Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Biol Psychiatry ; 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38061467

ABSTRACT

BACKGROUND: Polymorphisms in the gene encoding for metabotropic glutamate receptor 3 (mGlu3) are associated with an increased likelihood of schizophrenia diagnosis and can predict improvements in negative symptoms following treatment with antipsychotics. However, the mechanisms by which mGlu3 can regulate brain circuits involved in schizophrenia pathophysiology are not clear. METHODS: We employed selective pharmacological tools and a variety of approaches including whole-cell patch-clamp electrophysiology, slice optogenetics, and fiber photometry to investigate the effects of mGlu3 activation on phencyclidine (PCP)-induced impairments in thalamo-accumbal transmission and sociability deficits. A chemogenetic approach was used to evaluate the role of thalamo-accumbal transmission in PCP-induced sociability deficits. RESULTS: We first established that PCP treatment augmented excitatory transmission onto dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) in the nucleus accumbens (NAc) and induced sociability deficits. Our studies revealed a selective increase in glutamatergic synaptic transmission from thalamic afferents to D1-MSNs in the NAc shell. Chemogenetic silencing of thalamo-accumbal inputs rescued PCP-induced sociability deficits. Pharmacological activation of mGlu3 normalized PCP-induced impairments in thalamo-accumbal transmission and sociability deficits. Mechanistic studies revealed that mGlu3 activation induced robust long-term depression at synapses from the thalamic projections onto D1-MSNs in the NAc shell. CONCLUSIONS: These data demonstrate that activation of mGlu3 decreases thalamo-accumbal transmission and thereby rescues sociability deficits in mouse modeling schizophrenia-like symptoms. These findings provide novel insights into the NAc-specific mechanisms and suggest that agents modulating glutamatergic signaling in the NAc may provide a promising approach for treating negative symptoms in schizophrenia.

2.
Neuropharmacology ; 235: 109569, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37142158

ABSTRACT

Cellular responses to metabotropic glutamate (mGlu) receptor activation are shaped by mechanisms of receptor-receptor interaction. mGlu receptor subtypes form homodimers, intra- or inter-group heterodimers, and heteromeric complexes with other G protein-coupled receptors (GPCRs). In addition, mGlu receptors may functionally interact with other receptors through the ßγ subunits released from G proteins in response to receptor activation or other mechanisms. Here, we discuss the interactions between (i) mGlu1 and GABAB receptors in cerebellar Purkinje cells; (ii) mGlu2 and 5-HT2Aserotonergic receptors in the prefrontal cortex; (iii) mGlu5 and A2A receptors or mGlu5 and D1 dopamine receptors in medium spiny projection neurons of the indirect and direct pathways of the basal ganglia motor circuit; (iv) mGlu5 and A2A receptors in relation to the pathophysiology of Alzheimer's disease; and (v) mGlu7 and A1 adenosine or α- or ß1 adrenergic receptors. In addition, we describe in detail a novel form of non-heterodimeric interaction between mGlu3 and mGlu5 receptors, which appears to be critically involved in mechanisms of activity-dependent synaptic plasticity in the prefrontal cortex and hippocampus. Finally, we highlight the potential implication of these interactions in the pathophysiology and treatment of cerebellar disorders, schizophrenia, Alzheimer's disease, Parkinson's disease, l-DOPA-induced dyskinesias, stress-related disorders, and cognitive dysfunctions. This article is part of the Special Issue on "The receptor-receptor interaction as a new target for therapy".


Subject(s)
Alzheimer Disease , Parkinson Disease , Humans , Levodopa , Parkinson Disease/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Basal Ganglia/metabolism
3.
Pharmacol Biochem Behav ; 221: 173493, 2022 11.
Article in English | MEDLINE | ID: mdl-36402243

ABSTRACT

Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS) and abnormalities in the glutamatergic system underlie various CNS disorders. As metabotropic glutamate receptor 3 (mGlu3 receptor) regulates glutamatergic transmission in various brain areas, emerging literature suggests that targeting mGlu3 receptors can be a novel approach to the treatment of psychiatric and neurological disorders. For example, mGlu3 receptor negative allosteric modulators (NAMs) induce rapid antidepressant-like effects in both acute and chronic stress models. Activation of mGlu3 receptors can enhance cognition in the rodents modeling schizophrenia-like pathophysiology. The mGlu3 receptors expressed in the astrocytes induce neuroprotective effects. Although polymorphisms in GRM3 have been shown to be associated with addiction, there is not significant evidence about the efficacy of mGlu3 receptor ligands in rodent models of addiction. Collectively, drugs targeting mGlu3 receptors may provide an alternative approach to fill the unmet clinical need for safer and more efficacious therapeutics for CNS disorders.


