1
|
Chruścicka-Smaga B, Sowa-Kućma M, Pańczyszyn-Trzewik P, Bobula B, Korlatowicz A, Latocha K, Pabian P, Czechowska E, Lenda T, Faron-Górecka A, Stachowicz K. Evidence for functional interaction between the CB1 and the mGlu7 receptors mediated signaling in modulation of anxiety behavior and cognition. Life Sci 2025; 361:123313. [PMID: 39674270 DOI: 10.1016/j.lfs.2024.123313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Anxiety is a severe social problem. It is a disease entity that occurs alone or accompanies other diseases such as depression, phobia, or post-traumatic stress disorder. Our earlier studies demonstrated that blockage of arachidonic acid (AA) pathway via inhibition of cyclooxygenase-2 (COX-2) enzyme can modulate mGluRs-induced anxiety-like behavior. Here, we hypothesized that modulation of 2-arachidoglycerol (2-AG), a component of the AA pathway, concomitantly with modulation of mGluR7 signaling, should be adequate to trigger a similar response from the test organism. Since 2-AG is an endogenous agonist for CB1 receptors, we used a CB1/GPR55/μ-opioid receptor antagonist (AM251) alone and in combination with mGluR7 allosteric agonist (AMN082). Stress-induced hyperthermia (SIH) test was performed as a behavioral readout. AM251 has a dual mode on AMN082-mediated effects in SIH in CD-1 mice. Furthermore, the CB1 receptor ligand influenced adaptation to stress in repeated SIH procedures and learning possibilities of mice in the Barnes maze. We also found changes in mGluR7 protein expression levels in the prefrontal cortex (PFC) after mice were exposed to AM251, which showed the potential to attenuate the AMN082-induced decline in mGluR7 levels. The changes induced by AM251 on AMN082-mediated behavioral and biochemical effects were confirmed in electrophysiological experiments in which AM251 abolished AMN082-mediated LTP escalation in PFC. The mGluR7 overexpressed cell line was used to exclude the direct involvement of mGluR7 in AM251 activity. All the above results and the co-localization of CB1 and mGlu7 receptors detected in specific brain regions strongly suggest the specific interaction between CB1 and mGlu7 receptors and their signaling.
Collapse
Affiliation(s)
- Barbara Chruścicka-Smaga
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Magdalena Sowa-Kućma
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| | - Patrycja Pańczyszyn-Trzewik
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Agata Korlatowicz
- Department of Pharmacology, Laboratory of Biochemical Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Latocha
- Department of Pharmacology, Laboratory of Biochemical Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Paulina Pabian
- Department of Pharmacology, Laboratory of Biochemical Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Ewelina Czechowska
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| | - Tomasz Lenda
- Department of Neuro- and Psychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Agata Faron-Górecka
- Department of Pharmacology, Laboratory of Biochemical Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| |
Collapse
|
2
|
Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav 2022; 219:173446. [PMID: 35987339 DOI: 10.1016/j.pbb.2022.173446] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacological modulation of glutamate has long been considered to be of immense therapeutic utility. The metabotropic glutamate receptors (mGluRs) are potential targets for safely altering glutamate-driven excitation. Data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders. For each of the three mGluR groups, compounds have been constructed that produce either potentiation or functional blockade. PET ligands for mGlu5Rs have been studied in a range of patient populations and several mGlu5R antagonists have been tested for potential efficacy in patients including mavoglurant, diploglurant, basimglurant, GET 73, and ADX10059. Efficacy with mGlu5R antagonists has been reported in trials with patients with gastroesophageal reflux disease; data from patients with Parkinson's disease or Fragile X syndrome have not been as robust as hoped. Fenobam was approved for use as an anxiolytic prior to its recognition as an mGlu5R antagonist. mGlu2/3R agonists (pomaglumated methionil) and mGlu2R agonists (JNJ-40411813, AZD 8529, and LY2979165) have been studied in patients with schizophrenia with promising but mixed results. Antagonists of mGlu2/3Rs (decoglurant and TS-161) have been studied in depression where TS-161 has advanced into a planned Phase 2 study in treatment-resistant depression. The Group III mGluRs are the least developed of the mGluR receptor targets. The mGlu4R potentiator, foliglurax, did not meet its primary endpoint in patients with Parkinson's disease. Ongoing efforts to develop mGluR-targeted compounds continue to promise these glutamate modulators as medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | - Kamal P Pandey
- Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| |
Collapse
|
3
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders. SIGNIFICANCE STATEMENT: Allosteric modulation of metabotropic glutamate (mGlu) receptors represents a promising therapeutic strategy to normalize dysregulated cellular physiology associated with neuropsychiatric disease. This review summarizes preclinical and clinical studies using mGlu receptor allosteric modulators as experimental tools and potential therapeutic approaches for the treatment of neuropsychiatric diseases, including schizophrenia, stress, and substance use disorders.
Collapse
|
4
|
Stankiewicz A, Kaczorowska K, Bugno R, Kozioł A, Paluchowska MH, Burnat G, Chruścicka B, Chorobik P, Brański P, Wierońska JM, Duszyńska B, Pilc A, Bojarski AJ. New 1,2,4-oxadiazole derivatives with positive mGlu 4 receptor modulation activity and antipsychotic-like properties. J Enzyme Inhib Med Chem 2021; 37:211-225. [PMID: 34894953 PMCID: PMC8667925 DOI: 10.1080/14756366.2021.1998022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Considering the allosteric regulation of mGlu receptors for potential therapeutic applications, we developed a group of 1,2,4-oxadiazole derivatives that displayed mGlu4 receptor positive allosteric modulatory activity (EC50 = 282–656 nM). Selectivity screening revealed that they were devoid of activity at mGlu1, mGlu2 and mGlu5 receptors, but modulated mGlu7 and mGlu8 receptors, thus were classified as group III-preferring mGlu receptor agents. None of the compounds was active towards hERG channels or in the mini-AMES test. The most potent in vitro mGlu4 PAM derivative 52 (N-(3-chloro-4-(5-(2-chlorophenyl)-1,2,4-oxadiazol-3-yl)phenyl)picolinamide) was readily absorbed after i.p. administration (male Albino Swiss mice) and reached a maximum brain concentration of 949.76 ng/mL. Five modulators (34, 37, 52, 60 and 62) demonstrated significant anxiolytic- and antipsychotic-like properties in the SIH and DOI-induced head twitch test, respectively. Promising data were obtained, especially for N-(4-(5-(2-chlorophenyl)-1,2,4-oxadiazol-3-yl)-3-methylphenyl)picolinamide (62), whose effects in the DOI-induced head twitch test were comparable to those of clozapine and better than those reported for the selective mGlu4 PAM ADX88178.
Collapse
Affiliation(s)
- Anna Stankiewicz
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Kaczorowska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ryszard Bugno
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Aneta Kozioł
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Maria H Paluchowska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Grzegorz Burnat
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Barbara Chruścicka
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Paulina Chorobik
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Brański
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Beata Duszyńska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
5
|
Facilitating mGluR4 activity reverses the long-term deleterious consequences of chronic morphine exposure in male mice. Neuropsychopharmacology 2021; 46:1373-1385. [PMID: 33349673 PMCID: PMC8136479 DOI: 10.1038/s41386-020-00927-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Understanding the neurobiological underpinnings of abstinence from drugs of abuse is critical to allow better recovery and ensure relapse prevention in addicted subjects. By comparing the long-term transcriptional consequences of morphine and cocaine exposure, we identified the metabotropic glutamate receptor subtype 4 (mGluR4) as a promising pharmacological target in morphine abstinence. We evaluated the behavioral and molecular effects of facilitating mGluR4 activity in abstinent mice. Transcriptional regulation of marker genes of medium spiny neurons (MSNs) allowed best discriminating between 4-week morphine and cocaine abstinence in the nucleus accumbens (NAc). Among these markers, Grm4, encoding mGluR4, displayed down-regulated expression in the caudate putamen and NAc of morphine, but not cocaine, abstinent mice. Chronic administration of the mGluR4 positive allosteric modulator (PAM) VU0155041 (2.5 and 5 mg/kg) rescued social behavior, normalized stereotypies and anxiety and blunted locomotor sensitization in morphine abstinent mice. This treatment improved social preference but increased stereotypies in cocaine abstinent mice. Finally, the beneficial behavioral effects of VU0155041 treatment in morphine abstinent mice were correlated with restored expression of key MSN and neural activity marker genes in the NAc. This study reports that chronic administration of the mGluR4 PAM VU0155041 relieves long-term deleterious consequences of morphine exposure. It illustrates the neurobiological differences between opiate and psychostimulant abstinence and points to pharmacological repression of excessive activity of D2-MSNs in the NAc as a promising therapeutic lever in drug addiction.
