Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
J Med Chem ; 67(14): 11662-11687, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38691510

ABSTRACT

Metabotropic glutamate (mGlu) receptors play a key role in modulating most synapses in the brain. The mGlu7 receptors inhibit presynaptic neurotransmitter release and offer therapeutic possibilities for post-traumatic stress disorders or epilepsy. Screening campaigns provided mGlu7-specific allosteric modulators as the inhibitor XAP044 (Gee et al. J. Biol. Chem. 2014). In contrast to other mGlu receptor allosteric modulators, XAP044 does not bind in the transmembrane domain but to the extracellular domain of the mGlu7 receptor and not at the orthosteric site. Here, we identified the mode of action of XAP044, combining synthesis of derivatives, modeling and docking experiments, and mutagenesis. We propose a unique mode of action of these inhibitors, preventing the closure of the Venus flytrap agonist binding domain. While acting as a noncompetitive antagonist of L-AP4, XAP044 and derivatives act as apparent competitive antagonists of LSP4-2022. These data revealed more potent XAP044 analogues and new possibilities to target mGluRs.


Subject(s)
Receptors, Metabotropic Glutamate , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/chemistry , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Animals , Protein Domains , Allosteric Regulation/drug effects , HEK293 Cells , Binding Sites
2.
Cells ; 11(11)2022 06 02.
Article in English | MEDLINE | ID: mdl-35681512

ABSTRACT

Chronic psychosocial stress participates prominently in the etiology of various psychiatric conditions and comorbid somatic pathologies; however, suitable pharmacotherapy of these disorders is still of high medical need. During the last few decades, research on mGlu receptors advanced remarkably and much attention was given to the mGlu7 subtype. Here, genetic mGlu7 ablation, short-term pharmacological mGlu7 blockade, as well as siRNA-mediated knockdown of mGlu7 were shown to result in an acute anti-stress, antidepressant- and anxiolytic-like phenotype in mice. Moreover, we recently revealed a prominent stress-protective effect of genetic mGlu7 ablation also with respect to chronic psychosocial stress. In addition, we are able to demonstrate in the present study that the chronic pharmacological blockade of mGlu7 interferes with various chronic stress-induced alterations. For this, we used the chronic subordinate colony housing (CSC), a mouse model of chronic male subordination, in combination with chronic treatment with the mGlu7-selective orthosteric-like antagonist XAP044 (7-hydroxy-3-(4-iodophenoxy)-4H-chromen-4-one). Interestingly, XAP044 dose-dependently ameliorates hypothalamic-pituitary-adrenal axis dysfunctions, thymus atrophy, as well as the CSC-induced increase in innate anxiety. Taken together, our findings provide further evidence for the role of mGlu7 in chronic psychosocial stress-induced alterations and suggests the pharmacological blockade of mGlu7 as a promising therapeutic approach for the treatment of chronic stress-related pathologies in men.


Subject(s)
Droseraceae , Receptors, Metabotropic Glutamate , Animals , Droseraceae/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , Phenotype , Pituitary-Adrenal System/metabolism , Receptors, Metabotropic Glutamate/metabolism
3.
Genes Brain Behav ; 19(1): e12627, 2020 01.
Article in English | MEDLINE | ID: mdl-31793148

ABSTRACT

The group III metabotropic glutamate receptor subtype 7 (mGlu7) is an important regulator of glutamatergic and GABAergic neurotransmission and known to mediate emotionality and male social behavior. However, a possible regulatory role in maternal behavior remains unknown to date. Adequate expression of maternal behavior is essential for successful rearing and healthy development of the young. By understanding genetic and neural mechanisms underlying this important prosocial behavior, we gain valuable insights into possible dysregulations. Using genetic ablation as well as pharmacological modulation, we studied various parameters of maternal behavior in two different mouse strains under the influence of mGlu7. We can clearly show a regulatory role of mGlu7 in maternal behavior. Naïve virgin female C57BL/6 mGlu7 knockout mice showed more often nursing postures and less spontaneous maternal aggression compared to their heterozygous and wildtype littermates. In lactating C57BL/6 wildtype mice, acute central activation of mGlu7 by the selective agonist AMN082 reduced arched back nursing and accelerated pup retrieval without affecting maternal aggression. In addition, in lactating CD1 wildtype mice the selective mGlu7 antagonist XAP044 increased both pup retrieval and maternal aggression. With respect to receptor expression levels, mGlu7 mRNA expression was higher in lactating vs virgin C57BL/6 mice in the prefrontal cortex, but not hypothalamus or hippocampus. In conclusion, these findings highlight a significant role of the mGlu7 receptor subtype in mediating maternal behavior in mice. Region-dependent studies are warranted to further extend our knowledge on the specific function of the brain glutamate system in maternal behavior.


