Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Neuropharmacology ; 190: 108426, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33279506

ABSTRACT

For a long time metabotropic glutamate receptors (mGluRs) were thought to regulate neuronal functions as obligatory homodimers. Recent reports, however, indicate the existence of heterodimers between group-II and -III mGluRs in the brain, which differ from the homodimers in their signal transduction and sensitivity to negative allosteric modulators (NAMs). Whether the group-I mGluRs, mGlu1 and mGlu5, form functional heterodimers in the brain is still a matter of debate. We now show that mGlu1 and mGlu5 co-purify from brain membranes and hippocampal tissue and co-localize in cultured hippocampal neurons. Complementation assays with mutants deficient in agonist-binding or G protein-coupling reveal that mGlu1/5 heterodimers are functional in heterologous cells and transfected cultured hippocampal neurons. In contrast to heterodimers between group-II and -III mGluRs, mGlu1/5 receptors exhibit a symmetric signal transduction, with both protomers activating G proteins to a similar extent. NAMs of either protomer in mGlu1/5 receptors partially inhibit signaling, showing that both protomers need to be able to reach an active conformation for full receptor activity. Complete heterodimer inhibition is observed when both protomers are locked in their inactive state by a NAM. In summary, our data show that mGlu1/5 heterodimers exhibit a symmetric signal transduction and thus intermediate signaling efficacy and kinetic properties. Our data support the existence of mGlu1/5 heterodimers in neurons and highlight differences in the signaling transduction of heterodimeric mGluRs that influence allosteric modulation.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Animals , Brain/metabolism , Chromatography, Liquid , Hippocampus/cytology , Mice , Mice, Knockout , Protein Multimerization , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/genetics , Signal Transduction , Tandem Mass Spectrometry
2.
Neuropsychopharmacology ; 43(3): 503-512, 2018 02.
Article in English | MEDLINE | ID: mdl-28816242

ABSTRACT

Preclinical data suggest that inhibition of the metabotropic glutamate receptor 5 (mGluR5) receptor might hold therapeutic benefits in Fragile X syndrome (FXS). Treatment of Fmr1 knockout mice with mGluR5-negative allosteric modulators (NAMs) has been reported to correct a broad range of phenotypes related to FXS. The early short-term clinical trials with mGluR5 NAMs, including basimglurant, assessing the effects in individuals with FXS, were supportive of further exploration in larger, well-controlled trials. We evaluated basimglurant, a potent and selective mGluR5 NAM, in a 12-week, double-blind, parallel-group study of 183 adults and adolescents (aged 14-50, mean 23.4 years) with FXS. Individuals with an FMR1 full mutation were randomized to placebo or one of two doses of basimglurant. The primary efficacy endpoint was the change from baseline in behavioral symptoms using the Anxiety Depression and Mood Scale (ADAMS) total score. All treatment arms showed marked behavioral improvements from baseline to week 12 with less improvement in the basimglurant 1.5 mg arm than placebo; however, basimglurant 0.5 mg was inferior to placebo in the ADAMs total score. Treatment with basimglurant was overall well-tolerated. A higher incidence of adverse events classified as psychiatric disorders were reported in patients treated with basimglurant, including three patients with hallucinations or psychosis. In this phase 2 clinical trial, basimglurant did not demonstrate improvement over placebo. Evaluation of the overall risk-benefit in younger patient populations is an important consideration for the design of potential further investigations of efficacy with this class of medications.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/drug therapy , Imidazoles/therapeutic use , Psychotropic Drugs/therapeutic use , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Adolescent , Adult , DNA Methylation , Double-Blind Method , Excitatory Amino Acid Antagonists/adverse effects , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/psychology , Humans , Imidazoles/adverse effects , Male , Middle Aged , Psychotropic Drugs/adverse effects , Pyridines/adverse effects , RNA, Messenger/blood , Receptor, Metabotropic Glutamate 5/metabolism , Treatment Failure , Young Adult
3.
Neuropsychopharmacology ; 43(6): 1457-1465, 2018 05.
Article in English | MEDLINE | ID: mdl-29206810

