamide (CPPHA) are mediated by a distinct allosteric site in the 7TM domain different from the MPEP binding site. Experimental evidence confirms these findings through mutagenesis experiments involving residues F585 (TM1) and A809 (TM7). In an effort to investigate mGlu5 PAM selectivity for this alternative allosteric site distinct from MPEP binding, we employed in silico quantitative structure-activity relationship (QSAR) modeling. Subsequent ligand-based virtual screening prioritized a set of 63 candidate compounds predicted from a library of over 4 million commercially available compounds to bind exclusively to this novel site. Experimental validation verified the biological activity for seven of 63 selected candidates. Further, medicinal chemistry optimizations based on these molecules revealed compound VU6003586 with an experimentally validated potency of 174 nM. Radioligand binding experiments showed only partial inhibition at very high concentrations, most likely indicative of binding at a non-MPEP site. Selective positive allosteric modulators for mGlu5 have the potential for tremendous impact concerning devastating neurological disorders such as schizophrenia and Huntington's disease. These identified and validated novel selective compounds can serve as starting points for more specifically tailored lead and probe molecules and thus help the development of potential therapeutic agents with reduced adverse effects.


Subject(s)
Drug Discovery/methods , Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Animals , High-Throughput Screening Assays , Humans , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
14.
J Med Chem ; 62(3): 1246-1273, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30624919

ABSTRACT

Negative allosteric modulators (NAMs) of the metabotropic glutamate receptor 5 (mGlu5) hold great promise for the treatment of a variety of central nervous system disorders. We have recently reported that prop-2-ynylidenecycloalkylamine derivatives are potent and selective NAMs of the mGlu5 receptor. In this work, we explored the amide, carbamate, sulfonamide, and urea derivatives of prop-2-ynylidenecycloalkylamine compounds with the aim of improving solubility and metabolic stability. In silico and experimental analyses were performed on the synthesized series of compounds to investigate structure-activity relationships. Compounds 12, 32, and 49 of the carbamate, urea, and amide classes, respectively, showed the most suitable cytochrome inhibition and metabolic stability profiles. Among them, compound 12 showed excellent selectivity, solubility, and stability profiles as well as suitable in vitro and in vivo pharmacokinetic properties. It was highly absorbed in rats and dogs and was active in anxiety, neuropathic pain, and lower urinary tract models.


Subject(s)
Excitatory Amino Acid Agents/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation , Amides/chemistry , Animals , Biological Availability , CHO Cells , Carbamates/chemistry , Cricetulus , Dogs , Excitatory Amino Acid Agents/chemistry , Excitatory Amino Acid Agents/pharmacokinetics , Humans , Ligands , Rats , Structure-Activity Relationship , Sulfonamides/chemistry , Urea/chemistry
15.
Addict Biol ; 24(4): 696-706, 2019 07.
Article in English | MEDLINE | ID: mdl-29790622

ABSTRACT

In a longitudinal rat model of alcohol consumption, we showed that exposure to alcohol decreased the concentration of glutamate in the prefrontal cortex, whereas a normalization occurred during abstinence. 18F-FPEB PET scans revealed that pre-exposure mGluR5 availability in the nucleus accumbens was associated with future alcohol preference. Finally, alcohol exposure induced a decrease in mGluR5 availability in the bilateral hippocampus and amygdala compared with baseline, and in the hippocampus and striatum compared with saccharin (Figure).


Subject(s)
Amygdala/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hippocampus/drug effects , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Alcohol Abstinence , Alcoholism , Amygdala/diagnostic imaging , Amygdala/metabolism , Animals , Fluorine Radioisotopes , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Nitriles , Nucleus Accumbens/diagnostic imaging , Nucleus Accumbens/metabolism , Positron-Emission Tomography , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Proton Magnetic Resonance Spectroscopy , Pyridines , Rats , Receptor, Metabotropic Glutamate 5/metabolism
16.
Clin Sci (Lond) ; 132(21): 2323-2338, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30389826