Subject(s)
Aggression , Maternal Behavior , Motivation , Receptors, Metabotropic Glutamate/genetics , Animals , Benzhydryl Compounds/pharmacology , Chromones/pharmacology , Excitatory Amino Acid Agents/pharmacology , Female , Mice , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism
4.
Behav Brain Res ; 328: 57-61, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28392321

ABSTRACT

Pharmacological modulation of metabotropic glutamate receptor subtype 5 (mGluR5) and 7 (mGluR7) was shown to attenuate the acquisition and to facilitate the extinction of cued and contextual, non-social, fear. Using the allosteric mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) and the allosteric mGluR7 agonist N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082), we aimed to study how pharmacological blockade of mGluR5 and activation of mGluR7 influence acquisition and extinction of social fear in mice. We could show that when administered before social fear conditioning, neither MPEP nor AMN082 affected acquisition and extinction of social fear, suggesting that mGluR5 inactivation and mGluR7 activation do not alter social fear. However, when administered before social fear extinction, both MPEP and AMN082 impaired social fear extinction and extinction recall. These findings suggest that mGluR5 inactivation and mGluR7 activation are unlikely to prevent the formation of traumatic social memories. Furthermore, medication strategies aimed at augmenting exposure-based therapies for psychiatric disorders associated with social deficits via modulation of mGluR5 and mGluR7 must be pursued cautiously because of their potential to delay social fear extinction processes.


Subject(s)
Excitatory Amino Acid Agents/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Psychotropic Drugs/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptors, Metabotropic Glutamate/agonists , Animals , Benzhydryl Compounds/pharmacology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Fear/physiology , Male , Mental Recall/drug effects , Mental Recall/physiology , Mice , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Social Perception
5.
Brain Behav Immun ; 64: 23-32, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28012830

ABSTRACT

Chronic subordinate colony housing (CSC), an established mouse model for chronic psychosocial stress, promotes a microbial signature of gut inflammation, characterized by expansion of Proteobacteria, specifically Helicobacter spp., in association with colitis development. However, whether the presence of Helicobacter spp. during CSC is critically required for colitis development is unknown. Notably, during previous CSC studies performed at Regensburg University (University 1), male specific-pathogen-free (SPF) CSC mice lived in continuous subordination to a physically present and Helicobacter spp.-positive resident. Therefore, it is likely that CSC mice were colonized, during the CSC procedure, with Helicobacter spp. originating from the dominant resident. In the present study we show that employing SPF CSC mice and Helicobacter spp.-free SPF residents at Ulm University (University 2), results in physiological responses that are typical of chronic psychosocial stress, including increased adrenal and decreased thymus weights, decreased adrenal in vitro adrenocorticotropic hormone (ACTH) responsiveness, and increased anxiety-related behavior. However, in contrast to previous studies that used Helicobacter spp.-positive resident mice, use of Helicobacter spp.-negative resident mice failed to induce spontaneous colitis in SPF CSC mice. Consistent with the hypothesis that the latter is due to a lack of Helicobacter spp. transmission from dominant residents to subordinate mice during the CSC procedure, colonization of SPF residents with Helicobacter typhlonius at University 2, prior to the start of the CSC model, rescued the colitis-inducing potential of CSC exposure. Furthermore, using SPF CSC mice and H. typhlonius-free SPF residents at University 1 prevented CSC-induced colitis. In summary, our data support the hypothesis that the presence or absence of exposure to certain pathobionts contributes to individual variability in susceptibility to stress-/trauma-associated pathologies and to reproducibility of stress-related outcomes between laboratories.