Collapse
|
6
|
Meta-analysis of cognitive and behavioral tests in leptin- and leptin receptor-deficient mice. Neurosci Res 2020; 170:217-235. [PMID: 33316303 DOI: 10.1016/j.neures.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 01/19/2023]
Abstract
Leptin is a hormone produced by adipocytes that regulates food intake and metabolism. Leptin-related gene-deficient mice, such as db/db and ob/ob mice, are widely used to study diabetes and its related diseases. However, broad effects of leptin appear to cause variability in behavioral test results. We performed a meta-analysis of major behavioral tests in db/db and ob/ob mice. These mice exhibited significant impairments in the Morris water maze, forced swim, novel object recognition, Y-maze, tail suspension, and light-dark box tests, whereas the elevated plus maze and open field tests did not reveal significant changes. We also performed correlation and regression analyses between the animals' performances and the experimental protocols and conditions. The memory-related tests were characterized by the correlations of their results with animal age, while the performances in the elevated plus-maze and forced swim tests were affected by the width of the devices used. In conclusion, db/db and ob/ob mice mainly exhibit memory deficits and depression-like behavior, although experimenters should be aware of animal age and device size in conducting experiments.
Collapse
|
7
|
Langer E, Einat H, Stukalin Y. Similarities and dissimilarities in the effects of benzodiazepines and specific serotonin reuptake inhibitors (SSRIs) in the defensive marble burying test: A systematic review and meta-analysis. Eur Neuropsychopharmacol 2020; 36:38-49. [PMID: 32456852 DOI: 10.1016/j.euroneuro.2020.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/04/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
One problem areas of animal models and tests for neuropsychiatric disorders is unclear reproducibility, including both internal and external validity. One way to examine external validity is with systematic reviews and meta-analyses, a standard practice in clinical research that is relatively neglected in preclinical research. Considering the need to evaluate the validity and reproducibility of frequently used animal models, this study presents a meta-analysis of the effects of prototypic benzodiazepines and specific serotonin reuptake inhibitors (SSRIs) in the mouse defensive marble burying test (MBT). These drug groups were selected because although they differ in their biological targets as well as in their clinical use, they are both commonly used for the treatment of anxiety disorders. A PubMed literature search was performed to identify studies that examined the effects of benzodiazepines (diazepam, alprazolam, chlordiazepoxide, clonazepam) or SSRIs (fluoxetine, citalopram, escitalopram, fluvoxamine, paroxetine) in the MBT in mice. For benzodiazepines, 73 experiments were included. Benzodiazepines effect size was 2.04 and Q statistics was 1959 with a significant correlation between dose and effect size (r = 0.31, p = 0.007). For SSRIs we identified 47 experiments. Effect size of SSRIs was 2.24 and Q statistics was 493.38. No correlation was found between dose and effect size (r = 0.23, p = 0.12). The current results support the external validity of the defensive marble burying test as a screening test for anxiolytic effects. However, these results indicate that significant attention should be given to the administration schedules of benzodiazepines and SSRIs.
Collapse
Affiliation(s)
- Erez Langer
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel
| | - Haim Einat
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel.
| | - Yelena Stukalin
- School of Behavioral Sciences, Tel Aviv-Yaffo Academic College, Tel-Aviv, Israel
| |
Collapse
|
8
|
Cieślik P, Domin H, Chocyk A, Gruca P, Litwa E, Płoska A, Radulska A, Pelikant-Małecka I, Brański P, Kalinowski L, Wierońska JM. Simultaneous activation of mGlu 2 and muscarinic receptors reverses MK-801-induced cognitive decline in rodents. Neuropharmacology 2019; 174:107866. [PMID: 31785263 DOI: 10.1016/j.neuropharm.2019.107866] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 10/09/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022]
Abstract
The activity of an allosteric agonist of muscarinic M1 receptor, VU0357017, and a positive allosteric modulator (PAM) of M5 receptor, VU0238429, were investigated alone or in combination with the mGlu2 receptor PAM, LY487379 using the following behavioural tests: prepulse inhibition (PPI), novel object recognition (NOR), and spatial delayed alternation (SDA). VU0357017 (10 and 20 mg/kg) and VU0238429 (5 and 10 mg/kg) reversed deficits in PPI while VU0238429 (2.5 and 5 mg/kg) was effective in SDA. The simultaneous administration of subeffective doses of M1 or M5 activators (5, 1, or 0.25 mg/kg) with LY487379 (0.5 mg/kg) induced the same effect as that observed for the active dose of each compound. Selective M1 or M5 receptor blockers antagonized the effect exerted by these combinations, and pharmacokinetic studies confirmed independent transport through the blood-brain barrier. The expression of both receptors (M1 and M5) was established in brain structures involved in cognition (neocortex, hippocampus, and entorhinal cortex) in both the rat and the mouse brains by immunofluorescence staining. Specifically, double neuronal staining of mGlu2-M1 and mGlu2-M5 receptors was observed in many areas of the rat brain, while the number of double-stained mGlu2-M1 receptors was moderate in the mouse brain with no mGlu2-M5 colocalization. Finally, the combined administration of subeffective doses of the compounds did not alter prolactin levels or motor coordination, in contrast to the compounds given alone at the highest dose or in combination with standard neuroleptics.
Collapse
Affiliation(s)
- Paulina Cieślik
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Helena Domin
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Piotr Gruca
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Ewa Litwa
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Dębinki 7, 80-211, Gdańsk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdańsk, Poland
| | - Adrianna Radulska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Dębinki 7, 80-211, Gdańsk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdańsk, Poland
| | - Iwona Pelikant-Małecka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Dębinki 7, 80-211, Gdańsk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdańsk, Poland
| | - Piotr Brański
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Dębinki 7, 80-211, Gdańsk, Poland; Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdańsk, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343, Kraków, 12 Smetna Street, Poland.
| |
Collapse
|
9
|
Fulton MG, Loch MT, Cuoco CA, Rodriguez AL, Days E, Vinson PN, Kozek KA, Weaver CD, Blobaum AL, Conn PJ, Niswender CM, Lindsley CW. Challenges in the Discovery and Optimization of mGlu 2/4 Heterodimer Positive Allosteric Modulators. LETT DRUG DES DISCOV 2019; 16:1387-1394. [PMID: 32201485 PMCID: PMC7059629 DOI: 10.2174/1570180815666181017131349] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 11/22/2022]
Abstract
Background: This article describes the challenges in the discovery and optimization of mGlu2/4 heterodimer Positive Allosteric Modulators (PAMs). Methods: Initial forays based on VU0155041, a PAM of both the mGlu4 homodimer and the mGlu2/4 heterodimer, led to flat, intractable SAR that precluded advancement. Screening of a collection of 1,152 FDA approved drugs led to the discovery that febuxostat, an approved xanthine oxidase inhibitor, was a moderately potent PAM of the mGlu2/4 heterodimer (EC50 = 3.4 μM), but was peripherally restricted (rat Kp = 0.03). Optimization of this hit led to PAMs with improved potency (EC50s <800 nM) and improved CNS penetration (rat Kp >2, an ~100-fold increase). Results: However, these new amide analogs of febuxostat proved to be either GIRK1/2 and GIRK1/4 activators (primary carboxamide congeners) or mGlu2 PAMs (secondary and tertiary amides) and not selective mGlu2/4 heterodimer PAMs. Conclusion: These results required the team to develop a new screening cascade paradigm, and exemplified the challenges in developing allosteric ligands for heterodimeric receptors.
Collapse
Affiliation(s)
- Mark Gallant Fulton
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew Thomas Loch
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Caroline Anne Cuoco
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Alice Lambert Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Emily Days
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Paige Newton Vinson
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Krystian Andrezej Kozek
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Charles David Weaver
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna Louise Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Peter Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Colleen Marie Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Craig William Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
10
|
Chaki S, Koike H, Fukumoto K. Targeting of Metabotropic Glutamate Receptors for the Development of Novel Antidepressants. CHRONIC STRESS 2019; 3:2470547019837712. [PMID: 32500107 PMCID: PMC7243201 DOI: 10.1177/2470547019837712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022]
Abstract
Since discovering that ketamine has robust antidepressant effects, the
glutamatergic system has been proposed as an attractive target for the
development of novel antidepressants. Among the glutamatergic system,
metabotropic glutamate (mGlu) receptors are of interest because mGlu receptors
play modulatory roles in glutamatergic transmission, consequently, agents acting
on mGlu receptors might not exert the adverse effects associated with ketamine.
mGlu receptors have eight subtypes that are classified into three groups, and
the roles of each mGlu receptor subtype in depression are being investigated. To
date, the potential use of mGlu5 receptor antagonists and mGlu2/3 receptor
antagonists as antidepressants has been actively investigated, and the
mechanisms underlying these antidepressant effects are being delineated.