ABSTRACT

Metabotropic glutamate receptors belong to class C G-protein-coupled receptors and consist of eight subtypes that are ubiquitously expressed throughout the central nervous system. In recent years, the metabotropic glutamate receptor subtype 5 (mGlu5) has emerged as a promising target for a broad range of psychiatric and neurological disorders. Drug discovery programs targetting mGlu5 are primarily focused on development of allosteric modulators that interact with sites distinct from the endogenous agonist glutamate. Significant efforts have seen mGlu5 allosteric modulators progress into clinical trials; however, recent failures due to lack of efficacy or adverse effects indicate a need for a better understanding of the functional consequences of mGlu5 allosteric modulation. Biased agonism is an interrelated phenomenon to allosterism, describing how different ligands acting through the same receptor can differentially influence signaling to distinct transducers and pathways. Emerging evidence demonstrates that allosteric modulators can induce biased pharmacology at the level of intrinsic agonism as well as through differential modulation of orthosteric agonist-signaling pathways. Here, we present key considerations in the discovery and development of mGlu5 allosteric modulators and the opportunities and pitfalls offered by biased agonism and modulation.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System/drug effects , Excitatory Amino Acid Agonists/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Signal Transduction/drug effects , Animals , Binding Sites , Central Nervous System/metabolism , Central Nervous System Agents/chemistry , Central Nervous System Agents/metabolism , Excitatory Amino Acid Agonists/chemistry , Excitatory Amino Acid Agonists/metabolism , Glutamic Acid/metabolism , Humans , Ligands , Protein Binding , Protein Conformation , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
17.
J Neurochem ; 147(2): 222-239, 2018 10.
Article in English | MEDLINE | ID: mdl-30028018

ABSTRACT

Huntington's Disease (HD) is an autosomal-dominant neurodegenerative disorder, characterized by involuntary body movements, cognitive impairment, and psychiatric disorder. The metabotropic glutamate receptor 5 (mGluR5) plays an important role in HD and we have recently demonstrated that mGluR5-positive allosteric modulators (PAMs) can ameliorate pathology and the phenotypic signs of a mouse model of HD. In this study, we investigated the molecular mechanisms involved in mGluR5 PAMs effect on memory. Our results demonstrate that subchronic treatment with the mGluR5 PAM VU0409551 was effective in reversing the memory deficits exhibited by BACHD mice, a mouse model for HD. Moreover, VU0409551 treatment stabilized mGluR5 at the cellular plasma membrane of BACHD mice, increasing the expression of several genes important for synaptic plasticity, including c-Fos, brain-derived neurotrophic factor, Arc/Arg3.1, syntaxin 1A, and post-synaptic density-95. In addition, VU0409551 treatment also increased dendritic spine density and maturation and augmented the number of pre-synaptic sites. In conclusion, our results demonstrate that VU0409551 triggered the activation of cell signaling pathways important for synaptic plasticity, enhancing the level of dendritic spine maturation and rescuing BACHD memory impairment. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Huntington Disease/drug therapy , Huntington Disease/psychology , Memory Disorders/drug therapy , Memory Disorders/psychology , Neuronal Plasticity/drug effects , Oxazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Synapses/drug effects , Animals , Conditioning, Classical/drug effects , Dendritic Spines/drug effects , Gene Expression Regulation/drug effects , Huntington Disease/complications , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neuronal Plasticity/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Recognition, Psychology/drug effects , Signal Transduction/drug effects
18.
Brain Behav Immun ; 71: 93-107, 2018 07.
Article in English | MEDLINE | ID: mdl-29649522

ABSTRACT

Metabotropic glutamate receptor (mGlu)5 regulates microglia activation, which contributes to inflammation. However, the role of mGlu5 in neuroinflammation associated with Parkinson's disease (PD) remains unclear. Triptolide (T10) exerts potent immunosuppressive and anti-inflammatory effects and protects neurons by inhibiting microglia activation. In this study, we investigated the role of mGlu5 in the anti-inflammatory effect of T10 in a lipopolysaccharide (LPS)-induced PD model. In cultured BV2 cells and primary microglia, blocking mGlu5 activity or knocking down its expression abolished T10-inhibited release of proinflammatory cytokines induced by LPS. Moreover, T10 up-regulated mGlu5 expression decreased by LPS through enhancing mRNA expression and protein stability. T10 also reversed the reduction in mGlu5 membrane localization and modulated receptor-mediated mitogen-activated protein kinase activity induced by LPS. Pharmacological inhibition of signaling molecules increased nitric oxide level and inducible nitric oxide synthase (iNOS), tumor necrosis factor-α, and interleukin (IL)-1ß and -6 transcript levels that were downregulated by treatment with T10. Consistent with these in vitro findings, blocking mGlu5 attenuated the anti-inflammatory effects of T10 in an LPS-induced PD model and blocked the decreases in the number and morphology of ionized calcium binding adaptor molecule 1-positive microglia and LPS-induced iNOS protein expression caused by T10 treatment. Besides, mGlu5 mediated the effect of T10 on microglia-induced astrocyte activation in vitro and in vivo. The findings provide evidence for a novel mechanism by which mGlu5 regulates T10-inhibited microglia activation via modulating protein expression of the receptor and its intracellular signaling. The study might contribute to the biological effects of Chinese herbs as an approach for protecting against neurotoxicity in PD.