Subject(s)
Colitis/microbiology , Gastrointestinal Microbiome , Helicobacter/pathogenicity , Individuality , Stress, Psychological/microbiology , Adrenal Glands/pathology , Animals , Anxiety/microbiology , Colitis/complications , Helicobacter Infections , Inflammation/microbiology , Male , Mice, Inbred C57BL , Organ Size , Stress, Psychological/complications , Thymus Gland/pathology
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.
Neuropharmacology ; 113(Pt A): 343-353, 2017 02.
Article in English | MEDLINE | ID: mdl-27769854

ABSTRACT

We studied the interaction between mGlu7 and α1-adrenergic receptors in heterologous expression systems, brain slices, and living animals. L-2-Amino-4-phosphonobutanoate (L-AP4), and l-serine-O-phosphate (L-SOP), which activate group III mGlu receptors, restrained the stimulation of polyphosphoinositide (PI) hydrolysis induced by the α1-adrenergic receptor agonist, phenylephrine, in HEK 293 cells co-expressing α1-adrenergic and mGlu7 receptors. The inibitory action of L-AP4 was abrogated by (i) the mGlu7 receptor antagonist, XAP044; (ii) the C-terminal portion of type-2 G protein coupled receptor kinase; and (iii) the MAP kinase inhibitors, UO126 and PD98059. This suggests that the functional interaction between mGlu7 and α1-adrenergic receptors was mediated by the ßγ-subunits of the Gi protein and required the activation of the MAP kinase pathway. Remarkably, activation of neither mGlu2 nor mGlu4 receptors reduced α1-adrenergic receptor-mediated PI hydrolysis. In mouse cortical slices, both L-AP4 and L-SOP were able to attenuate norepinephrine- and phenylephrine-stimulated PI hydrolysis at concentrations consistent with the activation of mGlu7 receptors. L-AP4 failed to affect norepinephrine-stimulated PI hydrolysis in cortical slices from mGlu7-/- mice, but retained its inhibitory activity in slices from mGlu4-/- mice. At behavioural level, i.c.v. injection of phenylephrine produced antidepressant-like effects in the forced swim test. The action of phenylephrine was attenuated by L-SOP, which was inactive per se. Finally, both phenylephrine and L-SOP increased corticosterone levels in mice, but the increase was halved when the two drugs were administered in combination. Our data demonstrate that α1-adrenergic and mGlu7 receptors functionally interact and suggest that this interaction might be targeted in the treatment of stress-related disorders.


Subject(s)
Receptors, Adrenergic, alpha-1/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Adrenergic alpha-1 Receptor Agonists/metabolism , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/agonists , Signal Transduction/drug effects
8.
Neuropharmacology ; 115: 139-148, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27188844

ABSTRACT

Chronic stress-related psychiatric conditions and comorbid somatic pathologies are an enormous public health concern in modern society. The etiology of these disorders is complex, with stressors holding a chronic and psychosocial component representing the most acknowledged risk factor. During the last decades, research on the metabotropic glutamate receptor (mGlu) system advanced dramatically and much attention was given to the role of the metabotropic glutamate receptor subtype 7 (mGlu7) in acute stress-related behavior and physiology. However, virtually nothing is known about the potential involvement of mGlu7 in chronic psychosocial stress-related conditions. Using the chronic subordinate colony housing (CSC, 19 days) in male mice, we addressed whether central mGlu7 is altered upon chronic psychosocial stressor exposure and whether genetic ablation of mGlu7 interferes with the multitude of chronic stress-induced alterations. CSC exposure resulted in a downregulation of mGlu7 mRNA transcript levels in the prefrontal cortex, a brain region relevant for stress-related behaviors and physiology. Interestingly, mGlu7 deficiency relieved multiple chronic stress-induced alterations including the CSC-induced anxiety-prone phenotype; mGlu7 ablation also ameliorated CSC-induced physiological and immunological consequences such as hypothalamo-pituitary-adrenal (HPA) axis dysfunctions and colonic inflammation, respectively. Together, our findings provide first evidence for the involvement of mGlu7 in a wide range of behavioral and physiological alterations in response to chronic psychosocial stressor exposure. Moreover, the stress-protective phenotype of genetic mGlu7 ablation suggests mGlu7 pharmacological blockade to be a relevant option for the treatment of chronic stress-related emotional and somatic dysfunctions. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Subject(s)
Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Stress, Psychological/genetics , Stress, Psychological/metabolism , Adrenocorticotropic Hormone/blood , Animals , Anxiety/blood , Anxiety/metabolism , Anxiety/psychology , Body Weight/physiology , Corticosterone/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stress, Psychological/blood
9.
Curr Neuropharmacol ; 14(5): 514-39, 2016.
Article in English | MEDLINE | ID: mdl-27296643