ABSTRACT

Drugs targeting metabotropic glutamate receptor 5 (mGluR5) have therapeutic potential in autism spectrum disorders (ASD), including tuberous sclerosis complex (TSC). The question whether inhibition or potentiation of mGluR5 could be beneficial depends, among other factors, on the specific indication. To facilitate the development of mGluR5 treatment strategies, we tested the therapeutic utility of mGluR5 negative and positive allosteric modulators (an mGluR5 NAM and PAM) for TSC, using a mutant mouse model with neuronal loss of Tsc2 that demonstrates disease-related phenotypes, including behavioral symptoms of ASD and epilepsy. This model uniquely enables the in vivo characterization and rescue of the electrographic seizures associated with TSC. We demonstrate that inhibition of mGluR5 corrects hyperactivity, seizures, and elevated de novo synaptic protein synthesis. Conversely, positive allosteric modulation of mGluR5 results in the exacerbation of hyperactivity and epileptic phenotypes. The data suggest a meaningful therapeutic potential for mGluR5 NAMs in TSC, which warrants clinical exploration and the continued development of mGluR5 therapies.


Subject(s)
Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Tuberous Sclerosis/drug therapy , Allosteric Regulation , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Brain/drug effects , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Epilepsy/drug therapy , Epilepsy/metabolism , Excitatory Amino Acid Agents/pharmacology , Female , Imidazoles/pharmacology , Male , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Neurons/metabolism , Phenotype , Pyridines/pharmacology , Rats, Long-Evans , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/genetics
4.
Nat Rev Drug Discov ; 17(4): 280-299, 2018 04.
Article in English | MEDLINE | ID: mdl-29217836

ABSTRACT

Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.


Subject(s)
Fragile X Syndrome/drug therapy , Neurodevelopmental Disorders/drug therapy , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/therapeutic use , Animals , Clinical Trials as Topic , Drug Development/methods , Drug Evaluation, Preclinical , Humans , Randomized Controlled Trials as Topic
5.
Br J Clin Pharmacol ; 84(3): 445-455, 2018 03.
Article in English | MEDLINE | ID: mdl-29096426

ABSTRACT

AIM: The objectives of this first-in-human study were to evaluate the safety and tolerability, pharmacokinetics and pharmacodynamics, and maximum tolerated dose (MTD) of single ascending oral doses of RG7342, a positive allosteric modulator (PAM) of the metabotropic glutamate receptor 5 (mGlu5) for the treatment of schizophrenia, in healthy male subjects. METHODS: This was a single-centre, randomized, double-blind, adaptive study of 37 subjects receiving single ascending oral doses of RG7342 (ranging from 0.06-1.2 mg, n = 27) or placebo (n = 10). A modified continual reassessment method, with control for the probability of overdosing based on the occurrence of dose-limiting events (DLEs), was applied to inform the subsequent dose decisions for RG7342. RESULTS: DLEs consisted of dizziness, nausea and vomiting, and the incidence and severity of these adverse events increased in a concentration-dependent manner. RG7342 doses of 1.2 mg under fasting conditions, which reached a mean maximum plasma concentration (Cmax ) of 10.2 ng ml-1 , were not tolerated (four out of six subjects experienced DLEs). RG7342 showed dose-proportional pharmacokinetics, with rapid absorption and a biphasic decline, and a mean terminal half-life estimated to be >1000 h. CONCLUSIONS: Single oral doses of RG7342 were generally tolerated up to 0.6 mg under fasting and 0.9 mg under fed conditions in healthy subjects. Bayesian adaptive methods describing the probability of DLEs were applied effectively to support dose escalation. MTDs (fasting, fed) were associated with a Cmax of 6.5 ng ml-1 . The development of RG7342 was discontinued owing to the potential challenges associated with a long half-life in context of the observed adverse events.


Subject(s)
Allosteric Regulation/drug effects , Food-Drug Interactions , Receptor, Metabotropic Glutamate 5/drug effects , Administration, Oral , Adolescent , Adult , Bayes Theorem , Dose-Response Relationship, Drug , Double-Blind Method , Fasting , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Young Adult
6.
Brain Behav Immun ; 59: 79-92, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27524668