Subject(s)
Diterpenes/pharmacology , Microglia/metabolism , Phenanthrenes/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Animals , Cell Line , Disease Models, Animal , Diterpenes/metabolism , Dopaminergic Neurons/metabolism , Epoxy Compounds/metabolism , Epoxy Compounds/pharmacology , Inflammation/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Male , Mice , Microglia/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Parkinson Disease/metabolism , Phenanthrenes/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
19.
ACS Chem Neurosci ; 9(6): 1492-1502, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29522307

ABSTRACT

Metabotropic glutamate receptor 5 (mGlu5) plays a key role in synaptic information storage and memory, which is a well-known target for a variety of psychiatric and neurodegenerative disorders. In recent years, the increasing efforts have been focused on the design of allosteric modulators, and the negative allosteric modulators (NAMs) are the front-runners. Recently, the architecture of the transmembrane (TM) domain of mGlu5 receptor has been determined by crystallographic experiment. However, it has been not well understood how the pharmacophores of NAMs accommodated into the allosteric binding site. In this study, molecular dynamics (MD) simulations were performed on mGlu5 receptor bound with NAMs in preclinical or clinical development to shed light on this issue. In order to identify the key residues, the binding free energies as well as per-residue contributions for NAMs binding to mGlu5 receptor were calculated. Subsequently, the in silico site-directed mutagenesis of the key residues was performed to verify the accuracy of simulation models. As a result, the shared common features of the studied 5 clinically important NAMs (mavoglurant, dipraglurant, basimglurant, STX107, and fenobam) interacting with 11 residues in allosteric site were obtained. This comprehensive study presented a better understanding of mGlu5 receptor NAMs binding mechanism, which would be further used as a useful framework to assess and discover novel lead scaffolds for NAMs.


Subject(s)
Imidazoles/pharmacology , Molecular Dynamics Simulation , Protein Binding/drug effects , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Allosteric Regulation/drug effects , Humans , Molecular Docking Simulation/methods , Receptor, Metabotropic Glutamate 5/drug effects
20.
Toxicol Lett ; 288: 71-81, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29458170

ABSTRACT

Clinical epidemiological studies have shown that there is a link between Parkinson's disease (PD) and cancer, but how PD regulates cancer development remains unknown. In our study, the effect of metabotropic glutamate receptor 5 (mGlu5) on hepatoma was explored in a rotenone-induced PD model both in vitro and in vivo. We found that conditioned media derived from MN9D dopaminergic neuronal cells by rotenone-induced toxicity inhibited the growth, migration, invasion and promoted apoptosis of Hepa1-6 cells, which corresponded with decreased expression of mGlu5. Furthermore, treatment with 2-methyl-6-(phenylethynyl)pyridine (MPEP), a mGlu5 antagonist and knockdown of mGlu5, further reduced ATP levels and migration distance, and increased cleavage of caspase-3 in Hepa1-6 cells. Additionally, we found that conditioned media derived from rotenone-treated MN9D dopaminergic neuronal cells enhanced reactive oxygen species (ROS) generation and JNK phosphorylation, which could be further increased by MPEP treatment, and attenuated by mGlu5 agonist, (RS)-2-Chloro-5-hydroxyphenylglycine (CHPG) and ROS scavenger, N-acetyl-l-cysteine (NAC). The results indicated that down-regulation of mGlu5 promoted cell apoptosis through the intracellular ROS/JNK signaling pathway in a rotenone-induced cellular PD model. These findings were confirmed in vivo in a rotenone-induced rat model of PD combined with diethylnitrosamine (DEN)-induced hepatoma. Expression of Ki67 was decreased, and the levels of caspase-3 and p-JNK were increased in this model, which was accompanied by a decrease in protein expression of mGlu5. The study suggest that negative regulation of mGlu5 may inhibit hepatoma development in a rotenone-induced PD model, and as such may help with our further understanding of the correlation between PD and cancer.


Subject(s)
Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Neurotoxicity Syndromes/pathology , Parkinson Disease, Secondary/pathology , Receptor, Metabotropic Glutamate 5/drug effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Gene Knockdown Techniques , Male , Mice , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/psychology , Pyridines/toxicity , Rats , Rats, Sprague-Dawley , Reactive Nitrogen Species/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics , Rotenone , Signal Transduction/drug effects , Uncoupling Agents
SELECTION OF CITATIONS
SEARCH DETAIL
...