ABSTRACT

Chronic stress-related psychiatric conditions such as anxiety, depression, and alcohol abuse are an enormous public health concern. The etiology of these pathologies is complex, with psychosocial stressors being among the most frequently discussed risk factors. The brain glutamatergic neurotransmitter system has often been found involved in behaviors and pathophysiologies resulting from acute stress and fear. Despite this, relatively little is known about the role of glutamatergic system components in chronic psychosocial stress, neither in rodents nor in humans. Recently, drug discovery efforts at the metabotropic receptor subtypes of the glutamatergic system (mGlu1-8 receptors) led to the identification of pharmacological tools with emerging potential in psychiatric conditions. But again, the contribution of individual mGlu subtypes to the manifestation of physiological, molecular, and behavioral consequences of chronic psychosocial stress remains still largely unaddressed. The current review will describe animal models typically used to analyze acute and particularly chronic stress conditions, including models of psychosocial stress, and there we will discuss the emerging roles for mGlu receptor subtypes. Indeed, accumulating evidence indicates relevance and potential therapeutic usefulness of mGlu2/3 ligands and mGlu5 receptor antagonists in chronic stress-related disorders. In addition, a role for further mechanisms, e.g. mGlu7-selective compounds, is beginning to emerge. These mechanisms are important to be analyzed in chronic psychosocial stress paradigms, e.g. in the chronic subordinate colony housing (CSC) model. We summarize the early results and discuss necessary future investigations, especially for mGlu5 and mGlu7 receptor blockers, which might serve to suggest improved therapeutic strategies to treat stress-related disorders.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Stress Disorders, Traumatic/physiopathology , Stress, Psychological/physiopathology , Animals , Chronic Disease , Disease Models, Animal , Receptors, Metabotropic Glutamate/genetics , Stress Disorders, Traumatic/drug therapy , Stress, Psychological/drug therapy
10.
Neuropsychopharmacology ; 41(3): 726-35, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26149357

ABSTRACT

Metabotropic glutamate receptor subtype 7 (mGluR7) is a member of group III mGluRs, which localize to the presynaptic active zones of the mammalian central nervous system. Although histological, genetic, and electrophysiological studies ensure the importance of mGluR7, its roles in behavior and physiology remain largely unknown. Using a resident-intruder paradigm, we found a severe reduction in intermale aggressive behavior in mGluR7 knockout (KO) mice. We also found alterations in other social behaviors in male mGluR7 KO mice, including sexual behavior toward male intruders. Because olfaction is critical for rodent social behavior, including aggression, we performed an olfaction test, finding that mGluR7 KO mice failed to show interest in the smell of male urine. To clarify the olfactory deficit, we then exposed mice to urine and analyzed c-Fos-immunoreactivity, discovering a remarkable reduction in neural activity in the bed nucleus of the stria terminalis (BNST) of mGluR7 KO mice. Finally, intra-BNST administration of the mGluR7-selective antagonist 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazolo[4,5-c]pyridin-4(5H)-one (MMPIP) also reproduced the phenotype of mGluR7 KO mice, including reduced aggression and altered social interaction. Thus mGluR7 may work as an 'enhancer of neural activity' in the BNST and is important for intermale aggression. Our findings demonstrate that mGluR7 is essential for social behavior and innate behavior. Our study on mGluR7 in the BNST will shed light on future therapies for emotional disorders in humans.


Subject(s)
Aggression/physiology , Receptors, Metabotropic Glutamate/metabolism , Septal Nuclei/metabolism , Aggression/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Excitatory Amino Acid Antagonists/pharmacology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Neuropsychological Tests , Olfactory Perception/drug effects , Olfactory Perception/physiology , Physical Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Pyridones/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Septal Nuclei/drug effects , Urine
11.
Bioorg Med Chem ; 22(21): 5790-803, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25316499

ABSTRACT

Here we describe the identification, structure-activity relationship and the initial pharmacological characterization of AFQ056/mavoglurant, a structurally novel, non-competitive mGlu5 receptor antagonist. AFQ056/mavoglurant was identified by chemical derivatization of a lead compound discovered in a HTS campaign. In vitro, AFQ056/mavoglurant had an IC50 of 30 nM in a functional assay with human mGluR5 and was selective over the other mGluR subtypes, iGluRs and a panel of 238 CNS relevant receptors, transporter or enzymes. In vivo, AFQ056/mavoglurant showed an improved pharmacokinetic profile in rat and efficacy in the stress-induced hyperthermia test in mice as compared to the prototypic mGluR5 antagonist MPEP. The efficacy of AFQ056/mavoglurant in humans has been assessed in L-dopa induced dyskinesia in Parkinson's disease and Fragile X syndrome in proof of principle clinical studies.