ABSTRACT

Etiology and pharmacotherapy of stress-related psychiatric conditions and somatoform disorders are areas of high unmet medical need. Stressors holding chronic plus psychosocial components thereby bear the highest health risk. Although the metabotropic glutamate receptor subtype 5 (mGlu5) is well studied in the context of acute stress-induced behaviors and physiology, virtually nothing is known about its potential involvement in chronic psychosocial stress. Using the mGlu5 negative allosteric modulator CTEP (2-chloro-4-[2-[2,5-dimethyl-1-[4-(trifluoromethoxy)phenyl]imidazol-4yl]ethynyl]pyridine), a close analogue of the clinically active drug basimglurant - but optimized for rodent studies, as well as mGlu5-deficient mice in combination with a mouse model of male subordination (termed CSC, chronic subordinate colony housing), we demonstrate that mGlu5 mediates multiple physiological, immunological, and behavioral consequences of chronic psychosocial stressor exposure. For instance, CTEP dose-dependently relieved hypothalamo-pituitary-adrenal axis dysfunctions, colonic inflammation as well as the CSC-induced increase in innate anxiety; genetic ablation of mGlu5 in mice largely reproduced the stress-protective effects of CTEP and additionally ameliorated CSC-induced physiological anxiety. Interestingly, CSC also induced an upregulation of mGlu5 in the hippocampus, a stress-regulating brain area. Taken together, our findings provide evidence that mGlu5 is an important mediator for a wide range of chronic psychosocial stress-induced alterations and a potentially valuable drug target for the treatment of chronic stress-related pathologies in man.


Subject(s)
Imidazoles/therapeutic use , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Stress, Psychological/psychology , Adrenocorticotropic Hormone/blood , Animals , Anxiety/etiology , Anxiety/psychology , Chronic Disease , Dominance-Subordination , Dose-Response Relationship, Drug , Fever/etiology , Fever/physiopathology , Hydrocortisone/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Metabotropic Glutamate 5/genetics , Social Environment , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 112(41): 12852-7, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417096

ABSTRACT

A feature of early postnatal neocortical development is a transient peak in signaling via metabotropic glutamate receptor 5 (mGluR5). In visual cortex, this change coincides with increased sensitivity of excitatory synapses to monocular deprivation (MD). However, loss of visual responsiveness after MD occurs via mechanisms revealed by the study of long-term depression (LTD) of synaptic transmission, which in layer 4 is induced by acute activation of NMDA receptors (NMDARs) rather than mGluR5. Here we report that chronic postnatal down-regulation of mGluR5 signaling produces coordinated impairments in both NMDAR-dependent LTD in vitro and ocular dominance plasticity in vivo. The data suggest that ongoing mGluR5 signaling during a critical period of postnatal development establishes the biochemical conditions that are permissive for activity-dependent sculpting of excitatory synapses via the mechanism of NMDAR-dependent LTD.


Subject(s)
Dominance, Ocular/physiology , Long-Term Synaptic Depression/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Visual Cortex/metabolism , Animals , Mice , Mice, Mutant Strains , Receptor, Metabotropic Glutamate 5/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/genetics , Synaptic Transmission/physiology
8.
J Pharmacol Exp Ther ; 353(1): 213-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25665805

ABSTRACT

Major depressive disorder (MDD) is a serious public health burden and a leading cause of disability. Its pharmacotherapy is currently limited to modulators of monoamine neurotransmitters and second-generation antipsychotics. Recently, glutamatergic approaches for the treatment of MDD have increasingly received attention, and preclinical research suggests that metabotropic glutamate receptor 5 (mGlu5) inhibitors have antidepressant-like properties. Basimglurant (2-chloro-4-[1-(4-fluoro-phenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]-pyridine) is a novel mGlu5 negative allosteric modulator currently in phase 2 clinical development for MDD and fragile X syndrome. Here, the comprehensive preclinical pharmacological profile of basimglurant is presented with a focus on its therapeutic potential for MDD and drug-like properties. Basimglurant is a potent, selective, and safe mGlu5 inhibitor with good oral bioavailability and long half-life supportive of once-daily administration, good brain penetration, and high in vivo potency. It has antidepressant properties that are corroborated by its functional magnetic imaging profile as well as anxiolytic-like and antinociceptive features. In electroencephalography recordings, basimglurant shows wake-promoting effects followed by increased delta power during subsequent non-rapid eye movement sleep. In microdialysis studies, basimglurant had no effect on monoamine transmitter levels in the frontal cortex or nucleus accumbens except for a moderate increase of accumbal dopamine, which is in line with its lack of pharmacological activity on monoamine reuptake transporters. These data taken together, basimglurant has favorable drug-like properties, a differentiated molecular mechanism of action, and antidepressant-like features that suggest the possibility of also addressing important comorbidities of MDD including anxiety and pain as well as daytime sleepiness and apathy or lethargy.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Imidazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation , Animals , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/therapeutic use , Biogenic Monoamines/metabolism , Brain/metabolism , Cells, Cultured , Cricetulus , Depression/metabolism , Depression/psychology , Drug Inverse Agonism , Electroencephalography , Female , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Macaca fascicularis , Male , Mice , Pain/drug therapy , Pain/physiopathology , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Radioligand Assay , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Metabotropic Glutamate 5/metabolism , Urinary Bladder, Overactive/drug therapy , Urinary Bladder, Overactive/physiopathology
9.
Nat Neurosci ; 18(2): 182-4, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25581360