Although the outcomes of clinical trials using an mGlu5 receptor negative
allosteric modulator and an mGlu2/3 receptor negative allosteric modulator have
not been encouraging, these trials have been inconclusive, and additional trials
using other compounds with more appropriate profiles are needed. In contrast,
the roles of group III mGlu receptors have not yet been fully elucidated because
of a lack of suitable pharmacological tools. Nonetheless, investigations of the
use of mGlu4 and mGlu7 receptors as drug targets for the development of
antidepressants have been ongoing, and some interesting evidence has been
obtained.
Collapse
|
11
|
Chen Q, Ho JD, Ashok S, Vargas MC, Wang J, Atwell S, Bures M, Schkeryantz JM, Monn JA, Hao J. Structural Basis for ( S)-3,4-Dicarboxyphenylglycine (DCPG) As a Potent and Subtype Selective Agonist of the mGlu 8 Receptor. J Med Chem 2018; 61:10040-10052. [PMID: 30365309 DOI: 10.1021/acs.jmedchem.8b01120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
( S)-3,4-Dicarboxyphenylglycine (DCPG) was first reported in 2001 as a potent orthosteric agonist with high subtype selectivity for the mGlu8 receptor, but the structural basis for its high selectivity is not well understood. We have solved a cocrystal structure of recombinant human mGlu8 amino terminal domain (ATD) protein bound to ( S)-DCPG, which possesses the largest lobe opening angle observed to date among known agonist-bound mGlu ATD crystal structures. The binding conformation of ( S)-DCPG observed in the crystal structure is significantly different from that in the homology model built from an l-glutamate-bound rat mGlu1 ATD crystal structure, which has a smaller lobe opening angle. This highlights the importance of considering various lobe opening angles when modeling mGlu ATD-ligand complex. New homology models of other mGlu receptors based on the ( S)-DCPG-bound mGlu8 ATD crystal structure were explored to rationalize ( S)-DCPG's high mGlu8 receptor subtype selectivity.
Collapse
|
12
|
Cieślik P, Woźniak M, Kaczorowska K, Brański P, Burnat G, Chocyk A, Bobula B, Gruca P, Litwa E, Pałucha-Poniewiera A, Wąsik A, Pilc A, Wierońska J. Negative Allosteric Modulators of mGlu 7 Receptor as Putative Antipsychotic Drugs. Front Mol Neurosci 2018; 11:316. [PMID: 30294258 PMCID: PMC6158327 DOI: 10.3389/fnmol.2018.00316] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/16/2018] [Indexed: 01/08/2023] Open
Abstract
The data concerning antipsychotic-like activity of negative allosteric modulators (NAMs)/antagonists of mGlu7 receptors are limited. The only available ligands for this receptor are MMPIP and ADX71743. In the present studies, we used stable cell line expressing mGlu7 receptor and it was shown that both compounds dose-dependently potentiated forskolin elevated cAMP concentration in the T-REx 293 cells, showing their inverse agonist properties. Subsequently, pharmacokinetic studies were performed. Both compounds were given intraperitoneally (i.p.) at the dose of 10 mg/kg and reached Cmax 0.25-0.5 h after administration, and then they declined rapidly, ADX71743 being almost undetectable 2 h after administration, while the concentration of MMPIP was still observed, suggesting that the concentration of MMPIP was more stable. Finally, we investigated the role of both mGlu7 receptor NAMs in animal models of schizophrenia. Behavioral tests commonly used in antipsychotic drug discovery were conducted. Both tested compounds dose-dependently inhibited MK-801-induced hyperactivity (MMPIP at 15 mg/kg; ADX at 5 and 15 mg/kg) and DOI-induced head twitches (MMPIP at 5, 10, 15 mg/kg; ADX at 2.5, 5, 10 mg/kg). Moreover, the same effects were noticed in novel object recognition test, where MMPIP (5, 10, 15 mg/kg) and ADX71743 (1, 5, 15 mg/kg) reversed MK-801-induced disturbances. In the social interaction test, antipsychotic activity was observed only for ADX71743 (5, 15 mg/kg). ADX71743 at the dose 2.5 mg/kg reversed MK-801-induced disruption in prepulse inhibition while MMPIP at 10 mg/kg reversed MK-801-induced disruption in spatial delayed alternation. The present studies showed that mGlu7 receptor may be considered as a putative target for antipsychotic drugs, though more studies are needed due to limited number of available ligands.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joanna Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
13
|
Wierońska JM, Pilc A. Depression and schizophrenia viewed from the perspective of amino acidergic neurotransmission: Antipodes of psychiatric disorders. Pharmacol Ther 2018; 193:75-82. [PMID: 30149102 DOI: 10.1016/j.pharmthera.2018.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Depression and schizophrenia are burdensome, costly serious and disabling mental disorders. Moreover the existing treatments are not satisfactory. As amino-acidergic (AA) neurotransmitters built a vast majority of brain neurons, in this article we plan to focus on drugs influencing AA neurotransmission in both diseases: we will discuss several facts concerning glutamatergic and GABA-ergic neurotransmission in these diseases, based mainly on preclinical experiments that used stimulators and/or blockers of both neurotransmitter systems. In general a picture emerges showing, that treatments that increase excitatory effects (with either antagonists or agonists) tend to evoke antidepressant effects, while treatments that increase inhibitory effects tend to display antipsychotic properties. Moreover, it seems that the antidepressant activity of a given compound excludes it as a potential antipsychotic and vice versa.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland
| | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland.
| |
Collapse
|
14
|
Zussy C, Gómez-Santacana X, Rovira X, De Bundel D, Ferrazzo S, Bosch D, Asede D, Malhaire F, Acher F, Giraldo J, Valjent E, Ehrlich I, Ferraguti F, Pin JP, Llebaria A, Goudet C. Dynamic modulation of inflammatory pain-related affective and sensory symptoms by optical control of amygdala metabotropic glutamate receptor 4. Mol Psychiatry 2018; 23:509-520. [PMID: 27994221 DOI: 10.1038/mp.2016.223] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 10/06/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022]
Abstract
Contrary to acute pain, chronic pain does not serve as a warning signal and must be considered as a disease per se. This pathology presents a sensory and psychological dimension at the origin of affective and cognitive disorders. Being largely refractory to current pharmacotherapies, identification of endogenous systems involved in persistent and chronic pain is crucial. The amygdala is a key brain region linking pain sensation with negative emotions. Here, we show that activation of a specific intrinsic neuromodulatory system within the amygdala associated with type 4 metabotropic glutamate receptors (mGlu4) abolishes sensory and affective symptoms of persistent pain such as hypersensitivity to pain, anxiety- and depression-related behaviors, and fear extinction impairment. Interestingly, neuroanatomical and synaptic analysis of the amygdala circuitry suggests that the effects of mGlu4 activation occur outside the central nucleus via modulation of multisensory thalamic inputs to lateral amygdala principal neurons and dorso-medial intercalated cells. Furthermore, we developed optogluram, a small diffusible photoswitchable positive allosteric modulator of mGlu4. This ligand allows the control of endogenous mGlu4 activity with light. Using this photopharmacological approach, we rapidly and reversibly inhibited behavioral symptoms associated with persistent pain through optical control of optogluram in the amygdala of freely behaving animals. Altogether, our data identify amygdala mGlu4 signaling as a mechanism that bypasses central sensitization processes to dynamically modulate persistent pain symptoms. Our findings help to define novel and more precise therapeutic interventions for chronic pain, and exemplify the potential of optopharmacology to study the dynamic activity of endogenous neuromodulatory mechanisms in vivo.
Collapse
Affiliation(s)
- C Zussy
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - X Gómez-Santacana
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.,Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - X Rovira
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - D De Bundel
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - S Ferrazzo
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - D Bosch
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - D Asede
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - F Malhaire
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - F Acher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - J Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Network Biomedical Research Center on Mental Health (CIBERSAM), Madrid, Spain
| | - E Valjent
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - I Ehrlich
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - F Ferraguti
- Department of Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - J-P Pin
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| | - A Llebaria
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - C Goudet
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, Université de Montpellier, Montpellier, France.,INSERM, U1191, Montpellier, France
| |
Collapse
|
15
|
Ferraguti F. Metabotropic glutamate receptors as targets for novel anxiolytics. Curr Opin Pharmacol 2018; 38:37-42. [PMID: 29494817 DOI: 10.1016/j.coph.2018.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
Anxiety disorders are highly prevalent psychiatric illnesses posing an important social and economic burden. Their current pharmacotherapy shows short term efficacy, though nearly one third of patients do not achieve sustained remission. There is, therefore, a strong medical need for new therapeutic agents acting through novel mechanisms of action. Considerable work has focused on metabotropic glutamate (mGlu) receptors as potential targets for novel anxiolytics. Ligands acting at mGlu receptors showed promising results in preclinical studies, whereas their efficacy was dubious in clinical trials. Recent preclinical and clinical studies have opened new prospects for targeting mGlu receptors to treat anxiety disorders. This review provides an outlook on these progresses.