Subject(s)
Indoles/chemistry , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Animals , Brain/metabolism , Dyskinesia, Drug-Induced/drug therapy , Half-Life , High-Throughput Screening Assays , Humans , Hyperthermia, Induced , Indoles/pharmacokinetics , Indoles/pharmacology , Indoles/therapeutic use , Levodopa/toxicity , Male , Mice , Protein Binding/drug effects , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
12.
J Biol Chem ; 289(16): 10975-10987, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24596089

ABSTRACT

The metabotropic glutamate receptor subtype 7 (mGlu7) is an important presynaptic regulator of neurotransmission in the mammalian CNS. mGlu7 function has been linked to autism, drug abuse, anxiety, and depression. Despite this, it has been difficult to develop specific blockers of native mGlu7 signaling in relevant brain areas such as amygdala and limbic cortex. Here, we present the mGlu7-selective antagonist 7-hydroxy-3-(4-iodophenoxy)-4H-chromen-4-one (XAP044), which inhibits lateral amygdala long term potentiation (LTP) in brain slices from wild type mice with a half-maximal blockade at 88 nm. There was no effect of XAP044 on LTP of mGlu7-deficient mice, indicating that this pharmacological effect is mGlu7-dependent. Unexpectedly and in contrast to all previous mGlu7-selective drugs, XAP044 does not act via the seven-transmembrane region but rather via a binding pocket localized in mGlu7's extracellular Venus flytrap domain, a region generally known for orthosteric agonist binding. This was shown by chimeric receptor studies in recombinant cell line assays. XAP044 demonstrates good brain exposure and wide spectrum anti-stress and antidepressant- and anxiolytic-like efficacy in rodent behavioral paradigms. XAP044 reduces freezing during acquisition of Pavlovian fear and reduces innate anxiety, which is consistent with the phenotypes of mGlu7-deficient mice, the results of mGlu7 siRNA knockdown studies, and the inhibition of amygdala LTP by XAP044. Thus, we present an mGlu7 antagonist with a novel molecular mode of pharmacological action, providing significant application potential in psychiatry. Modeling the selective interaction between XAP044 and mGlu7's Venus flytrap domain, whose three-dimensional structure is already known, will facilitate future drug development supported by computer-assisted drug design.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Behavior, Animal , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Stress, Psychological/metabolism , Amygdala/pathology , Animals , Anxiety/drug therapy , Anxiety/genetics , Anxiety/pathology , CHO Cells , Cricetinae , Cricetulus , L Cells , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Mice , Mice, Mutant Strains , Protein Structure, Tertiary , Receptors, Metabotropic Glutamate/genetics , Stress, Psychological/drug therapy , Stress, Psychological/genetics , Stress, Psychological/pathology
13.
Neuropharmacology ; 72: 215-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23664812

ABSTRACT

Glutamate transmission and synaptic plasticity in the amygdala are essential for the learning and expression of conditioned fear. Glutamate activates both ionotropic glutamate receptors and eight subtypes of metabotropic glutamate receptors (mGlu1-8). In the present study, we investigated the roles of mGlu7 and mGlu8 in amygdala-dependent behavior and synaptic plasticity. We show that ablation of mGlu7 but not mGlu8 attenuates long-term potentiation (LTP) at thalamo-lateral amygdala (LA) synapses where a strong association between LTP and learning has been demonstrated. mGlu7-deficient mice express a general deficit in conditioned fear whereas mGlu8-deficient mice show a dramatic reduction in contextual fear. The mGlu7 agonist AMN082 reduced thalamo-LA LTP and intra-amygdala administration blocked conditioned fear learning. In contrast, the mGlu8 agonist DCPG decreased synaptic transmission but not LTP at thalamo-LA synapses. Intra-amygdala DCPG selectively reduced the expression of contextual fear but did not affect the acquisition and expression of cued fear. Taken together, these data revealed very different roles for mGlu7 and mGlu8 in amygdala synaptic transmission, fear learning and its expression. These receptors seem promising targets for treating anxiety disorders with different underlying pathologies with exaggerated fear learning (mGlu7) or contextual fear (mGlu8).