Subject(s)
Nervous System Diseases , Receptors, Metabotropic Glutamate , Humans , Nervous System Diseases/drug therapy , Central Nervous System , Glutamic Acid
4.
J Biol Chem ; 298(10): 102458, 2022 10.
Article in English | MEDLINE | ID: mdl-36063995

ABSTRACT

Glutamate acts at eight metabotropic glutamate (mGlu) receptor subtypes expressed in a partially overlapping fashion in distinct brain circuits. Recent evidence indicates that specific mGlu receptor protomers can heterodimerize and that these heterodimers can exhibit different pharmacology when compared to their homodimeric counterparts. Group III mGlu agonist-induced suppression of evoked excitatory potentials and induction of long-term potentiation at Schaffer collateral-CA1 (SC-CA1) synapses in the rodent hippocampus can be blocked by the selective mGlu7 negative allosteric modulator (NAM), ADX71743. Curiously, a different mGlu7 NAM, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one, failed to block these responses in brain slices despite its robust activity at mGlu7 homodimers in vitro. We hypothesized that this might result from heterodimerization of mGlu7 with another mGlu receptor protomer and focused on mGlu8 as a candidate given the reported effects of mGlu8-targeted compounds in the hippocampus. Here, we used complemented donor acceptor-resonance energy transfer to study mGlu7/8 heterodimer activation in vitro and observed that ADX71743 blocked responses of both mGlu7/7 homodimers and mGlu7/8 heterodimers, whereas 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one only antagonized responses of mGlu7/7 homodimers. Taken together with our electrophysiology observations, these results suggest that a receptor with pharmacology consistent with an mGlu7/8 heterodimer modulates the activity of SC-CA1 synapses. Building on this hypothesis, we identified two additional structurally related mGlu7 NAMs that also differ in their activity at mGlu7/8 heterodimers, in a manner consistent with their ability to inhibit synaptic transmission and plasticity at SC-CA1. Thus, we propose that mGlu7/8 heterodimers are a key molecular target for modulating the activity of hippocampal SC-CA1 synapses.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate , Synapses , Hippocampus/metabolism , Long-Term Potentiation , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Animals , Rodentia , Saccharomyces cerevisiae , Electrophysiology
5.
Mol Pharmacol ; 101(5): 275-285, 2022 05.
Article in English | MEDLINE | ID: mdl-35246479

ABSTRACT

Accumulating evidence of glutamatergic abnormalities in the brains of schizophrenia patients has led to efforts to target various components of glutamatergic signaling as potential new approaches for schizophrenia. Exciting research suggests that metabotropic glutamate (mGlu) receptors could provide a fundamentally new approach for better symptomatic relief in patients with schizophrenia. In preclinical studies, the mGlu5 receptor positive allosteric modulators (PAMs) show efficacy in animal models relevant for all symptom domains in schizophrenia. Interestingly, biased pure mGlu5 receptor PAMs that do not potentiate coupling of mGlu5 receptors to N-methyl-D-aspartate (NMDA) receptors lack neurotoxic effects associated with mGlu5 PAMs that enhance coupling to NMDA receptors or have allosteric agonist activity. This provides a better therapeutic profile for treating schizophrenia-like symptoms. Additionally, the mGlu1 receptor PAMs modulate dopamine release in the striatum, which may contribute to their antipsychotic-like effects. Besides group I mGlu (mGlu1 and mGlu5) receptors, agonists of mGlu2/3 receptors also induce robust antipsychotic-like and procognitive effects in rodents and may be effective in treating symptoms of schizophrenia in a selective group of patients. Additionally, mGlu2/4 receptor heterodimers modulate glutamatergic neurotransmission in the prefrontal cortex at selective synapses activated in schizophrenia and therefore hold potential as novel antipsychotics. Excitingly, the mGlu3 receptor activation can enhance cognition in rodents, suggesting that mGlu3 receptor agonist/PAM could provide a novel approach for the treatment of cognitive deficits in schizophrenia. Collectively, the development of mGlu receptor-specific ligands may provide an alternative approach to meet the clinical need for safer and more efficacious therapeutics for schizophrenia. SIGNIFICANCE STATEMENT: The currently available antipsychotic medications do not show significant efficacy for treating negative symptoms and cognitive deficits in schizophrenia. Emerging preclinical and clinical literature suggests that pharmacological targeting of metabotropic glutamate receptors could potentially provide an alternative approach for designing safer and more efficacious therapeutics for treating schizophrenia.