Collapse
Affiliation(s)
- Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1A, 6020 Innsbruck, Austria.
| |
Collapse
|
16
|
Chaki S, Fukumoto K. mGlu receptors as potential targets for novel antidepressants. Curr Opin Pharmacol 2018; 38:24-30. [DOI: 10.1016/j.coph.2018.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/09/2018] [Indexed: 12/21/2022]
|
17
|
Dadkhah T, Rahimi-Aliabadi S, Jamshidi J, Ghaedi H, Taghavi S, Shokraeian P, Akhavan-Niaki H, Tafakhori A, Ohadi M, Darvish H. A genetic variant in miRNA binding site of glutamate receptor 4, metabotropic (GRM4) is associated with increased risk of major depressive disorder. J Affect Disord 2017; 208:218-222. [PMID: 27792966 DOI: 10.1016/j.jad.2016.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 09/29/2016] [Accepted: 10/16/2016] [Indexed: 01/06/2023]
Abstract
BACKGROUND Glutamate receptor 4, metabotropic (GRM4) expression is increased in the brain of patients with depression. The poorly conserved miR-1202 is downregulated in depression and is negatively correlated with GRM4. A variation located at the 3' UTR of the GRM4 gene may influence the interaction between miR-1202 and GRM4. The aim of this study was to determine the possible association between GRM4 3' UTR variant (rs2229901) and major depressive disorder (MDD). METHODS A total of 500 subjects comprising 250 patients with MDD and 250 healthy controls were included in our study. The single nucleotide polymorphism rs2229901 was genotyped using PCR-RFLP method. Allele and genotype frequencies were compared between the two groups using chi-square test and logistic regression models. The impact of rs2229901 on GRM4/miR-1202 hybrid stability and local GRM4-3' UTR secondary structure were assessed using RNAsnp program. RESULTS Genotype and allele frequency of rs2229901were significantly different in patients with MDD comparing to the control group (p=0.018 and p=0.007, respectively). The G-allele was more prevalent among patients with MDD. The rs2229901 variant was predicted to be structure-disruptive. LIMITATIONS The relatively small sample size and lack of functional experiments are the major limitations of this study. CONCLUSION Our results suggest that rs2229901 is associated with MDD risk. This variant probably impacts the interaction between GRM4 and miR-1202. Functional studies are needed to clarify the possible mechanisms by which rs2229901 influences MDD risk.
Collapse
Affiliation(s)
- Tahereh Dadkhah
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Simin Rahimi-Aliabadi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Jamshidi
- Noncommunicable Diseases Research Center, Fasa University ofMedical Sciences, Fasa, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghyegh Taghavi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Haleh Akhavan-Niaki
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Imam Khomeini Hospital and Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Ohadi
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hossein Darvish
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Abstract
Contemporary biological psychiatry uses experimental animal models to increase our understanding of affective disorder pathogenesis. Modern anxiolytic drug discovery mainly targets specific pathways and molecular determinants within a single phenotypic domain. However, greater understanding of the mechanisms of action is possible through animal models. Primarily developed with rats, animal models in anxiety have been adapted with mixed success for mice, easy-to-use mammals with better genetic possibilities than rats. In this review, we focus on the three most common animal models of anxiety in mice used in the screening of anxiolytics. Both conditioned and unconditioned models are described, in order to represent all types of animal models of anxiety. Behavioral studies require careful attention to variable parameters linked to environment, handling, or paradigms; this is also discussed. Finally, we focus on the consequences of re-exposure to the apparatus. Test-retest procedures can provide new answers, but should be intensively studied in order to revalidate the entire paradigm as an animal model of anxiety.
Collapse
|
19
|
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, Conn PJ. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev 2016; 116:6707-41. [PMID: 26882314 PMCID: PMC4988345 DOI: 10.1021/acs.chemrev.5b00656] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.
Collapse
Affiliation(s)
- Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Corey R. Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville VIC 3052, Australia
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
20
|
Peterlik D, Flor PJ, Uschold-Schmidt N. The Emerging Role of Metabotropic Glutamate Receptors in the Pathophysiology of Chronic Stress-Related Disorders. Curr Neuropharmacol 2016; 14:514-39. [PMID: 27296643 PMCID: PMC4983752 DOI: 10.2174/1570159x13666150515234920] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/04/2015] [Accepted: 05/12/2015] [Indexed: 12/28/2022] Open
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.
Collapse
Affiliation(s)
| | - Peter J Flor
- Faculty of Biology and Preclinical Medicine, University of Regensburg, D-93053 Regensburg, Germany.
| | - Nicole Uschold-Schmidt
- Faculty of Biology and Preclinical Medicine, University of Regensburg, D-93053 Regensburg, Germany.
| |
Collapse
|
21
|
Potential of Glutamate-Based Drug Discovery for Next Generation Antidepressants. Pharmaceuticals (Basel) 2015; 8:590-606. [PMID: 26393618 PMCID: PMC4588184 DOI: 10.3390/ph8030590] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/27/2015] [Accepted: 09/03/2015] [Indexed: 01/17/2023] Open
Abstract
Recently, ketamine has been demonstrated to exert rapid-acting antidepressant effects in patients with depression, including those with treatment-resistant depression, and this discovery has been regarded as the most significant advance in drug development for the treatment of depression in over 50 years. To overcome unwanted side effects of ketamine, numerous approaches targeting glutamatergic systems have been vigorously investigated. For example, among agents targeting the NMDA receptor, the efficacies of selective GluN2B receptor antagonists and a low-trapping antagonist, as well as glycine site modulators such as GLYX-13 and sarcosine have been demonstrated clinically. Moreover, agents acting on metabotropic glutamate receptors, such as mGlu2/3 and mGlu5 receptors, have been proposed as useful approaches to mimicking the antidepressant effects of ketamine. Neural and synaptic mechanisms mediated through the antidepressant effects of ketamine have been being delineated, most of which indicate that ketamine improves abnormalities in synaptic transmission and connectivity observed in depressive states via the AMPA receptor and brain-derived neurotrophic factor-dependent mechanisms. Interestingly, some of the above agents may share some neural and synaptic mechanisms with ketamine. These studies should provide important insights for the development of superior pharmacotherapies for depression with more potent and faster onsets of actions.
Collapse
|
22
|
Podkowa K, Rzeźniczek S, Marciniak M, Acher F, Pilc A, Pałucha-Poniewiera A. A novel mGlu4 selective agonist LSP4-2022 increases behavioral despair in mouse models of antidepressant action. Neuropharmacology 2015; 97:338-45. [PMID: 26074092 DOI: 10.1016/j.neuropharm.2015.05.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/26/2015] [Accepted: 05/30/2015] [Indexed: 11/16/2022]
Abstract
Numerous data have indicated that metabotropic glutamate (mGlu) receptor ligands may be potentially useful as novel antidepressant drugs (ADs). The Group III mGlu receptor has not been explored much because of the limited access to selective ligands, but some behavioral studies have indicated that modulating group III mGlu receptors may result in benefits for the therapy of depression. Here, we investigated the potential antidepressant-like effects of a new mGlu4 selective orthosteric agonist, LSP4-2022. We found that the drug induced pro-depressant effects in the tail suspension test (TST) and the forced swim test (FST) in mice at doses that did not change the locomotor activity of the animals. Additional experiments that used knock-out (KO) mice and aimed to verify the selectivity of LSP4-2022 revealed that the drug induced strong pro-depressant-like effects in mGlu7 KO mice but did not affect the behavior of mGlu4 KO mice in the TST, suggesting that the activation of the mGlu4 receptor plays a role in producing the pro-depressant activity of the tested drug. The results of our study indicate that the inhibition rather than activation of mGlu4 receptors might induce antidepressant effects, but this hypothesis should be verified using a selective mGlu4 receptor antagonist, which is currently not available.