Subject(s)
Amygdala/physiology , Conditioning, Psychological/physiology , Receptors, Metabotropic Glutamate/metabolism , Amygdala/drug effects , Animals , Biophysics , Conditioning, Psychological/drug effects , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Fear/drug effects , Fear/physiology , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement/drug effects , Movement/physiology , Receptors, Metabotropic Glutamate/deficiency , Time Factors
14.
Neuropharmacology ; 72: 66-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603202

ABSTRACT

Our knowledge regarding the molecular pathophysiology underlying anxiety disorders remains incomplete. Increasing evidence points to a role of glutamate in anxiety. The group III metabotropic glutamate receptors (mGlu4, mGlu6, mGlu7 and mGlu8 receptors) remain the least investigated glutamate receptor subtypes partially due to a delay in the development of specific pharmacological tools. Early work using knockout animals and pharmacological tools aimed at investigating the role of mGlu7 receptor in the pathophysiology of anxiety disorders has yielded exciting yet not always consistent results. To further investigate the role this receptor plays in anxiety-like behaviour, we knocked down mGlu7 receptor mRNA levels in the adult mouse brain using siRNA delivered via an osmotic minipump. This reduced anxiety-like behaviour in the light-dark box coupled with an attenuation of stress-induced hyperthermia (SIH) and a reduction of the acoustic startle response (ASRs) in the fear-potentiated startle paradigm (FPS). These effects on anxiety-like behaviour were independent of any impairment of locomotor activity and surprisingly, no behavioural changes were observed in the forced swim test (FST), which is in contrast to mGlu7 receptor knockout animals. Furthermore, the previously reported epilepsy-prone phenotype seen in mGlu7 receptor knockout animals was not observed following siRNA-induced knockdown of the receptor. These data suggest targeting mGlu7 receptors with selective antagonist drugs may be an effective and safe strategy for the treatment of anxiety disorders.


Subject(s)
Anxiety/drug therapy , Anxiety/metabolism , RNA, Small Interfering/therapeutic use , Receptors, Metabotropic Glutamate/metabolism , Adaptation, Ocular/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Fear/drug effects , Hyperthermia, Induced/psychology , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Pentylenetetrazole/toxicity , Receptors, Metabotropic Glutamate/genetics , Reflex, Startle/drug effects , Seizures/chemically induced , Seizures/drug therapy , Stress, Physiological/physiology , Swimming/psychology
15.
Addict Biol ; 18(1): 66-77, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23126471

ABSTRACT

Chronic stress is known to enhance the susceptibility for addiction disorders including alcoholism. While these findings have been recapitulated in animal models, the majority of these studies have utilized non-social rather than social stress paradigms; the latter of which are believed to be more relevant to the human situation. Therefore, the major aim of this study was to investigate, if 14 days of chronic subordinate colony housing (CSC), a pre-clinically validated psychosocial stress paradigm relevant for human psychiatric and somatic disorders, enhances ethanol (EtOH) consumption in male mice. To assess this, we employed the well-established two-bottle free-choice paradigm where mice were given access to water and 2, 4, 6 and 8% EtOH solutions (with the concentrations increasing each fourth day) following termination of the stress procedure. After 14 days of CSC, stressed mice consumed significantly more EtOH at all concentrations tested and displayed increased EtOH preference at concentrations of 6 and 8%. This effect was not due to an altered taste preference in CSC mice as assessed by saccharine- and quinine-preference tests, but was accompanied by increased anxiety-related behavior. Systemic administration of baclofen (2.5 mg/kg) or oxytocin (OXT; 10 mg/kg) reduced the EtOH intake in single housed control (baclofen, OXT) and CSC (baclofen) mice, whereas intracerebroventricular OXT (0.5 µg/2 µl) was ineffective in both groups. Taken together, these results suggest that (i) chronic psychosocial stress enhances EtOH consumption, and (ii) baclofen and OXT differentially affect EtOH intake in mice.


Subject(s)
Alcohol Drinking , Baclofen/pharmacology , Ethanol/administration & dosage , GABA-B Receptor Agonists/pharmacology , Oxytocin/pharmacology , Stress, Psychological/psychology , Adolescent , Alcoholism/drug therapy , Alcoholism/psychology , Analysis of Variance , Animals , Baclofen/administration & dosage , Disease Models, Animal , Drinking Behavior , Ethanol/adverse effects , Food Preferences , GABA-B Receptor Agonists/administration & dosage , Housing, Animal , Humans , Injections, Intraperitoneal , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Oxytocin/administration & dosage , Self Administration
16.
Biochem Pharmacol ; 84(4): 414-24, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22554564