ABSTRACT

Human chromosome 16p11.2 microdeletion is the most common gene copy number variation in autism, but the synaptic pathophysiology caused by this mutation is largely unknown. Using a mouse with the same genetic deficiency, we found that metabotropic glutamate receptor 5 (mGluR5)-dependent synaptic plasticity and protein synthesis was altered in the hippocampus and that hippocampus-dependent memory was impaired. Notably, chronic treatment with a negative allosteric modulator of mGluR5 reversed the cognitive deficit.


Subject(s)
Autistic Disorder/physiopathology , Chromosome Disorders/physiopathology , Hippocampus/physiopathology , Imidazoles/pharmacology , Intellectual Disability/physiopathology , Memory Disorders/physiopathology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/physiology , Animals , Autistic Disorder/genetics , Autistic Disorder/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Chromosome Deletion , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 16/metabolism , Chromosomes, Mammalian , Disease Models, Animal , Hippocampus/metabolism , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
10.
J Med Chem ; 58(3): 1358-71, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25565255

ABSTRACT

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor 5 (mGlu5) have potential for the treatment of psychiatric diseases including depression, fragile X syndrome (FXS), anxiety, obsessive-compulsive disorders, and levodopa induced dyskinesia in Parkinson's disease. Herein we report the optimization of a weakly active screening hit 1 to the potent and selective compounds chloro-4-[1-(4-fluorophenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]pyridine (basimglurant, 2) and 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine (CTEP, 3). Compound 2 is active in a broad range of anxiety tests reaching the same efficacy but at a 10- to 100-fold lower dose compared to diazepam and is characterized by favorable DMPK properties in rat and monkey as well as an excellent preclinical safety profile and is currently in phase II clinical studies for the treatment of depression and fragile X syndrome. Analogue 3 is the first reported mGlu5 NAM with a long half-life in rodents and is therefore an ideal tool compound for chronic studies in mice and rats.


Subject(s)
Depression/drug therapy , Drug Discovery , Fragile X Syndrome/drug therapy , Imidazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Macaca mulatta , Male , Mice , Mice, Inbred Strains , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship
11.
Neuropsychopharmacology ; 40(5): 1222-33, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25409593

ABSTRACT

Stress-induced psychiatric disorders, such as depression, have recently been linked to changes in glutamate transmission in the central nervous system. Glutamate signaling is mediated by a range of receptors, including metabotropic glutamate receptors (mGluRs). In particular, mGluR subtype 5 (mGluR5) is highly implicated in stress-induced psychopathology. The major scaffold protein Homer1 critically interacts with mGluR5 and has also been linked to several psychopathologies. Yet, the specific role of Homer1 in this context remains poorly understood. We used chronic social defeat stress as an established animal model of depression and investigated changes in transcription of Homer1a and Homer1b/c isoforms and functional coupling of Homer1 to mGluR5. Next, we investigated the consequences of Homer1 deletion, overexpression of Homer1a, and chronic administration of the mGluR5 inverse agonist CTEP (2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine) on the effects of chronic stress. In mice exposed to chronic stress, Homer1b/c, but not Homer1a, mRNA was upregulated and, accordingly, Homer1/mGluR5 coupling was disrupted. We found a marked hyperactivity behavior as well as a dysregulated hypothalamic-pituitary-adrenal axis activity in chronically stressed Homer1 knockout (KO) mice. Chronic administration of the selective and orally bioavailable mGluR5 inverse agonist, CTEP, was able to recover behavioral alterations induced by chronic stress, whereas overexpression of Homer1a in the hippocampus led to an increased vulnerability to chronic stress, reflected in an increased physiological response to stress as well as enhanced depression-like behavior. Overall, our results implicate the glutamatergic system in the emergence of stress-induced psychiatric disorders, and support the Homer1/mGluR5 complex as a target for the development of novel antidepressant agents.