Subject(s)
Antipsychotic Agents , Schizophrenia , Allosteric Regulation , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Glutamic Acid , Humans , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/drug therapy
6.
Neuron ; 110(6): 1068-1083.e5, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35045338

ABSTRACT

Inhibitory interneurons orchestrate prefrontal cortex (PFC) activity, but we have a limited understanding of the molecular and experience-dependent mechanisms that regulate synaptic plasticity across PFC microcircuits. We discovered that mGlu5 receptor activation facilitates long-term potentiation at synapses from the basolateral amygdala (BLA) onto somatostatin-expressing interneurons (SST-INs) in mice. This plasticity appeared to be recruited during acute restraint stress, which induced intracellular calcium mobilization within SST-INs and rapidly potentiated postsynaptic strength onto SST-INs. Restraint stress and mGlu5 receptor activation each augmented BLA recruitment of SST-IN phasic feedforward inhibition, shunting information from other excitatory inputs, including the mediodorsal thalamus. Finally, studies using cell-type-specific mGlu5 receptor knockout mice revealed that mGlu5 receptor function in SST-expressing cells is necessary for restraint stress-induced changes to PFC physiology and related behaviors. These findings provide new insights into interneuron-specific synaptic plasticity mechanisms and suggest that SST-IN microcircuits may be promising targets for treating stress-induced psychiatric diseases.


Subject(s)
Interneurons , Somatostatin , Animals , Interneurons/physiology , Long-Term Potentiation , Mice , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Somatostatin/metabolism , Synapses/physiology
7.
Neuropharmacology ; 196: 108687, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34175327

ABSTRACT

The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".


Subject(s)
Anxiety Disorders/drug therapy , Brain/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Stress, Psychological/drug therapy , Allosteric Regulation , Animals , Humans , Ketamine/therapeutic use , Molecular Targeted Therapy , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
8.
Biol Psychiatry ; 90(6): 385-398, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33965197

ABSTRACT

BACKGROUND: Polymorphisms in GRM3, the gene encoding the mGlu3 metabotropic glutamate receptor, are associated with impaired cognition and neuropsychiatric disorders such as schizophrenia. Limited availability of selective genetic and molecular tools has hindered progress in developing a clear understanding of the mechanisms through which mGlu3 receptors regulate synaptic plasticity and cognition. METHODS: We examined associative learning in mice with trace fear conditioning, a hippocampal-dependent learning task disrupted in patients with schizophrenia. Underlying cellular mechanisms were assessed using ex vivo hippocampal slice preparations with selective pharmacological tools and selective genetic deletion of mGlu3 receptor expression in specific neuronal subpopulations. RESULTS: mGlu3 receptor activation enhanced trace fear conditioning and reversed deficits induced by subchronic phencyclidine. Mechanistic studies revealed that mGlu3 receptor activation induced metaplastic changes, biasing afferent stimulation to induce long-term potentiation through an mGlu5 receptor-dependent, endocannabinoid-mediated, disinhibitory mechanism. Selective genetic deletion of either mGlu3 or mGlu5 from hippocampal pyramidal cells eliminated effects of mGlu3 activation, revealing a novel mechanism by which mGlu3 and mGlu5 interact to enhance cognitive function. CONCLUSIONS: These data demonstrate that activation of mGlu3 receptors in hippocampal pyramidal cells enhances hippocampal-dependent cognition in control and impaired mice by inducing a novel form of metaplasticity to regulate circuit function, providing a clear mechanism through which genetic variation in GRM3 can contribute to cognitive deficits. Developing approaches to positively modulate mGlu3 receptor function represents an encouraging new avenue for treating cognitive disruption in schizophrenia and other psychiatric diseases.