Collapse
Affiliation(s)
- Karolina Podkowa
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Szymon Rzeźniczek
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Marcin Marciniak
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland
| | - Francine Acher
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, UMR8601-CNRS, Paris Descartes University, Sorbonne Paris Cite,45, rue des Saints-Peres, 75270 Paris Cedex 06, France
| | - Andrzej Pilc
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland; Jagiellonian University Medical College, Department of Drug Management, Faculty of Health Sciences, Grzegórzecka 20, 31-531 Kraków, Poland
| | - Agnieszka Pałucha-Poniewiera
- Institute of Pharmacology Polish Academy of Sciences, Department of Neurobiology, Smętna 12, 31-343 Kraków, Poland.
| |
Collapse
|
23
|
Moloney RD, Golubeva AV, O'Connor RM, Kalinichev M, Dinan TG, Cryan JF. Negative allosteric modulation of the mGlu7 receptor reduces visceral hypersensitivity in a stress-sensitive rat strain. Neurobiol Stress 2015; 2:28-33. [PMID: 26844237 PMCID: PMC4721404 DOI: 10.1016/j.ynstr.2015.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 03/15/2015] [Accepted: 04/03/2015] [Indexed: 11/19/2022] Open
Abstract
Glutamate, the main excitatory neurotransmitter in the central nervous system, exerts its effect through ionotropic and metabotropic receptors. Of these, group III mGlu receptors (mGlu 4, 6, 7, 8) are among the least studied due to a lack of pharmacological tools. mGlu7 receptors, the most highly conserved isoform, are abundantly distributed in the brain, especially in regions, such as the amygdala, known to be crucial for the emotional processing of painful stimuli. Visceral hypersensitivity is a poorly understood phenomenon manifesting as an increased sensitivity to visceral stimuli. Glutamate has long been associated with somatic pain processing leading us to postulate that crossover may exist between these two modalities. Moreover, stress has been shown to exacerbate visceral pain. ADX71743 is a novel, centrally penetrant, negative allosteric modulator of mGlu7 receptors. Thus, we used this tool to explore the possible involvement of this receptor in the mediation of visceral pain in a stress-sensitive model of visceral hypersensitivity, namely the Wistar Kyoto (WKY) rat. ADX71743 reduced visceral hypersensitivity in the WKY rat as exhibited by increased visceral sensitivity threshold with concomitant reductions in total number of pain behaviours. Moreover, AD71743 increased total distance and distance travelled in the inner zone of the open field. These findings show, for what is to our knowledge, the first time, that mGlu7 receptor signalling plays a role in visceral pain processing. Thus, negative modulation of the mGlu7 receptor may be a plausible target for the amelioration of stress-induced visceral pain where there is a large unmet medical need.
Collapse
Affiliation(s)
- Rachel D. Moloney
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Psychiatry, University College Cork, Ireland
| | - Anna V. Golubeva
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
| | | | | | - Timothy G. Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Psychiatry, University College Cork, Ireland
| | - John F. Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- Corresponding author. Dept Anatomy & Neuroscience, Room 386, Western Gateway Building, University College Cork, Western Rd., Cork, Ireland.
| |
Collapse
|
24
|
Viral vector mediated expression of mutant huntingtin in the dorsal raphe produces disease-related neuropathology but not depressive-like behaviors in wildtype mice. Brain Res 2015; 1608:177-90. [PMID: 25732261 DOI: 10.1016/j.brainres.2015.02.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 02/10/2015] [Accepted: 02/14/2015] [Indexed: 12/13/2022]
Abstract
Huntington׳s disease (HD) is a neurodegenerative disorder caused by a mutation in the HTT gene (mHTT) encoding the protein huntingtin. An expansion in the gene׳s CAG repeat length renders a misfolded, dysfunctional protein with an abnormally long glutamine (Q) stretch at the N terminus that often incorporates into inclusion bodies and leads to neurodegeneration in many regions of the brain. HD is characterized by motor and cognitive decline as well as mood disorders, with depression being particularly common. Approximately 40% of the HD population suffers from depressive symptoms. Because these symptoms often manifest a decade or more prior to the knowledge that the person is at risk for the disease, a portion of the early depression in HD appears to be a consequence of the pathology arising from expression of the mutant gene. While the depression in HD patients is often treated with serotonin agonists, there is scant experimental evidence that the depression in HD responds well to these serotonin treatments or in a similar manner to how non-HD depression tends to respond. Additionally, at very early sub-threshold depression levels, abnormal changes in several neuronal populations are already detectable in HD patients, suggesting that a variety of brain structures may be involved. Taken together, the serotonin system is a viable candidate. However, at present there is limited evidence of the precise nuclei or circuits that play a role in HD depression. With this in mind, the current study was designed to control for the widespread brain neuropathology that occurs in HD and in transgenic mouse models of HD and focuses specifically on the influence of the midbrain dorsal raphe nucleus (DRN). The DRN provides the majority of the serotonin to the forebrain and exhibits cell loss in non-HD depression. Therefore, we employed a viral vector delivery system to investigate whether the over-expression of mHTT in the DRN׳s ventral sub-nuclei alone is sufficient to produce depressive-like behaviors. Wildtype mice were injected with an adeno-associated virus (AAV2/1) encoding HTT containing either a pathogenic (N171-82Q) or control (N171-16Q) CAG repeat length into the ventral DRN and depressive-like behaviors and motor behaviors were assessed for 12 weeks post-surgery. Quantitative PCR and immunohistochemistry (IHC) verified positive transduction in the ventral aspects of the DRN, including the ventral sub-nucleus (DRv) and interfascicular sub-nucleus (DRif). IHC demonstrated microgliosis in and around the injection site and mHTT-positive inclusions in serotonin-producing neurons and a small percentage of astrocytes in animals injected with N171-82Q compared to controls. Moreover, N171-82Q injected mice showed a 75% reduction in cells that stained positive for the serotonin synthesis enzyme, tryptophan hydroxylase-2 (TPH2) compared to controls (p<0.05). Despite mHTT-mediated pathology in the DRv and DRif, no significant changes in depressive-like behavior were detected. Consequently, we conclude that 12 weeks of N171-82Q expression in the ventral sub-nuclei of the DRN of wildtype mice causes characteristic disease-related cellular neuropathology but is not sufficient to elicit depressive-like behaviors. Ongoing studies are investigating whether a larger injection volume that transfects a larger percentage of the DRN and/or a longer time course of mHTT expression might elicit depressive-like behaviors. Moreover, mHTT expression in other regions of the brain, such as the hippocampal dentate gyrus and/or the frontal cortex might be necessary to elicit HD depression. Together, these results may prove helpful in addressing which therapeutic and/or pharmacological strategies might be most efficacious when treating depressive symptomology in patients suffering from HD.
Collapse
|
25
|
Jalan-Sakrikar N, Field JR, Klar R, Mattmann M, Gregory KJ, Zamorano R, Engers DW, Bollinger SR, Weaver CD, Days EL, Lewis LM, Utley TJ, Hurtado M, Rigault D, Acher F, Walker AG, Melancon BJ, Wood MR, Lindsley C, Conn PJ, Xiang Z, Hopkins CR, Niswender CM. Identification of positive allosteric modulators VU0155094 (ML397) and VU0422288 (ML396) reveals new insights into the biology of metabotropic glutamate receptor 7. ACS Chem Neurosci 2014; 5:1221-37. [PMID: 25225882 PMCID: PMC4306484 DOI: 10.1021/cn500153z] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/15/2014] [Indexed: 11/29/2022] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is a member of the group III mGlu receptors (mGlus), encompassed by mGlu4, mGlu6, mGlu7, and mGlu8. mGlu7 is highly expressed in the presynaptic active zones of both excitatory and inhibitory synapses, and activation of the receptor regulates the release of both glutamate and GABA. mGlu7 is thought to be a relevant therapeutic target for a number of neurological and psychiatric disorders, and polymorphisms in the GRM7 gene have been linked to autism, depression, ADHD, and schizophrenia. Here we report two new pan-group III mGlu positive allosteric modulators, VU0155094 and VU0422288, which show differential activity at the various group III mGlus. Additionally, both compounds show probe dependence when assessed in the presence of distinct orthosteric agonists. By pairing studies of these nonselective compounds with a synapse in the hippocampus that expresses only mGlu7, we have validated activity of these compounds in a native tissue setting. These studies provide proof-of-concept evidence that mGlu7 activity can be modulated by positive allosteric modulation, paving the way for future therapeutics development.