Subject(s)
Carrier Proteins/metabolism , Depressive Disorder/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Resilience, Psychological , Stress, Psychological/metabolism , Animals , Brain/drug effects , Brain/metabolism , Carrier Proteins/genetics , Chronic Disease , Disease Models, Animal , Dominance-Subordination , Drug Inverse Agonism , Excitatory Amino Acid Antagonists/pharmacology , Homer Scaffolding Proteins , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms , Pyridines/pharmacology , RNA, Messenger/metabolism , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Resilience, Psychological/drug effects
12.
Curr Opin Pharmacol ; 20: 124-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25488569

ABSTRACT

Fragile X syndrome (FXS) is the most common monogenic form of inherited mental retardation caused by a trinucleotid repeat expansion and transcriptional shutdown of the FMR1 gene. FXS patients present a complex and often severe neuropsychiatric phenotype yet have mild somatic symptoms, normal life expectancies, and no indications of neurodegeneration. The therapeutic potential of mGlu5 inhibitors was proposed in the 'mGluR theory of FXS' based on early insights into the molecular pathophysiology of FXS. Studies in Fragile X mental retardation 1 (Fmr1) knock-out mice, a widely used disease model, demonstrated that mGlu5 inhibitors can correct a broad range of disease-related phenotypes. Recent clinical trials, however, with two different mGlu5 inhibitors (basimglurant and mavoglurant) showed no therapeutic benefit in FXS patients for reasons as yet unclear.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Fragile X Syndrome/drug therapy , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Animals , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/physiopathology , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Mice , Mice, Knockout , Molecular Targeted Therapy , Phenotype , Pyridines/pharmacology , Pyridines/therapeutic use
13.
Biol Psychiatry ; 75(3): 189-97, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23910948

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is the most common genetic cause for intellectual disability. Fmr1 knockout (KO) mice are an established model of FXS. Chronic pharmacological inhibition of metabotropic glutamate receptor 5 (mGlu5) in these mice corrects multiple molecular, physiological, and behavioral phenotypes related to patients' symptoms. To better understand the pathophysiology of FXS and the effect of treatment, brain activity was analyzed using functional magnetic resonance imaging in relation to learning and memory performance. METHODS: Wild-type (WT) and Fmr1 KO animals receiving chronic treatment with the mGlu5 inhibitor CTEP or vehicle were evaluated consecutively for 1) learning and memory performance in the inhibitory avoidance and extinction test, and 2) for the levels of brain activity using continuous arterial spin labeling based functional magnetic resonance imaging. Neural activity patterns were correlated with cognitive performance using a multivariate regression analysis. Furthermore, mGlu5 receptor expression in brains of untreated mice was analyzed by autoradiography and saturation analysis using [(3)H]-ABP688. RESULTS: Chronic CTEP treatment corrected the learning deficit observed in Fmr1 KO mice in the inhibitory avoidance and extinction test and prevented memory extinction in WT and Fmr1 KO animals. Chronic CTEP treatment normalized perfusion in the amygdala and the lateral hypothalamus in Fmr1 KO mice and furthermore decreased perfusion in the hippocampus and increased perfusion in primary sensorimotor cortical areas. No significant differences in mGlu5 receptor expression levels between Fmr1 WT and KO mice were detected. CONCLUSIONS: Chronic mGlu5 inhibition corrected the learning deficits and partially normalized the altered brain activity pattern in Fmr1 KO mice.


Subject(s)
Brain/drug effects , Cognition/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/drug therapy , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Animals , Avoidance Learning/drug effects , Brain/blood supply , Disease Models, Animal , Electroshock/adverse effects , Excitatory Amino Acid Antagonists/pharmacokinetics , Extinction, Psychological/drug effects , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Imidazoles/therapeutic use , Mice , Mice, Knockout , Oximes/pharmacokinetics , Oxygen/blood , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5/metabolism , Tritium/pharmacokinetics
14.
Nat Rev Drug Discov ; 12(10): 777-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24080699

ABSTRACT

The rising rates of autism spectrum disorder (ASD) and the lack of effective medications to treat its core symptoms have led to an increased sense of urgency to identify therapies for this group of neurodevelopmental conditions. Developing drugs for ASD, however, has been challenging because of a limited understanding of its pathophysiology, difficulties in modelling the disease in vitro and in vivo, the heterogeneity of symptoms, and the dearth of prior experience in clinical development. In the past few years these challenges have been mitigated by considerable advances in our understanding of forms of ASD caused by single-gene alterations, such as fragile X syndrome and tuberous sclerosis. In these cases we have gained insights into the pathophysiological mechanisms underlying these conditions. In addition, they have aided in the development of animal models and compounds with the potential for disease modification in clinical development. Moreover, genetic studies are illuminating the molecular pathophysiology of ASD, and new tools such as induced pluripotent stem cells offer novel possibilities for drug screening and disease diagnostics. Finally, large-scale collaborations between academia and industry are starting to address some of the key barriers to developing drugs for ASD. Here, we propose a conceptual framework for drug discovery in ASD encompassing target identification, drug profiling and considerations for clinical trials in this novel area.


Subject(s)
Child Development Disorders, Pervasive/drug therapy , Drug Design , Drug Discovery , Animals , Child , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/physiopathology , Clinical Trials as Topic , Cooperative Behavior , Disease Models, Animal , Drug Industry/methods , Drug Industry/organization & administration , Humans , Molecular Targeted Therapy
15.
PLoS One ; 8(7): e68722, 2013.
Article in English | MEDLINE | ID: mdl-23844236

ABSTRACT

Brain-derived neurotrophic factor (BDNF) importantly regulates learning and memory and supports the survival of injured neurons. Reduced BDNF levels have been detected in the brains of Alzheimer's disease (AD) patients but the exact role of BDNF in the pathophysiology of the disorder remains obscure. We have recently shown that reduced signaling of BDNF receptor TrkB aggravates memory impairment in APPswe/PS1dE9 (APdE9) mice, a model of AD. The present study examined the influence of Bdnf gene deficiency (heterozygous knockout) on spatial learning, spontaneous exploratory activity and motor coordination/balance in middle-aged male and female APdE9 mice. We also studied brain BDNF protein levels in APdE9 mice in different ages showing progressive amyloid pathology. Both APdE9 and Bdnf mutations impaired spatial learning in males and showed a similar trend in females. Importantly, the effect was additive, so that double mutant mice performed the worst. However, APdE9 and Bdnf mutations influenced spontaneous locomotion in contrasting ways, such that locomotor hyperactivity observed in APdE9 mice was normalized by Bdnf deficiency. Obesity associated with Bdnf deficiency did not account for the reduced hyperactivity in double mutant mice. Bdnf deficiency did not alter amyloid plaque formation in APdE9 mice. Before plaque formation (3 months), BDNF protein levels where either reduced (female) or unaltered (male) in the APdE9 mouse cortex. Unexpectedly, this was followed by an age-dependent increase in mature BDNF protein. Bdnf mRNA and phospho-TrkB levels remained unaltered in the cortical tissue samples of middle-aged APdE9 mice. Immunohistological studies revealed increased BDNF immunoreactivity around amyloid plaques indicating that the plaques may sequester BDNF protein and prevent it from activating TrkB. If similar BDNF accumulation happens in human AD brains, it would suggest that functional BDNF levels in the AD brains are even lower than reported, which could partially contribute to learning and memory problems of AD patients.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain-Derived Neurotrophic Factor/genetics , Memory Disorders/etiology , Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Animals , Body Weight/genetics , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Environment , Female , Humans , Hyperkinesis/genetics , Male , Maze Learning , Memory , Mice , Mice, Transgenic , Plaque, Amyloid , Presenilin-1/genetics , Psychomotor Performance
16.
Hum Mol Genet ; 22(10): 2041-54, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23396537