Subject(s)
Receptors, Metabotropic Glutamate , Schizophrenia , Animals , Cognition , Hippocampus/metabolism , Long-Term Potentiation , Mice , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/genetics
9.
Neuropsychopharmacology ; 46(12): 2148-2157, 2021 11.
Article in English | MEDLINE | ID: mdl-34035469

ABSTRACT

Clinical and translational studies suggest that prefrontal cortex (PFC) dysregulation is a hallmark feature of several affective disorders. Thus, investigating the mechanisms involved in the regulation of PFC function and synaptic plasticity could aid in developing new medications. In recent years, the mGlu2 and mGlu3 subtypes of metabotropic glutamate (mGlu) receptors have emerged as exciting potential targets for the treatment of affective disorders, as mGlu2/3 antagonists exert antidepressant-like effects across many rodent models. Several recent studies suggest that presynaptic mGlu2 receptors may contribute to these effects by regulating excitatory transmission at synapses from the thalamus to the PFC. Interestingly, we found that mGlu3 receptors also inhibit excitatory drive to the PFC but act by inducing long-term depression (LTD) at amygdala-PFC synapses. It remains unclear, however, whether blockade of presynaptic, postsynaptic, or glial mGlu3 receptors contribute to long-term effects on PFC circuit function and antidepressant-like effects of mGlu2/3 antagonists. To address these outstanding questions, we leveraged transgenic Grm3fl/fl mice and viral-mediated gene transfer to genetically ablate mGlu3 receptors from pyramidal cells in the frontal cortex of adult mice of all sexes. Consistent with a role for mGlu3 in PFC pyramidal cells, mGlu3-dependent amygdala-cortical LTD was eliminated following mGlu3 receptor knockdown. Furthermore, knockdown mice displayed a modest, task-specific anxiolytic phenotype and decreased passive coping behaviors. These studies reveal that postsynaptic mGlu3 receptors are critical for mGlu3-dependent LTD and provide convergent genetic evidence suggesting that modulating cortical mGlu3 receptors may provide a promising new approach for the treatment of mood disorders.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate/genetics , Animals , Mice , Neuronal Plasticity , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism
10.
J Med Chem ; 62(9): 4638-4655, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30998358

ABSTRACT

The histamine 3 receptor (H3R) is a presynaptic receptor, which modulates several neurotransmitters including histamine and various essential physiological processes, such as feeding, arousal, cognition, and pain. The H3R is considered as a drug target for the treatment of several central nervous system disorders. We have synthesized and identified a novel series of 4-aryl-6-methyl-5,6,7,8-tetrahydroquinazolinamines that act as selective H3R antagonists. Among all the synthesized compounds, in vitro and docking studies suggested that the 4-methoxy-phenyl-substituted tetrahydroquinazolinamine compound 4c has potent and selective H3R antagonist activity (IC50 < 0.04 µM). Compound 4c did not exhibit any activity on the hERG ion channel and pan-assay interference compounds liability. Pharmacokinetic studies showed that 4c crosses the blood brain barrier, and in vivo studies demonstrated that 4c induces anorexia and weight loss in obese, but not in lean mice. These data reveal the therapeutic potential of 4c as an anti-obesity candidate drug via antagonizing the H3R.