Collapse
Affiliation(s)
- Nidhi Jalan-Sakrikar
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Julie R. Field
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Rebecca Klar
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Margrith
E. Mattmann
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Karen J. Gregory
- Drug
Discovery Biology, Monash Institute of Pharmaceutical
Sciences, Parkville, VIC 3052, Australia
| | - Rocio Zamorano
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Darren W. Engers
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Sean R. Bollinger
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - C. David Weaver
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Emily L. Days
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - L. Michelle Lewis
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Thomas J. Utley
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Miguel Hurtado
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | | | | | - Adam G. Walker
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Bruce J. Melancon
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Michael R. Wood
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Craig
W. Lindsley
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - P. Jeffrey Conn
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Zixiu Xiang
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Corey R. Hopkins
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Colleen M. Niswender
- Department of Pharmacology and Vanderbilt Center
for Neuroscience Drug Discovery, Department of Pharmacology and
Vanderbilt Institute of Chemical Biology, and Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37212, United States
| |
Collapse
|
26
|
Rovira X, Harrak Y, Trapero A, González-Bulnes P, Malhaire F, Pin JP, Goudet C, Giraldo J, Llebaria A. Exploring the active conformation of cyclohexane carboxylate positive allosteric modulators of the type 4 metabotropic glutamate receptor. ChemMedChem 2014; 9:2685-98. [PMID: 25196639 DOI: 10.1002/cmdc.201402190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Indexed: 11/07/2022]
Abstract
The active conformation of a family of metabotropic glutamate receptor subtype 4 (mGlu4 ) positive allosteric modulators (PAMs) with the cyclohexane 1,2-dicarboxylic scaffold present in cis-2-(3,5-dichlorophenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041) was investigated by testing structurally similar six-membered ring compounds that have a locked conformation. The norbornane and cyclohexane molecules designed as mGlu4 conformational probes and the enantiomers of the trans diastereomer were computationally characterized and tested in mGlu4 pharmacological assays. The results support a VU0155041 active conformation, with the chair cyclohexane having the aromatic amide substituent in an axial position and the carboxylate in an equatorial position. Moreover, the receptor displays enantiomeric discrimination of the chiral PAMs. The constructed pharmacophore characterized a highly constrained mGlu4 allosteric binding site, thus providing a step forward in structure-based drug design for mGlu4 PAMs.
Collapse
Affiliation(s)
- Xavier Rovira
- Laboratory of Molecular Neuropharmacology & Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Universitat Autònoma de Barcelona, 08193 Bellaterra (Spain); Institut de Génomique Fonctionnelle, CNRS UMR5203, Université de Montpellier; U661, INSERM, 141 Rue de la Cardonille, 34094 Montpellier (France)
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kalinichev M, Le Poul E, Boléa C, Girard F, Campo B, Fonsi M, Royer-Urios I, Browne SE, Uslaner JM, Davis MJ, Raber J, Duvoisin R, Bate ST, Reynolds IJ, Poli S, Celanire S. Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. J Pharmacol Exp Ther 2014; 350:495-505. [PMID: 24947466 PMCID: PMC4152882 DOI: 10.1124/jpet.114.214437] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/19/2014] [Indexed: 11/22/2022] Open
Abstract
There is growing evidence that activation of metabotropic glutamate receptor 4 (mGlu4) leads to anxiolytic- and antipsychotic-like efficacy in rodent models, yet its relevance to depression-like reactivity remains unclear. Here, we present the pharmacological evaluation of ADX88178 [5-methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine], a novel potent, selective, and brain-penetrant positive allosteric modulator of the mGlu4 receptor in rodent models of anxiety, obsessive compulsive disorder (OCD), fear, depression, and psychosis. ADX88178 dose-dependently reduced the number of buried marbles in the marble burying test and increased open-arm exploration in the elevated plus maze (EPM) test, indicative of anxiolytic-like efficacy. Target specificity of the effect in the EPM test was confirmed using male and female mGlu4 receptor knockout mice. In mice, ADX88178 reduced the likelihood of conditioned freezing in the acquisition phase of the fear conditioning test, yet had no carryover effect in the expression phase. Also, ADX88178 dose-dependently reduced duration of immobility in the forced swim test, indicative of antidepressant-like efficacy. ADX88178 reduced DOI (2,5-dimethoxy-4-iodoamphetamine)-mediated head twitches (albeit with no dose-dependency), and MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine]-induced locomotor hyperactivity in mice, but was inactive in the conditioned avoidance response test in rats. The compound showed good specificity as it had no effect on locomotor activity in mice and rats at efficacious doses. Thus, allosteric activation of mGlu4 receptors can be a promising new therapeutic approach for treatment of anxiety, OCD, fear-related disorders, and psychosis.
Collapse
Affiliation(s)
- Mikhail Kalinichev
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Emmanuel Le Poul
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Christelle Boléa
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Françoise Girard
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Brice Campo
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Massimiliano Fonsi
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Isabelle Royer-Urios
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Susan E Browne
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Jason M Uslaner
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Matthew J Davis
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Jacob Raber
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Robert Duvoisin
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Simon T Bate
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Ian J Reynolds
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Sonia Poli
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| | - Sylvain Celanire
- Addex Therapeutics, Plan-les-Ouates, Geneva, Switzerland (M.K., E.L.P., C.B., F.G., B.C., M.F., I.R.-U., S.P., S.C.); Merck Research Laboratories, West Point, Pennsylvania (S.E.B., J.M.U., M.J.D., I.J.R.); Oregon Health & Science University, Portland, Oregon (M.J.D., J.R., R.D.); and Huntingdon Life Sciences Ltd., Huntingdon Research Centre, Huntingdon, United Kingdom (S.T.B.)
| |
Collapse
|
28
|
Autistic-like syndrome in mu opioid receptor null mice is relieved by facilitated mGluR4 activity. Neuropsychopharmacology 2014; 39:2049-60. [PMID: 24619243 PMCID: PMC4104328 DOI: 10.1038/npp.2014.59] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 02/28/2014] [Accepted: 02/28/2014] [Indexed: 02/03/2023]
Abstract
The etiology of Autism Spectrum Disorders (ASDs) remains largely unknown. Identifying vulnerability genes for autism represents a major challenge in the field and allows the development of animal models for translational research. Mice lacking the mu opioid receptor gene (Oprm1(-/-)) were recently proposed as a monogenic mouse model of autism, based on severe deficits in social behavior and communication skills. We confirm this hypothesis by showing that adult Oprm1(-/-) animals recapitulate core and multiple comorbid behavioral symptoms of autism and also display anatomical, neurochemical, and genetic landmarks of the disease. Chronic facilitation of mGluR4 signaling, which we identified as a novel pharmacological target in ASDs in these mice, was more efficient in alleviating behavioral deficits than the reference molecule risperidone. Altogether, our data provide first evidence that disrupted mu opioid receptor signaling is sufficient to trigger a comprehensive autistic syndrome, maybe through blunted social reward processes, and this mouse model opens promising avenues for therapeutic innovation.
Collapse
|
29
|
Pomierny-Chamioło L, Rup K, Pomierny B, Niedzielska E, Kalivas PW, Filip M. Metabotropic glutamatergic receptors and their ligands in drug addiction. Pharmacol Ther 2014; 142:281-305. [DOI: 10.1016/j.pharmthera.2013.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023]
|
30
|
Gubellini P, Melon C, Dale E, Doller D, Kerkerian-Le Goff L. Distinct effects of mGlu4 receptor positive allosteric modulators at corticostriatal vs. striatopallidal synapses may differentially contribute to their antiparkinsonian action. Neuropharmacology 2014; 85:166-77. [PMID: 24866785 DOI: 10.1016/j.neuropharm.2014.05.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/21/2014] [Accepted: 05/15/2014] [Indexed: 12/20/2022]
Abstract
Metabotropic glutamate 4 (mGlu4) receptor is a promising target for the treatment of motor deficits in Parkinson's disease (PD). This is due in part to its localization at key basal ganglia (BG) synapses that become hyperactive in this pathology, particularly striatopallidal synapses. In this context, mGlu4 receptor activation using either orthosteric agonists or positive allosteric modulators (PAMs) improves motor symptoms in rodent PD models in certain conditions. However, literature data show that mGlu4 receptor PAMs have no effect at striatopallidal GABAergic synapses (unless combined with an orthosteric agonist) and on the firing activity of pallidal neurons, and fail to provide significant motor improvement in relevant PD models. This questions the mechanistic hypothesis that mGlu4 receptor PAMs should act at striatopallidal synapses to alleviate PD motor symptoms. To shed light on this issue, we performed brain slice electrophysiology experiments. We show that Lu AF21934, an mGlu4 PAM small-molecule probe-compound, was ineffective at striatopallidal synapses at all concentrations tested, while it significantly inhibited corticostriatal synaptic transmission. Similarly, Lu AF21934 did not affect electrophysiology readouts at striatopallidal synapses in the presence of haloperidol or in 6-hydroxydopamine-lesioned rats. Interestingly, co-application of Lu AF21934 with a glutamate transporter inhibitor revealed a significant inhibitory action at striatopallidal synapses. Possibly, this effect could rely on increased level/permanence of glutamate in the synaptic cleft. Such differential efficacy of mGlu4 receptor PAMs at corticostriatal vs. striatopallidal synapses raises several issues regarding the synaptic target(s) of these drugs in the BG, and challenges the mechanisms by which they alleviate motor deficits in experimental PD models.