ABSTRACT

Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the loss-of-function of fragile X mental retardation protein (FMRP). The loss of FMRP function in neurons abolishes its suppression on mGluR1/5-dependent dendritic protein translation, enhancing mGluR1/5-dependent synaptic plasticity and other disease phenotypes in FXS. In this study, we describe a new activation function of FMRP in regulating protein expression in astroglial cells. We found that astroglial glutamate transporter subtype glutamate transporter 1 (GLT1) and glutamate uptake is significantly reduced in the cortex of fmr1(-/-) mice. Correspondingly, neuronal excitability is also enhanced in acute fmr1(-/-) (but not in fmr1(+/+) control) cortical slices treated with low doses (10 µm) of the GLT1-specific inhibitor dihydrokainate (DHK). Using mismatched astrocyte and neuron co-cultures, we demonstrate that the loss of astroglial (but not neuronal) FMRP particularly reduces neuron-dependent GLT1 expression and glutamate uptake in co-cultures. Interestingly, protein (but not mRNA) expression and the (S)-3,5-dihydroxyphenylglycine-dependent Ca(2+) responses of astroglial mGluR5 receptor are also selectively reduced in fmr1(-/-) astrocytes and brain slices, attenuating neuron-dependent GLT1 expression. Subsequent FMRP immunoprecipitation and QRT-PCR analysis showed that astroglial mGluR5 (but not GLT1) mRNA is associated with FMRP. In summary, our results provide evidence that FMRP positively regulates translational expression of mGluR5 in astroglial cells, and FMRP-dependent down-regulation of mGluR5 underlies GLT1 dysregulation in fmr1(-/-) astrocytes. The dysregulation of GLT1 and reduced glutamate uptake may potentially contribute to enhanced neuronal excitability observed in the mouse model of FXS.


Subject(s)
Astrocytes/metabolism , Down-Regulation , Excitatory Amino Acid Transporter 2/biosynthesis , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Protein Biosynthesis , Receptors, Metabotropic Glutamate/metabolism , Animals , Astrocytes/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Excitatory Amino Acid Transporter 2/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Gene Knockdown Techniques , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Mice , Neurons/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics
17.
Trends Pharmacol Sci ; 33(12): 669-84, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23084458

ABSTRACT

Autism and autism spectrum disorders (ASDs) affect millions of individuals worldwide. Despite increased autism diagnoses over the past 30 years, therapeutic intervention is often 'trial and error'. This approach has identified some beneficial agents, but complex heterogeneous disorders require a more personalized treatment regimen. Many ASD risk factors are genetic, implicating impaired synaptic development and function. Monogenetic disorders (e.g., fragile X syndrome, Rett syndrome, and neurofibromatosis) that have phenotypic overlap with autism provide insights into ASD pathology through the identification novel drug targets (e.g., glutamatergic receptors). Encouragingly, some of these novel drug targets provide symptomatic improvement, even in patients who have lived with ASDs for protracted periods of time. Consequently, a targeted drug discovery approach is expected to deliver improved agents for the treatment and management of ASDs. Here, we review the opportunities and challenges in drug development for autism and provide insight into the neurobiology of ASDs.


Subject(s)
Autistic Disorder/drug therapy , Autistic Disorder/physiopathology , Child Development Disorders, Pervasive/drug therapy , Child Development Disorders, Pervasive/physiopathology , Synapses/physiology , Animals , Child , Drug Discovery , Humans , Psychotropic Drugs/therapeutic use
18.
Behav Brain Funct ; 8: 30, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-22686184

ABSTRACT

BACKGROUND: Previous studies have demonstrated a profound lack of habituation in 129P3 mice compared to the habituating, but initially more anxious, BALB/c mice. The present study investigated whether this non-adaptive phenotype of 129P3 mice is primarily based on anxiety-related characteristics. METHODS: To test this hypothesis and extend our knowledge on the behavioural profile of 129P3 mice, the effects of the anxiolyticdiazepam (1, 3 and 5 mg/kg) and the putative anxiolytic metabotropic glutamate receptor 5 (mGlu5R) antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP, 3, 10 and 30 mg/kg) treatment on within-trial (intrasession) habituation, object recognition (diazepam: 1 mg/kg; MPEP 10 mg/kg) and on the central-nervous expression of the immediate early gene c-Fos (diazepam: 1 mg/kg; MPEP 10 mg/kg) were investigated. RESULTS: Behavioural findings validated the initially high, but habituating phenotype of BALB/c mice, while 129P3 mice were characterized by impaired intrasession habituation. Diazepam had an anxiolytic effect in BALB/c mice, while in higher doses caused behavioural inactivity in 129P3 mice. MPEP revealed almost no anxiolytic effects on behaviour in both strains, but reduced stress-induced corticosterone responses only in 129P3 mice. These results were complemented by reduced expression of c-Fos after MPEP treatment in brain areas related to emotional processes, and increased c-Fos expression in higher integrating brain areas such as the prelimbic cortex compared to vehicle-treated 129P3 mice. CONCLUSIONS: These results suggest that the strain differences observed in (non)adaptive anxiety behaviour are at least in part mediated by differences in gamma-aminobutyric acid- A and mGluR5 mediated transmission.