Subject(s)
Anti-Obesity Agents/therapeutic use , Histamine H3 Antagonists/therapeutic use , Obesity/drug therapy , Quinazolines/therapeutic use , Receptors, Histamine H3/metabolism , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Blood Glucose/metabolism , Diet, High-Fat , HEK293 Cells , Histamine H3 Antagonists/chemical synthesis , Histamine H3 Antagonists/pharmacokinetics , Humans , Male , Mice, Inbred C57BL , Molecular Structure , Proto-Oncogene Proteins c-fos/metabolism , Quinazolines/chemical synthesis , Quinazolines/pharmacokinetics , Stereoisomerism , Structure-Activity Relationship , Weight Loss/drug effects
11.
Bioorg Med Chem Lett ; 29(4): 585-590, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30600206

ABSTRACT

Isocoumarins are lactone ring-containing natural products, are quite abundant in microbes and higher plants, and have been shown to exhibit a broad range of pharmacological properties. However, the molecular mechanism or target of this class of molecules is not known. In this study, we have synthesized 14 isocoumarin derivatives and evaluated for their activity at TrkB receptor in transiently transfected HEK293T cells. We identified 8-hydroxy-3-aryl isocoumarin (1) as a high-affinity agonist at the TrkB receptor. We also demonstrated that isocoumarin 1 activated endogenously TrkB receptor in primary cortical neurons and modulated various markers of synaptic plasticity, and increased dendritic arborization. These results indicate therapeutic potential and molecular target of 8-hydroxy-3-aryl isocoumarin 1 for the treatment of various CNS disorders.


Subject(s)
Isocoumarins/pharmacology , Neurons/drug effects , Receptor, trkB/agonists , HEK293 Cells , Hippocampus/cytology , Hippocampus/drug effects , Humans , Isocoumarins/chemistry , MAP Kinase Signaling System , Neurons/physiology , Phosphorylation , Receptor, trkB/metabolism
12.
Psychoneuroendocrinology ; 101: 128-137, 2019 03.
Article in English | MEDLINE | ID: mdl-30458370

ABSTRACT

The last two decades of research has established histamine (HA) as a neurotransmitter. Since H3R antagonists are known to modulate several neurotransmitters besides HA, H3R antagonists have shown potential for the treatment of different central nervous system disorders, including depression. However, molecular mechanisms underlying the beneficial effects of H3R antagonism in depression are not clear, yet. In the present study, we investigated the antidepressant potential of ciproxifan, a selective H3R antagonist, in chronic unpredictable stress (CUS) model of depression in C57BL/6 J mice. We observed that chronic treatment of CUS mice with ciproxifan (3 mg/kg i.p.; for three weeks) alleviates depression-like symptoms such as helplessness measured by forced swim and tail suspension test (FST and TST), anhedonia measured by sucrose preference test (SPT) and social deficit measured in social behavior test. Chronic ciproxifan treatment restored CUS induced BDNF expression in the prefrontal cortex (PFC) and hippocampus. We also observed that ciproxifan modulates CUS induced NUCB2/nesfatin-1 and CRH expression in the hypothalamus and plasma corticosterone. We also determined the direct effect of HA on BDNF expression in neurons by western blotting and immunocytochemistry, and found that HA significantly induced BDNF expression, which was blocked by the H4R selective antagonist, but not by other HA receptor selective antagonists. Furthermore, ciproxifan significantly modulated NMDA glutamate receptor subunits NR2B and NR2A. Thus, these results suggest that increased HA signaling in the brain produces antidepressant-like effects in mice and modulates BDNF expression and HPA-axis.


Subject(s)
Depression/drug therapy , Histamine Antagonists/pharmacology , Receptors, Histamine H3/metabolism , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/blood , Depression/metabolism , Depressive Disorder/metabolism , Disease Models, Animal , Hippocampus/metabolism , Histamine/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/metabolism , Stress, Psychological/metabolism
13.
Neurobiol Dis ; 118: 94-107, 2018 10.
Article in English | MEDLINE | ID: mdl-29981843

ABSTRACT

GPR40 (Free fatty acid receptor 1) has emerged as an important therapeutic target for diabetes. Several studies have demonstrated the association of comorbid psychiatric conditions with decreased n-3 polyunsaturated fatty acids, which may act as an agonist for GPR40. In this study, we for the first time provide evidence of reduced GPR40 signaling in the hippocampus and cortex which may be a critical underlying mechanism mediating cognitive deficits in diabesity (diabetes and obesity together). Specifically, we showed decreased GPR40 and brain-derived neurotrophic factor (BDNF) expression in the brain regions of high-fat-diet-induced obese and db/db mice. Next, we demonstrated that chronic treatment with docosahexaenoic acid (DHA) or the synthetic GPR40 agonist, GW9508, significantly alleviates cognitive functions in mice, which correlates with increased BDNF expression in the hippocampus. This supports the hypothesis that DHA improves cognitive function in diabesity via GPR40 agonism. We also showed that DHA specifically activates GPR40 and modulates BDNF expression in primary cortical neurons mediated by the extracellular receptor kinase (ERK) and P38-mitogen-activated protein kinase (MAPK) pathways. Finally, the central nervous system (CNS)-specific blockade of GPR40 signaling abrogated the memory potentiating effects of DHA, and induction of BDNF expression in the hippocampus. Thus, we provided evidence that DHA stimulation of GPR40 mediate some of DHA's beneficial effects in metabolic syndrome and identify GPR40 as a viable therapeutic target for the treatment of CNS-related comorbidities associated with diabesity.


Subject(s)
Association Learning/physiology , Brain-Derived Neurotrophic Factor/biosynthesis , Docosahexaenoic Acids/therapeutic use , Memory Disorders/metabolism , Obesity/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Animals , Association Learning/drug effects , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/agonists , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diet, High-Fat/adverse effects , Docosahexaenoic Acids/pharmacology , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology , Receptors, G-Protein-Coupled/agonists
14.
ChemMedChem ; 13(4): 384-395, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29319226

ABSTRACT

Muscarinic acetylcholine receptors (mAChRs) are important therapeutic targets for several diseases of the central nervous system and periphery. However, the lack of subtype-selective ligands for these receptors is a major challenge. A novel approach involving the integration of a natural product framework with a bioactive molecule (iNPBM) by using gephyrotoxin and the isoindoline framework is demonstrated for the discovery of new and selective mAChR modulators. We established a scalable and versatile synthetic scheme to enable the synthesis of various analogues that provided the first structure-activity relationship study of this class of compounds. Pharmacological profiling of these compounds demonstrated several ligands with high affinity and selectivity for mAChRs. Specifically, RG-06 and RG-09 were found to be antagonists of M3-mAChR, whereas RG-02 was found to be an agonist at M2-mAChR. Furthermore, RG-02 exhibited salutary effects in an established pharmacological model of a cognitive deficit in mice.


Subject(s)
Biological Products/metabolism , Receptors, Muscarinic/metabolism , Alkaloids/chemistry , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Biological Products/chemistry , Biological Products/pharmacology , Catalysis , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Isoindoles/chemistry , Isoindoles/metabolism , Isoindoles/pharmacology , Locomotion/drug effects , Male , Maze Learning/drug effects , Metals/chemistry , Mice , Mice, Inbred C57BL , Molecular Conformation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, Muscarinic/chemistry , Structure-Activity Relationship
15.
Nat Nanotechnol ; 12(12): 1190-1198, 2017 12.
Article in English | MEDLINE | ID: mdl-28967893

ABSTRACT

Beta-arrestins (ßarrs) critically mediate desensitization, endocytosis and signalling of G protein-coupled receptors (GPCRs), and they scaffold a large number of interaction partners. However, allosteric modulation of their scaffolding abilities and direct targeting of their interaction interfaces to modulate GPCR functions selectively have not been fully explored yet. Here we identified a series of synthetic antibody fragments (Fabs) against different conformations of ßarrs from phage display libraries. Several of these Fabs allosterically and selectively modulated the interaction of ßarrs with clathrin and ERK MAP kinase. Interestingly, one of these Fabs selectively disrupted ßarr-clathrin interaction, and when expressed as an intrabody, it robustly inhibited agonist-induced endocytosis of a broad set of GPCRs without affecting ERK MAP kinase activation. Our data therefore demonstrate the feasibility of selectively targeting ßarr interactions using intrabodies and provide a novel framework for fine-tuning GPCR functions with potential therapeutic implications.


Subject(s)
Endocytosis/drug effects , Immunoglobulin Fab Fragments , Peptide Library , Receptors, G-Protein-Coupled/metabolism , Single-Chain Antibodies , Clathrin/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/pharmacology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology
16.
J Med Chem ; 60(15): 6733-6750, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28726402

ABSTRACT

To obtain selective and potent opioid receptor ligands, we synthesized dehydro derivatives of alvimopan and found compound (28f), a selective but modest affinity MOR antagonist weaker than alvimopan (1). We replaced the arylpiperidine unit by an arylpiperazine to obtain the 1-(α-carboxycinnamyl)-4-arylpiperazines like 13h, which to our surprise had no MOR or DOR activity but was a KOR agonist with moderate affinity. In contrast, literature examples of arylpiperazines 4 and 5 were reported to be pan opioid receptor antagonists, while 6 was a MOR agonist. Two compounds (13l and 11b) showed analgesic response in tail flick test which was blocked by pretreatment with norbinaltorphimine (norBNI). Among 10 1-(α-carboxycinnamyl)-4-arylpiperidines, compound 28g and five others were specific MOR antagonists. Interestingly, compound 26b of this series was found to be more potent than naloxone but weaker than 1. Docking studies have explained differential activities of the above piperazines and piperidines.


Subject(s)
Cinnamates/pharmacology , Piperazines/pharmacology , Piperidines/pharmacology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Cinnamates/chemical synthesis , HEK293 Cells , Humans , Ligands , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Naloxone/pharmacology , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/pharmacology , Piperazines/chemical synthesis , Piperidines/chemical synthesis
17.
Mol Biol Cell ; 28(8): 1003-1010, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28228552

ABSTRACT

G protein-coupled receptors (GPCRs) exhibit highly conserved activation and signaling mechanisms by which agonist stimulation leads to coupling of heterotrimeric G proteins and generation of second messenger response. This is followed by receptor phosphorylation, primarily in the carboxyl terminus but also in the cytoplasmic loops, and subsequent binding of arrestins. GPCRs typically recruit arrestins through two different sets of interactions, one involving phosphorylated receptor tail and the other mediated by the receptor core. The engagement of both set of interactions (tail and core) is generally believed to be necessary for arrestin-dependent functional outcomes such as receptor desensitization, endocytosis, and G protein-independent signaling. Here we demonstrate that a vasopressin receptor (V2R) mutant with truncated third intracellular loop (V2RΔICL3) can interact with ß-arrestin 1 (ßarr1) only through the phosphorylated tail without engaging the core interaction. Of interest, such a partially engaged V2RΔICL3-ßarr1 complex can efficiently interact with clathrin terminal domain and ERK2 MAPK in vitro. Furthermore, this core interaction-deficient V2R mutant exhibits efficient endocytosis and ERK activation upon agonist stimulation. Our data suggest that core interaction with ßarr is dispensable for V2R endocytosis and ERK activation and therefore provide novel insights into refining the current understanding of functional requirements in biphasic GPCR-ßarr interaction.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Receptors, Vasopressin/metabolism , beta-Arrestin 1/metabolism , Amino Acid Sequence , Arrestins/metabolism , Clathrin/metabolism , Endocytosis , Enzyme Activation , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , MAP Kinase Signaling System , Phosphorylation , Protein Binding , Receptors, G-Protein-Coupled/metabolism
18.
Sci Rep ; 6: 33401, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27634008

ABSTRACT

Psychotomimetic and prodepressive effect by kappa opioid receptor (KOR) activation in rodents and human is widely known. Significantly, recent clinical investigations demonstrated the salutary effects of KOR antagonists in patients with treatment resistant depression, indicating essential role of KOR signaling in refractory depression. This study was undertaken to reveal the molecular determinant of KOR mediated depression and antidepressant response of KOR antagonist. We observed that chronic KOR activation by U50488, a selective KOR agonist, significantly increased depression like symptoms (behavioral despair, anhedonia and sociability) in C57BL/6J mice, which were blocked by KOR antagonist norBNI and antidepressant imipramine, but not by fluoxetine or citalopram. Further, chronic KOR activation increased phosphorylation of NR2B subunit of NMDA at tyrosine 1472 (pNR2B NMDA) in the hippocampus, but not in the cortex. Similar to behavioral effects norBNI and imipramine, but not SSRIs, blocked NR2B phosphorylation. Moreover, KOR induced depression like behaviors were reversed by NR2B selective inhibitor Ro 25-6981. Mechanistic studies in primary cultured neurons and brain tissues using genetic and pharmacological approaches revealed that stimulation of KOR modulates several molecular correlates of depression. Thus, these findings elucidate molecular mechanism of KOR signaling in treatment resistant depression like behaviors in mice.