Collapse
Affiliation(s)
- Paolo Gubellini
- Aix-Marseille Université, CNRS, IBDM UMR7288, 13009 Marseille, France.
| | - Christophe Melon
- Aix-Marseille Université, CNRS, IBDM UMR7288, 13009 Marseille, France
| | - Elena Dale
- Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | - Dario Doller
- Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | | |
Collapse
|
31
|
Ossowska K, Wardas J, Berghauzen-Maciejewska K, Głowacka U, Kuter K, Pilc A, Zorn SH, Doller D. Lu AF21934, a positive allosteric modulator of mGlu4 receptors, reduces the harmaline-induced hyperactivity but not tremor in rats. Neuropharmacology 2014; 83:28-35. [PMID: 24726309 DOI: 10.1016/j.neuropharm.2014.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/18/2014] [Accepted: 03/31/2014] [Indexed: 12/19/2022]
Abstract
Harmaline induces tremor in animals resembling essential tremor which has been suggested to result from activation of the glutamatergic olivo-cerebellar projection. The aim of the present study was to examine the effects of systemic administration of Lu AF21934, a brain-penetrating positive allosteric modulator of the metabotropic glutamate receptor 4 (mGlu4), on the harmaline-induced tremor and other forms of motor activity in rats using fully automated Force Plate Actimeters. The influence of harmaline on the mGlu4 mRNA expression in the cerebellum and inferior olive was analysed by in situ hybridization. Harmaline at a dose of 15 mg/kg (ip) triggered tremor which was manifested by an increase in the power within 9-15 Hz band and in the tremor index (a difference in power between bands 9-15 Hz and 0-8 Hz). Harmaline induced a biphasic effect on mobility, initially inhibiting the exploratory locomotor activity of rats (0-30 min after administration), followed by an increase in their basic activity. Lu AF21934 (0.5-5 mg/kg sc) did not influence tremor but at doses of 0.5 and 2.5 mg/kg reversed harmaline-induced hyperactivity. MGlu4 mRNA expression was high in the cerebellar cortex and low in the inferior olive. Repeated harmaline (15 mg/kg ip once a day for 5 days] decreased mGlu4 mRNA in the cerebellum and inferior olive. The present study indicates that the mGlu4 stimulation counteracts hyperactivity induced by harmaline which suggests the involvement of cerebellar glutamatergic transmission in this process. In contrast, neuronal mechanisms involved in tremor seem to be insensitive to the stimulation of mGlu4.
Collapse
Affiliation(s)
- Krystyna Ossowska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland.
| | - Jadwiga Wardas
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Klemencja Berghauzen-Maciejewska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Urszula Głowacka
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Katarzyna Kuter
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 12, Smętna St., 31-343 Kraków, Poland
| | - Stevin H Zorn
- Discovery Chemistry & DMPK, Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| | - Dario Doller
- Discovery Chemistry & DMPK, Lundbeck Research USA, 215 College Road, Paramus, NJ 07652, USA
| |
Collapse
|
32
|
Zaniewska M, Przegaliński E, Filip M, Pilc A, Doller D. Inhibitory actions of mGlu4 receptor ligands on cocaine-, but not nicotine-, induced sensitizing and conditioning locomotor responses in rats. Pharmacol Rep 2014; 66:205-11. [PMID: 24911071 DOI: 10.1016/j.pharep.2013.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Male Wistar rats were used to verify the hypothesis that metabotropic glutamate 4 (mGlu4) receptor ligands may modulate the locomotor effects evoked by cocaine or nicotine. METHODS The preferential mGlu4 receptor orthosteric agonist (2S)-2-amino-4-[hydroxy[hydroxy(4-hydroxy-3-methoxy-5-nitrophenyl)methyl]phosphoryl]butanoic acid (LSP1-2111) and the mGlu4 receptor positive allosteric modulator (+)-cis-N(1)-(3,4-dichlorophenyl)cyclohexane-1,2-dicarboxamide (Lu AF21934) were used in the study. Rats were given repeated pairings of a test environment with cocaine (10mg/kg), nicotine (0.4 mg/kg) or the respective vehicles for 5 days. On day 10, animals were challenged with cocaine (10mg/kg, cocaine sensitization), nicotine (0.4 mg/kg, nicotine sensitization) or vehicle (conditioned hyperlocomotion) in experimental cages. RESULTS Given on day 10, LSP1-2111 (3mg/kg) as well as Lu AF21934 (2.5-5mg/kg) decreased the expression of cocaine sensitization. In another set of experiments, LSP1-2111 (3mg/kg) and Lu AF21934 (5mg/kg) administered on day 10 attenuated the conditioned hyperlocomotion in rats treated repeatedly with cocaine. Neither LSP1-2111 (1-3mg/kg) nor Lu AF21934 (2.5-5mg/kg) changed the expression of nicotine sensitization and conditioned hyperlocomotion in rats treated repeatedly with nicotine. None of the mGlu4 receptor agonist/modulator altered the basal locomotor activity or acute hyperactivity to cocaine or nicotine. CONCLUSIONS The present data indicate that pharmacological stimulation of mGlu4 receptors reduces the cocaine-induced expression of sensitization as well as conditioned hyperactivity. In contrast, mGlu4 receptor activation seems to be devoid of any effect on the locomotor effects of nicotine.
Collapse
Affiliation(s)
- Magdalena Zaniewska
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.
| | - Edmund Przegaliński
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Darío Doller
- Chemical and Pharmacokinetic Sciences, Lundbeck Research USA, Paramus, USA
| |
Collapse
|
33
|
Finlay C, Duty S. Therapeutic potential of targeting glutamate receptors in Parkinson's disease. J Neural Transm (Vienna) 2014; 121:861-80. [PMID: 24557498 DOI: 10.1007/s00702-014-1176-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/05/2014] [Indexed: 12/28/2022]
Abstract
Glutamate plays a complex role in many aspects of Parkinson's disease including the loss of dopaminergic neurons, the classical motor symptoms as well as associated non-motor symptoms and the treatment-related side effect, L-DOPA-induced dyskinesia. This widespread involvement opens up possibilities for glutamate-based therapies to provide a more rounded approach to treatment than is afforded by current dopamine replacement therapies. Beneficial effects of blocking postsynaptic glutamate transmission have already been noted in a range of preclinical studies using antagonists of NMDA receptors or negative allosteric modulators of metabotropic glutamate receptor 5 (mGlu5), while positive allosteric modulators of mGlu4 in particular, although at an earlier stage of investigation, also look promising. This review addresses each of the key features of Parkinson's disease in turn, summarising the contribution glutamate makes to that feature and presenting an up-to-date account of the potential for drugs acting at ionotropic or metabotropic glutamate receptors to provide relief. Whilst only a handful of these have progressed to clinical trials to date, notably NMDA and NR2B antagonists against motor symptoms and L-DOPA-induced dyskinesia, with mGlu5 negative allosteric modulators also against L-DOPA-induced dyskinesia, the mainly positive outcomes of these trials, coupled with supportive preclinical data for other strategies in animal models of Parkinson's disease and L-DOPA-induced dyskinesia, raise cautious optimism that a glutamate-based therapeutic approach will have significant impact on the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Clare Finlay
- Wolfson Centre for Age-Related Diseases, King's College London, WW1.28. Hodgkin Building, Guy's Campus, London, SE1 1UL, UK
| | | |
Collapse
|
34
|
Sławińska A, Wierońska JM, Stachowicz K, Marciniak M, Lasoń-Tyburkiewicz M, Gruca P, Papp M, Kusek M, Tokarski K, Doller D, Pilc A. The antipsychotic-like effects of positive allosteric modulators of metabotropic glutamate mGlu4 receptors in rodents. Br J Pharmacol 2014; 169:1824-39. [PMID: 23714045 DOI: 10.1111/bph.12254] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/24/2013] [Accepted: 05/12/2013] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Because agonists at metabotropic glutamate receptors exert beneficial effects in schizophrenia, we have assessed the actions of Lu AF21934 and Lu AF32615, two chemically distinct, selective and brain-penetrant positive allosteric modulators (PAMs) of the mGlu4 receptor, in several tests reflecting positive, negative and cognitive symptoms of schizophrenia in rodents. EXPERIMENTAL APPROACH Hyperactivity induced by MK-801 or amphetamine and head twitches induced by 2,5-dimethoxy-4-iodoamphetamine (DOI) in mice were used as models for positive symptoms. Disruption of social interaction and spatial delayed alternation tests induced by MK-801 in rats were used as models for negative and cognitive symptoms of schizophrenia, respectively. KEY RESULTS Lu AF21934 (0.1-5 mg·kg(-1) ) and Lu AF32615 (2-10 mg·kg(-1) ) dose-dependently inhibited hyperactivity induced by MK-801 or amphetamine. They also antagonized head twitches and increased frequency of spontaneous excitatory postsynaptic currents (EPSCs) in brain slices, induced by DOI. In mice lacking the mGlu4 receptor (mGlu4 (-/-) ) mice, Lu AF21934 did not antagonize DOI-induced head twitches. MK-801-induced disruption in the social interaction test was decreased by Lu AF21934 at 0.5 mg·kg(-1) and by Lu AF32615 at 10 mg·kg(-1) . In the delayed spatial alternation test, Lu AF21934 was active at 1 and 2 mg·kg(-1) , while Lu AF32615 was active at 10 mg·kg(-1) . CONCLUSIONS AND IMPLICATIONS We propose that activation by PAMs of the mGlu4 receptor is a promising approach to the discovery of novel antipsychotic drugs.