Subject(s)
Avoidance Learning/drug effects , Avoidance Learning/physiology , Diazepam/pharmacology , Habituation, Psychophysiologic/drug effects , Habituation, Psychophysiologic/genetics , Pyridines/pharmacology , Animals , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred Strains , Random Allocation
19.
Neuron ; 74(1): 49-56, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22500629

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. Previous studies have implicated mGlu5 in the pathogenesis of the disease, but a crucial unanswered question is whether pharmacological mGlu5 inhibition is able to reverse an already established FXS phenotype in mammals. Here we have used the novel, potent, and selective mGlu5 inhibitor CTEP to address this issue in the Fmr1 knockout mouse. Acute CTEP treatment corrects elevated hippocampal long-term depression, protein synthesis, and audiogenic seizures. Chronic treatment that inhibits mGlu5 within a receptor occupancy range of 81% ± 4% rescues cognitive deficits, auditory hypersensitivity, aberrant dendritic spine density, overactive ERK and mTOR signaling, and partially corrects macroorchidism. This study shows that a comprehensive phenotype correction in FXS is possible with pharmacological intervention starting in young adulthood, after development of the phenotype. It is of great interest how these findings may translate into ongoing clinical research testing mGlu5 inhibitors in FXS patients.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/drug therapy , Imidazoles/therapeutic use , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/drug effects , Age Factors , Animals , Disease Models, Animal , Drug Administration Schedule , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Phenotype , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism
20.
Neuropharmacology ; 62(1): 391-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21867718

ABSTRACT

Accumulating evidence suggests that biogenic amine-based antidepressants act, at least in part, via regulation of brain-derived neurotrophic factor (BDNF) signaling. Biogenic amine-based antidepressants increase BDNF synthesis and activate its signaling pathway through TrkB receptors. Moreover, the antidepressant-like effects of these molecules are abolished in BDNF deficient mice. Glutamate-based drugs, including the NMDA antagonist ketamine, and the AMPA receptor potentiator LY 451646, mimic the effects of antidepressants in preclinical tests with high predictive validity. In humans, a single intravenous dose of ketamine produces an antidepressant effect that is rapid, robust and persistent. In this study, we examined the role of BDNF in expression of the antidepressant-like effects of ketamine and an AMPA receptor potentiator (LY 451646) in the forced swim test (FST). Ketamine and LY 451646 produced antidepressant-like effects in the FST in mice at 45 min after a single injection, but no effects were observed one week after a single ketamine injection. As previously reported, the effects of imipramine in the forced swim test were blunted in heterozygous BDNF knockout (bdnf(+/-)) mice. However ketamine and LY 451646 produced similar antidepressant-like responses in wildtype and bdnf(+/-) mice. Neither ketamine nor LY 451646 significantly influenced the levels BDNF or TrkB phosphorylation in the hippocampus when assessed at 45 min or 7 days after the drug administration. These data demonstrate that under the conditions tested, neither ketamine nor the AMPA-potentiator LY 451656 activate BDNF signaling, but produce a characteristic antidepressant-like response in heterozygous bdnf(+/-) mice. These data indicate that unlike biogenic amine-based agents, BDNF signaling does not play a pivotal role in the antidepressant effects of glutamate-based compounds. This article is part of a Special Issue entitled 'Anxiety and Depression'.


Subject(s)
Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/deficiency , Depression/drug therapy , Ketamine/pharmacology , Sulfonamides/pharmacology , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/metabolism , Depression/genetics , Depression/pathology , Depression/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Imipramine/pharmacology , Imipramine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reaction Time/drug effects , Reaction Time/genetics , Receptor, trkA/metabolism , Swimming/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...