Subject(s)
Depressive Disorder, Treatment-Resistant/therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Down-Regulation/drug effects , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice, Inbred C57BL , Neurons/metabolism , Phenols/pharmacology , Phenols/therapeutic use , Phosphorylation/drug effects , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
19.
Biochim Biophys Acta ; 1860(10): 2178-90, 2016 10.
Article in English | MEDLINE | ID: mdl-27180173

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) inhibitor gefitinib (Iressa) is used for treating non-small cell lung cancer. Gefitinib also induces differentiation in acute myeloid leukemia (AML) cell lines and patient samples lacking EGFR by an unknown mechanism. Here we dissected the mechanism of gefitinib action responsible for its EGFR-independent effects. METHODS: Signaling events were analyzed by homogenous time-resolved fluorescence and immunoblotting. Cellular proliferation and differentiation were assessed by ATP measurement, trypan blue exclusion, 5-bromo-2'-deoxyuridine incorporation and flow-cytometry. Gefitinib and G protein-coupled receptor (GPCR) interactions were assessed by ß-arrestin recruitment, luciferase and radioligand competition assays. Role of histamine receptors (HR) in gefitinib actions were assessed by HR knockdown or pharmacological modulation. EGFR and HR interaction was assessed by co-immunoprecipitation. RESULTS: Gefitinib reduced cyclic AMP content in both AML and EGFR-expressing cells and induced ERK phosphorylation in AML cells. Dibutyryl-cAMP or PD98059 suppressed gefitinib-induced AML cell cytostasis and differentiation. Gefitinib bound to and modulated HRs with subtype selectivity. Pharmacological or genetic modulations of H2 and H4 HRs (H2R and H4R) not only suppressed gefitinib-induced cytostasis and differentiation of AML cells but also blocked EGFR and ERK1/2 inhibition in MDA-MB-231 cells. Moreover, in MDA-MB-231 cells gefitinib enhanced EGFR interaction with H4R that was blocked by H4R agonist 4-methyl histamine (4MH). CONCLUSION: HRs play critical roles in anti-cancer effects of gefitinib in both EGFR-deficient and EGFR-rich environments. GENERAL SIGNIFICANCE: We furnish fresh insights into gefitinib functions which may provide new molecular clues to its efficacy and safety issues.


Subject(s)
ErbB Receptors/genetics , Leukemia, Myeloid, Acute/drug therapy , Quinazolines/administration & dosage , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine H2/genetics , Receptors, Histamine/genetics , Antineoplastic Agents/administration & dosage , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , ErbB Receptors/antagonists & inhibitors , Gefitinib , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Receptors, Histamine/metabolism , Receptors, Histamine H2/metabolism , Receptors, Histamine H4
20.
Int J Biochem Cell Biol ; 77(Pt B): 226-39, 2016 08.
Article in English | MEDLINE | ID: mdl-27046448

ABSTRACT

G protein-coupled receptors (GPCRs) act as a relay center through which extracellular signals, in the form of neurotransmitters or therapeutics, are converted into an intracellular response, which ultimately shapes the overall response at the tissue and behavioral level. Remarkably in similar ways, epigenetic mechanisms also modulate the expression pattern of a large number of genes in response to the dynamic environment inside and outside of the body, and consequently overall response. Emerging evidences from the pharmacogenomics and preclinical studies clearly suggest that these two distinct mechanisms criss-cross each other in several neurological disorders. At one hand such cross-talks between two distinct mechanisms make disease etiology more challenging to understand, while on the other hand if dealt appropriately, such situations might provide an opportunity to find novel druggable target and strategy for the treatment of complex diseases. In this review article, we have summarized and highlighted the main findings that tie epigenetic mechanisms to GPCR mediated signaling in the pathophysiology of central nervous system (CNS) disorders, including depression, addiction and pain.


Subject(s)
Epigenesis, Genetic , Mental Disorders/genetics , Mental Disorders/pathology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Animals , Humans , Mental Disorders/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...