ABSTRACT

Group-III metabotropic glutamate receptors (mGluRs) comprise four structurally related brain and retinal G protein-coupled receptors (GPCRs), mGluR4, mGluR6, mGluR7 and mGluR8, which receive much attention as promising targets for nervous system drugs. In particular, activation of mGluR4 is a major focus for the development of new therapeutics in Parkinson's disease, while mGluR7 activation is considered a potential approach for future treatments of specific psychiatric conditions. The first generation group-III mGluR agonists, e.g.l-AP4 and l-SOP, are characterized by an essential phosphonate functional group, which became a major limitation for the development of systemically active, potent and receptor subtype-selective drugs. Recently however, two approaches emerged in parallel providing resolution to this constraint: in silico high-throughput screening of chemical libraries against a 3D-model of the mGluR4 extracellular domain identified a hit that was optimized into a series of potent and subtype-selective orthosteric agonists with drug-like properties and novel chemotype structures; secondly, high-throughput random screening of chemical libraries against recombinantly expressed group-III receptors identified diverse chemical sets of allosteric agonists and positive modulators, which are drug-like, display selectivity for mGluR4, mGluR7, or mGluR8 and act via novel pharmacological sites. Here, we illustrate new scientific insights obtained via the use of those strategies. Also, we compare advantages and disadvantages of both approaches to identify the desired group-III mGluR activators and we conclude with suggestions how to employ those discovery strategies with success for the identification, optimization, and development of clinical drug candidates; this may have important implications for the entire field of GPCR research.


Subject(s)
Central Nervous System Agents/chemistry , Receptors, Metabotropic Glutamate/agonists , Allosteric Regulation , Animals , Central Nervous System Agents/pharmacology , Humans , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism
17.
Addict Biol ; 17(2): 235-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21392179

ABSTRACT

Growing evidence supports a role for the central nervous system (CNS) neurotransmitter L-glutamate and its metabotropic receptors (mGluRs) in drug addiction in general and alcohol-use disorders in particular. Alcohol dependence, for instance, has a genetic component, and the recent discovery that variations in the gene coding for mGluR7 modulate alcohol consumption further validates involvement of the L-glutamate system. Consequently, increasing interest emerges in developing L-glutamatergic therapies for the treatment of alcohol abuse and dependence. To this end, we performed a detailed behavioral pharmacology study to investigate the regulation of alcohol consumption and preference following administration of the mGluR7-selective drugs N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082) and 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP). Upon administration of the allosteric agonist AMN082 (10 mg/kg, i.p.) in rats, there was a significant decrease in ethanol consumption and preference, without affecting ethanol blood metabolism. In contrast, mGluR7 blockade with MMPIP (10 mg/kg, i.p.) showed an increase in alcohol intake and reversed AMN082's effect on ethanol consumption and preference. Both mGluR7-directed pharmacological tools had no effect on total fluid intake, taste preference, or on spontaneous locomotor activity. In conclusion, these findings support a specific regulatory role for mGluR7 on alcohol drinking and preference and provide evidence for the use of AMN082-type drugs as potential new treatments for alcohol-use disorders in man.


Subject(s)
Alcohol Drinking/drug therapy , Benzhydryl Compounds/pharmacology , Pyridones/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Alcohol Drinking/psychology , Animals , Ethanol/metabolism , Food Preferences/drug effects , Male , Motor Activity/drug effects , Quinine/pharmacology , Rats , Rats, Wistar , Saccharin/pharmacology , Self Administration , Sweetening Agents/pharmacology , Taste/drug effects
18.
Neuropharmacology ; 62(4): 1619-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22079159

ABSTRACT

Fear extinction is defined as the attenuation of a conditioned-fear memory by re-exposing animals to the conditioned stimulus without the aversive stimulus. This process is known to be effectively enhanced via administration of D-cycloserine (DCS), a partial NMDA-receptor agonist. However, other glutamatergic mechanisms, such as interference with metabotropic glutamate receptor (mGluR) subtypes 5 and 7 in the extinction of aversive memories are insufficiently understood. Using the allosteric mGluR5 receptor antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP), the mGluR7 allosteric agonist N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082), and DCS for comparison, we aimed to study how pharmacological blockade of mGluR5 and activation of mGluR7 influenced within- and between-session conditioned-fear extinction training and extinction retention in rats. We show that when injected before extinction training, mGluR7 activation with AMN082 enhanced freezing and thereby attenuated within-session fear extinction, whereas both DCS and the mGluR5 receptor antagonist MPEP had no effect on this process. However, these differential drug effects were not long lasting, as no difference in extinction retention were observed 24 h later. Therefore, we assessed whether the compounds affect 24 h consolidation of extinction training following incomplete extinction training (between-session extinction). Similar to DCS, AMN082- but not MPEP-treated rats showed facilitated extinction retention, as exhibited by decreased freezing. Finally, using fluoxetine, we provide evidence that the effect of AMN082 on between-session extinction retention is most likely not via increasing 5-HT transmission. These findings demonstrate that mGluR7 activation differentially modulates conditioned-fear extinction, in dependence on the protocol employed, and suggests drugs with AMN082-like mechanisms as potential add-on drugs following exposure-based psychotherapy for fear-related human disorders.