Collapse
Affiliation(s)
- Anna Sławińska
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nickols HH, Conn PJ. Development of allosteric modulators of GPCRs for treatment of CNS disorders. Neurobiol Dis 2014; 61:55-71. [PMID: 24076101 PMCID: PMC3875303 DOI: 10.1016/j.nbd.2013.09.013] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 12/14/2022] Open
Abstract
The discovery of allosteric modulators of G protein-coupled receptors (GPCRs) provides a promising new strategy with potential for developing novel treatments for a variety of central nervous system (CNS) disorders. Traditional drug discovery efforts targeting GPCRs have focused on developing ligands for orthosteric sites which bind endogenous ligands. Allosteric modulators target a site separate from the orthosteric site to modulate receptor function. These allosteric agents can either potentiate (positive allosteric modulator, PAM) or inhibit (negative allosteric modulator, NAM) the receptor response and often provide much greater subtype selectivity than orthosteric ligands for the same receptors. Experimental evidence has revealed more nuanced pharmacological modes of action of allosteric modulators, with some PAMs showing allosteric agonism in combination with positive allosteric modulation in response to endogenous ligand (ago-potentiators) as well as "bitopic" ligands that interact with both the allosteric and orthosteric sites. Drugs targeting the allosteric site allow for increased drug selectivity and potentially decreased adverse side effects. Promising evidence has demonstrated potential utility of a number of allosteric modulators of GPCRs in multiple CNS disorders, including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, as well as psychiatric or neurobehavioral diseases such as anxiety, schizophrenia, and addiction.
Collapse
Key Words
- (+)-6-(2,4-dimethylphenyl)-2-ethyl-6,7-dihydrobenzo[d]oxazol-4(5H)-one
- (1-(4-cyano-4-(pyridine-2-yl)piperidine-1-yl)methyl-4-oxo-4H-quinolizine-3-carboxylic acid)
- (1S,2S)-N(1)-(3,4-dichlorophenyl)cyclohexane-1,2-dicarboxamide
- (1S,3R,4S)-1-aminocyclo-pentane-1,3,4-tricarboxylic acid
- (3,4-dihydro-2H-pyrano[2,3]b quinolin-7-yl)(cis-4-methoxycyclohexyl) methanone
- (3aS,5S,7aR)-methyl 5-hydroxy-5-(m-tolylethynyl)octahydro-1H-indole-1-carboxylate
- 1-(1′-(2-methylbenzyl)-1,4′-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-one
- 1-[3-(4-butyl-1-piperidinyl)propyl]-3,4-dihydro-2(1H)-quinolinone
- 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- 2-(2-(3-methoxyphenyl)ethynyl)-5-methylpyridine
- 2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1Himidazol-4-yl)ethynyl)pyridine
- 2-methyl-6-(2-phenylethenyl)pyridine
- 2-methyl-6-(phenylethynyl)-pyridine
- 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide
- 3-cyclohexyl-5-fluoro-6-methyl-7-(2-morpholin-4-ylethoxy)-4H-chromen-4-one
- 3[(2-methyl-1,3-thiazol-4-yl)ethylnyl]pyridine
- 4-((E)-styryl)-pyrimidin-2-ylamine
- 4-[1-(2-fluoropyridin-3-yl)-5-methyl-1H-1,2,3-triazol-4-yl]-N-isopropyl-N-methyl-3,6-dihydropyridine-1(2H)-carboxamide
- 4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]-piperidine
- 5-methyl-6-(phenylethynyl)-pyridine
- 5MPEP
- 6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one
- 6-OHDA
- 6-hydroxydopamine
- 6-methyl-2-(phenylazo)-3-pyridinol
- 77-LH-28-1
- 7TMR
- AC-42
- ACPT-1
- AChE
- AD
- ADX71743
- AFQ056
- APP
- Allosteric modulator
- Alzheimer's disease
- BINA
- BQCA
- CDPPB
- CFMMC
- CNS
- CPPHA
- CTEP
- DA
- DFB
- DHPG
- Drug discovery
- ERK1/2
- FMRP
- FTIDC
- FXS
- Fragile X syndrome
- GABA
- GPCR
- JNJ16259685
- L-AP4
- L-DOPA
- Lu AF21934
- Lu AF32615
- M-5MPEP
- MMPIP
- MPEP
- MPTP
- MTEP
- Metabotropic glutamate receptor
- Muscarinic acetylcholine receptor
- N-[4-chloro-2[(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl]phenyl]-2-hydrobenzamide
- N-methyl-d-aspartate
- N-phenyl-7-(hydroxylimino)cyclopropa[b]chromen-1a-carboxamide
- NAM
- NMDA
- PAM
- PCP
- PD
- PD-LID
- PET
- PHCCC
- PQCA
- Parkinson's disease
- Parkinson's disease levodopa-induced dyskinesia
- SAM
- SIB-1757
- SIB-1893
- TBPB
- [(3-fluorophenyl)methylene]hydrazone-3-fluorobenzaldehyde
- acetylcholinesterase
- amyloid precursor protein
- benzylquinolone carboxylic acid
- central nervous system
- dihydroxyphenylglycine
- dopamine
- extracellular signal-regulated kinase 1/2
- fragile X mental retardation protein
- l-(+)-2-amino-4-phosphonobutyric acid
- l-3,4-dihydroxyphenylalanine
- mGlu
- metabotropic glutamate receptor
- negative allosteric modulator
- phencyclidine
- positive allosteric modulator
- positron emission tomography
- potassium 30-([(2-cyclopentyl-6-7-dimethyl-1-oxo-2,3-dihydro-1H-inden-5yl)oxy]methyl)biphenyl l-4-carboxylate
- seven transmembrane receptor
- silent allosteric modulator
- γ-aminobutyric acid
Collapse
Affiliation(s)
- Hilary Highfield Nickols
- Division of Neuropathology, Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, 37232, USA
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
36
|
Abstract
Mood disorders are common and debilitating, resulting in a significant public health burden. Current treatments are only partly effective and patients who have failed to respond to trials of existing antidepressant agents (eg, those who suffer from treatment-resistant depression [TRD]) require innovative therapeutics with novel mechanisms of action. Although neuroscience research has elucidated important aspects of the basic mechanisms of antidepressant action, most antidepressant drugs target monoaminergic mechanisms identified decades ago. Glutamate, the major excitatory neurotransmitter in the central nervous system, and glutamatergic dysfunction has been implicated in mood disorders. These data provide a rationale for the pursuit of glutamatergic agents as novel therapeutic agents. Here, we review preclinical and clinical investigations of glutamatergic agents in mood disorders with a focus on depression. We begin with discussion of evidence for the rapid antidepressant effects of ketamine, followed by studies of the antidepressant efficacy of the currently marketed drugs riluzole and lamotrigine. Promising novel agents currently in development, including N-methyl-D-aspartate (NMDA) receptor modulators, 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid (AMPA) receptor modulators, and drugs with activity at the metabotropic glutamate (mGlu) receptors are then reviewed. Taken together, both preclinical and clinical evidence exists to support the pursuit of small molecule modulators of the glutamate system as novel therapeutic agents in mood disorders. It is hoped that by targeting neural systems outside of the monoamine system, more effective and perhaps faster acting therapeutics can be developed for patients suffering from these disabling disorders.
Collapse
Affiliation(s)
- Kyle Ab Lapidus
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|