Subject(s)
Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , Fear/drug effects , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Benzhydryl Compounds/pharmacology , Conditioning, Classical/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/physiology , Fear/physiology , Fluoxetine/pharmacology , Male , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Selective Serotonin Reuptake Inhibitors/pharmacology
19.
Pharmacol Rev ; 63(1): 35-58, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228260

ABSTRACT

Metabotropic glutamate (mGlu) receptors are G-protein-coupled receptors expressed primarily on neurons and glial cells, where they are located in the proximity of the synaptic cleft. In the central nervous system (CNS), mGlu receptors modulate the effects of l-glutamate neurotransmission in addition to that of a variety of other neurotransmitters. However, mGlu receptors also have a widespread distribution outside the CNS that has been somewhat neglected to date. Based on this expression, diverse roles of mGlu receptors have been suggested in a variety of processes in health and disease including controlling hormone production in the adrenal gland and pancreas, regulating mineralization in the developing cartilage, modulating lymphocyte cytokine production, directing the state of differentiation in embryonic stem cells, and modulating gastrointestinal secretory function. Understanding the role of mGlu receptors in the periphery will also provide a better insight into potential side effects of drugs currently being developed for neurological and psychiatric conditions. This review summarizes the new potential roles of mGlu receptors and raises the possibility of novel pharmacological targets for various disorders.


Subject(s)
Receptors, Metabotropic Glutamate/physiology , Animals , Glutamic Acid/metabolism , Humans , Molecular Targeted Therapy , Organ Specificity , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors
20.
Eur J Pharmacol ; 639(1-3): 123-31, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20371242

ABSTRACT

Understanding the complex interaction between stress and genetics that leads to the manifestation of disorders such as depression, anxiety, and cognitive dysfunction is one of the key areas of research in modern neuroscience. Growing evidence suggests that the glutamatergic system may be a relevant therapeutic target for such disorders. Glutamate is the neurotransmitter at the vast majority of excitatory synapses in the brain, and metabotropic glutamate (mGlu) receptor subtypes (mGlu(1) receptor-mGlu(8) receptor) act as important pre- and postsynaptic regulators of neurotransmission in the central nervous system (CNS), providing a mechanism by which fast synaptic responses through ligand-gated cation channels can be fine-tuned. Thus mGlu receptors are poised to participate in a wide variety of functions of the CNS. The presynaptic mGlu(7) receptor shows the highest evolutionary conservation within the family and it is thought to regulate neurotransmitter release. The mGlu(7) receptor is also the most widely distributed of the presynaptic mGlu receptors and is present at a broad range of synapses that are postulated to be critical for both normal CNS function and a range of psychiatric and neurological disorders. A growing body of evidence suggests that the mGlu(7) receptor is a key player in shaping synaptic responses at glutamatergic synapses as well as being a key regulator of inhibitory GABAergic transmission. The development of selective pharmacological and genetic tools has allowed for the unravelling of mGlu(7) receptor function in a host of physiological and behavioural processes. Knockout mice and siRNA knockdown has pointed to a role of the mGlu(7) receptor in anxiety, extinction of fear and aversion learning, spatial memory and the hormonal response to stress. In addition, these studies are largely supported by pharmacological manipulation of mGlu(7) receptor using the selective modulator N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082), although paradoxical effects with this agonist have also emerged. Together these data suggest that the mGlu(7) receptor is an important regulator of glutamatergic function, of fear and aversion and cognition and thus this receptor represents an innovative therapeutic target for stress-related disorders at the interface of cognition and anxiety.


Subject(s)
Cognition/physiology , Emotions/physiology , Excitatory Amino Acid Agents/therapeutic use , Receptors, Metabotropic Glutamate/metabolism , Synaptic Transmission/drug effects , Animals , Cognition/drug effects , Emotions/drug effects , Excitatory Amino Acid Agents/pharmacology , Humans , Mental Disorders/drug therapy , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL